Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT OF LEUKEMIA WITH HISTONE DEACETYLASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2016/007423
Kind Code:
A1
Abstract:
Provided herein are combinations comprising an HDAC inhibitor and azacitidine for the treatment of leukemia in a subject in need thereof. Provided herein are combinations comprising an HDAC inhibitor and azacitidine for the treatment of acute myelogenous leukemia in a subject in need thereof. Also provided herein are methods for treating leukemia in a subject in need thereof, comprising administering to the subject an effective amount of the above combination or an HDAC inhibitor, as well as methods for treating acute myelogenous leukemia in a subject in need thereof, comprising administering to the subject an effective amount of the above combination or an HDAC inhibitor.

Inventors:
TAMANG DAVID LEE (US)
JONES SIMON S (US)
MIN CHENGYIN (US)
YANG MIN (US)
Application Number:
PCT/US2015/039225
Publication Date:
January 14, 2016
Filing Date:
July 06, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ACETYLON PHARMACEUTICALS INC (US)
TAMANG DAVID LEE (US)
International Classes:
A61K31/706; A61K31/4184; A61P35/02
Domestic Patent References:
WO2012068109A22012-05-24
Foreign References:
US20140128391A12014-05-08
US20110300134A12011-12-08
US20120094927A12012-04-19
Other References:
ABUJAMRA A. L. ET AL.: "Histone deacetylase inhibitors: a new perspective for the treatment of leukemia.", LEUK RES., vol. 34, no. 6, 2010, pages 687 - 695, XP027015253
See also references of EP 3166603A4
Attorney, Agent or Firm:
TRINQUE, Brian, C. et al. (28 State StreetBoston, MA, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A pharmaceutical combination for treating leukemia comprising a therapeutically effective amount of a histone deacetylase (HDAC) inhibitor or a pharmaceutically acceptable salt thereof, and azacitidine or a pharmaceutically acceptable salt thereof.

2. The combination of claim 1 , wherein the leukemia is acute myelogenous leukemia

(AML).

3. The combination of claim 1 , wherein the HDAC inhibitor is an HDAC6-specific inhibitor.

4. The combination of claim 1 , wherein the HDAC inhibitor is an HDACl/2-specific inhibitor.

5. The combination of claim 1 , wherein the HDAC inhibitor is an HDACl/2/6-specific inhibitor.

6. The combination of claim 3, wherein the HDAC6-specific inhibitor is a compound of Formula I:

(I)

or a pharmaceutically acceptable salt thereof,

wherein,

ring B is aryl or heteroaryl;

Ri is an aryl or heteroaryl, each of which may be optionally substituted by OH, halo, or Ci_6-alkyl; and

R is H or Ci_6-alkyl. The combination of claim 6, wherein the compound of Formula I

or a pharmaceutically acceptable salt thereof.

The combination of claim 6, wherein the compound of Formula I

or a pharmaceutically acceptable salt thereof.

9. The combination of claim 3, wherein the HDAC6-specific inhibitor is a compound of Formula II:

or a pharmaceutically acceptable salt thereof,

wherein,

Rx and Ry together with the carbon to which each is attached, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl;

each RA is independently Ci_6-alkyl, Ci_6-alkoxy, halo, OH, -N02, -CN, or -NH2; and m is 0 or 1. 10. The combination of claim 9, wherein the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

11. The combination of claim 9, wherein the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

12. The combination of claim 4, wherein the HDACl/2-specific inhibitor is a compound of Formula III:

(III)

or a pharmaceutically acceptable salt thereof,

wherein,

R1 is aryl or heteroaryl;

R2 and R3 are each independently selected from C3-6-cycloalkyl, Ci_6-alkyl-OR , Ci-6- alkyl-C _6-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, C2_6-alkenyl;

R6 is H or Ci_6-alkyl; and

R7 is H or C3_6-cycloalkyl.

13. The combination of claim 12, wherein the compound of Formula III is:

or a pharmaceutically acceptable salt thereof.

14. The combination of claim 4, wherein the HDACl/2-specific inhibitor is the compound:

or a pharmaceutically acceptable sa t thereof. 15. The combination of claim 5, wherein the HDACl/2/6-specific inhibitor is a compound of Formula IV:

(IV)

or a pharmaceutically acceptable salts thereof,

wherein,

Rx is independently selected from the group consisting of -CO2R1, and - C(0)N(R1)2;

Ry is selected from the group consisting of H, Ci_6-alkyl, Ci_6-alkoxy, halo, -OH, - N02, -CN, -NH2, -C(0)R1, -CC^R1, and -C(0)N(R1)2;

each R1 is, independently for each occurrence, selected from the group consisting of

H, Ci_6-alkyl, C3-8-cycloalkyl, C3_7-heterocycloalkyl, aryl, heteroaryl, Ci_6-alkyl-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, Ci_6-alkyl-aryl, and Ci_6-alkyl-heteroaryl; and

Rz is selected from the group consisting of Ci_6-alkyl, C3-8-cycloalkyl, C3-7- heterocycloalkyl, aryl, and heteroaryl.

16. The combination of claim 15, wherein the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof.

17. The combination of claim 1, where hibitor is:

or a pharmaceutically acceptable salt thereof.

18. The combination of claim 1, wherein the combination further comprises a

pharmaceutically acceptable carrier.

19. A method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a histone deacetylase (HDAC) inhibitor or a pharmaceutically acceptable salt thereof, and azacitidine or a pharmaceutically acceptable salt thereof.

20. The method of claim 19, wherein the leukemia is acute myelogenous leukemia (AML).

21. The method of claim 19, wherein the HDAC inhibitor is an HDAC6-specific inhibitor.

22. The method of claim 19, wherein the HDAC inhibitor is an HDACl/2-specific inhibitor.

23. The method of claim 19, wherein the HDAC inhibitor is an HDACl/2/6-specific inhibitor.

24. The method of claim 21, wherein the HDAC6-specific inhibitor is a compound of Formula I:

(I)

a pharmaceutically acceptable salt thereof,

wherein,

ring B is aryl or heteroaryl;

Ri is an aryl or heteroaryl, each of which may be optionally substituted by OH, halo, Ci_6-alkyl; and

R is H or Ci_6-alkyl.

The method of claim 24, wherein the compound of Formula I

or a pharmaceutically acceptable salt thereof.

The method of claim 24, wherein the compound of Formula I

or a pharmaceutically acceptable salt thereof.

27. The method of claim 21, wherein the HDAC6-specific inhibitor is a compound of Formula II:

or a pharmaceutically acceptable salt thereof,

wherein,

Rx and Ry together with the carbon to which each is attached, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl;

each RA is independently Ci_6-alkyl, Ci_6-alkoxy, halo, OH, -N02, -CN, or -NH2; and m is 0 or 1.

28. The method of claim 27, wherein the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

29. The method of claim 27, wherein the compound of Formula II

or a pharmaceutically acceptable salt thereof.

30. The method of claim 22, wherein the HDACl/2-specific inhibitor is a compound of Formula III:

(III)

or a pharmaceutically acceptable salt thereof,

wherein,

R1 is aryl or heteroaryl;

R2 and R3 are each independently selected from C3_6-cycloalkyl, Ci_6-alkyl-OR6, Ci_6- alkyl-C3-6-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, C2_6-alkenyl;

R6 is H or Ci_6-alkyl; and R7 is H or C3-6-cycloalkyl.

The method of claim 30, wherein the compound of Formula III

or a pharmaceutically acceptable salt thereof.

The method of claim 22, wh -specific inhibitor is the compound:

or a pharmaceutically acceptable salt thereof.

33. The method of claim 23, wherein the HDACl/2/6-specific inhibitor is a compound of Formula IV:

(IV)

or a pharmaceutically acceptable salts thereof,

wherein,

Rx is independently selected from the group consisting of -CO2R1, and C(0)N(R1)2;

Ry is selected from the group consisting of H, Ci_6-alkyl, Ci_6-alkoxy, halo, -OH, - N02, -CN, -NH2, -C(0)R1, -CO2R1, and -C(0)N(R1)2; each R is, independently for each occurrence, selected from the group consisting of

H, Ci_6-alkyl, C3-8-cycloalkyl, C3_7-heterocycloalkyl, aryl, heteroaryl, Ci_6-alkyl-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, Ci_6-alkyl-aryl, and Ci_6-alkyl-heteroaryl; and

Rz is selected from the group consisting of Ci_6-alkyl, C3-8-cycloalkyl, C3-7- heterocycloalkyl, aryl, and heteroaryl.

34. The method of claim 33, wherein the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof.

35. The method of claim 19, wherein the HDAC inhibitor is:

or a pharmaceutically acceptable salt thereof.

36. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula I:

(I)

or a pharmaceutically acceptable salt thereof,

wherein, ring B is aryl or heteroaryl;

Ri is an aryl or heteroaryl, each of which may be optionally substituted by OH, halo, Ci_6-alkyl; and

R is H or Ci_6-alkyl.

The method of claim 36, wherein the compound of Formula I is:

or a pharmaceutically acceptable salt thereof.

The method of claim 36, wherein the compound of Formula I

or a pharmaceutically acceptable salt thereof.

39. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula II:

or a pharmaceutically acceptable salt thereof,

wherein,

Rx and Ry together with the carbon to which each is attached, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl;

each RA is independently Ci_6-alkyl, Ci_6-alkoxy, halo, OH, -N02, -CN, or -NH2; and m is 0 or 1.

40. The method of claim 39, wherein the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

41. The method of claim 39, wherein the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

42. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula III:

(III)

or a pharmaceutically acceptable salt thereof,

wherein,

R1 is aryl or heteroaryl;

R2 and R3 are each independently selected from C3_6-cycloalkyl, Ci_6-alkyl-OR6, Ci_6- alkyl-C3-6-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, C2_6-alkenyl;

R6 is H or Ci_6-alkyl; and

R • 7 i :s H or C3-6-cycloalkyl.

43. The method of claim 42, wherein the compound of Formula III is:

or a pharmaceutically acceptable salt thereof.

44. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound:

or a pharmaceutically acceptable salt thereof.

45. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound:

or a pharmaceutically acceptable salt thereof.

46. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula IV:

(IV)

or a pharmaceutically acceptable salts thereof,

wherein,

Rx is independently selected from the group consisting of -CO2R1, and -

C(0)N(R1)2;

Ry is selected from the group consisting of H, Ci_6-alkyl, Ci_6-alkoxy, halo, -OH, - N02, -CN, -NH2, -C(0)R1, -CO2R1, and -C(0)N(R1)2;

each R1 is, independently for each occurrence, selected from the group consisting of H, Ci_6-alkyl, C3-8-cycloalkyl, C3-7-heterocycloalkyl, aryl, heteroaryl, Ci_6-alkyl-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, Ci_6-alkyl-aryl, and Ci_6-alkyl-heteroaryl; and

Rz is selected from the group consisting of Ci_6-alkyl, C3-8-cycloalkyl, C3-7- heterocycloalkyl, aryl, and heteroaryl. 47. The method of claim 46, wherein the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof. 48. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a HDAC1/2- specific inhibitor.

49. A method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a HD AC 1/2/6- specific inhibitor.

50. The method of any one of claims 36-49, wherein the method further comprises administering azacitidine to the subject.

5 1 . A method for decreasing cell viability of cancer cells comprising administering a combination comprising an HDAC inhibitor and azacitidine.

52. A method for inducing differentiat ion of cancer cells comprising administering a combination comprising an HDAC inhibitor and azacitidine.

53. A method for inducing apoptosis of cancer cells comprising admin istering a combination comprising an HDAC inhibitor and azacitidine.

Description:
TREATMENT OF LEUKEMIA WITH HISTONE DEACETYLASE INHIBITORS

CROSS-REFERENCE TO RELATED APPLICATIONS

This application is related to U.S. Provisional Application No. 62/021,473, filed July 7, 2014, U.S. Provisional Application No. 62/061,233, filed October 8, 2014, and U.S.

Provisional Application No. 62/147,218, filed April 14, 2015. The contents of each of these applications are incorporated herein by reference in their entirety.

BACKGROUND

Cancer is distinguished by uncontrolled proliferation of cells. The cellular components of blood originate from pluripotent hematopoietic stem cells. Via their regenerative and differentiating capacities, stem cells generate lymphoid and myeloid precursors, which then produce lymphocytes, neutrophils, eosinophils, basophils,

erythrocytes, and platelets. In leukemia, high levels of immature white blood cells, or blasts, are present. Four main types of leukemia are recognized: acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL) and chronic myeloid leukemia (CML); although less common types are known as well.

Leukemia has an average 5-year mortality rate of 40%, and in 2012 developed in over 350,000 people globally. Therefore, there remains a continued and urgent need for therapies directed toward treatment of leukemia.

SUMMARY

In one aspect, provided herein is a pharmaceutical combination for treating leukemia, comprising a therapeutically effective amount of a histone deacetylase (HDAC) inhibitor or a pharmaceutically acceptable salt thereof, and azacitidine or a pharmaceutically acceptable salt thereof. In one embodiment, the HDAC inhibitor is an HDAC6-specific inhibitor. In another embodiment, the HDAC inhibitor is an HDACl/2-specific inhibitor. In another embodiment, the HDAC inhibitor is an HDACl/2/6-specific inhibitor. embodiment, the HDAC6-specific inhibitor is a compound of Formula I

(I)

or a pharmaceutically acceptable salt thereof.

In another embodimen -specific inhibitor is a compound of Formula II:

(II)

or a pharmaceutically acceptable salt thereof.

In another embodiment, the HDACl/2-specific inhibitor is a compound of Formula

III:

(HI)

or a pharmaceutically acceptable salt thereof.

In another embodiment, the H is:

or a pharmaceutically acceptable salt thereof.

In another embodiment, the HDACl/2/6-specific inhibitor is a compound of Formula

IV:

(IV)

or a pharmaceutically acceptable salt thereof.

In another embodiment, the combination further comprises a pharmaceutically acceptable carrier.

In another aspect, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a pharmaceutical combination comprising a histone deacetylase (HDAC) inhibitor or a pharmaceutically acceptable salt thereof, and azacitidine or a pharmaceutically acceptable salt thereof. In one embodiment, the HDAC inhibitor is an HDAC6-specific inhibitor. In another embodiment, the HDAC inhibitor is an HDACl/2-specific inhibitor. In another embodiment, the HDAC inhibitor is an HDACl/2/6-specific inhibitor.

In yet another embodiment, the HDAC6-specific inhibitor is a compound of Formula

I:

(I)

or a pharmaceutically acceptable salt thereof.

In another embodimen -specific inhibitor is a compound of Formula II:

(II)

or a pharmaceutically acceptable salt thereof,

In another embodiment, the HDACl/2-specific inhibitor is a compound of Formula III:

(III)

or a pharmaceutically acceptable salt thereof.

In another embodiment, the H :

or a pharmaceutically acceptable salt thereof.

In another embodiment, the HDACl/2/6-specific inhibitor is a compound of Formula

IV:

(IV)

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula I:

(I)

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula II:

(II)

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula III:

(HI)

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound:

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula IV:

(IV)

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof.

In another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a HDACl/2-specific inhibitor.

In yet another aspect, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a HDACl/2/6-specific inhibitor.

BRIEF DESCRIPTION OF THE FIGURES

Figures 1A-D are a set of four graphs that show synergy of HDAC inhibitors and azacitidine on AML cells. Each of the graphs shows the CI values plotted as a function of Fa. Figure 1A shows data for azacitidine and Compound A on HL-60 cells, Figure IB shows data for azacitidine and Compound C on HL-60 cells, Figure 1C shows data for azacitidine and Compound E on HL-60 cells, and Figure ID shows data for azacitidine and Compound F on HL-60 cells.

Figures 2A-D are a set of three graphs and pictures showing that HDAC inhibition increases apoptosis and suppresses AML1/ETO in AML. Figures 2A-C show data for the Kasumi-1 cell cycle at 72 hours. Figure 2A shows data for Compound B, Figure 2B shows data for Compound G, and Figure 2C shows data for Compound E. Figure 2D shows pictures of gels and the expression of the fusion protein AMLl/ETO or ACTB. Data is shown for Compound A and panobinostat.

Figures 3A-D are a set of four graphs that show the single agent activity on viability in AML cell lines. 6 AML cell lines: HL-60 (large filled circles), THP-1 (upright filled triangles), MV-4-11 (small filled diamonds), Kasumi-1 (open squares), NB4 (open upside- down triangles), and MOLM-13 (open diamonds) were exposed to increasing concentrations of either Compound B (Figure 3A), Compound A (Figure 3B), Compound E (Figure 3C), or azacitidine (Figure 3D) to determine their response to drug treatment.

Figures 4A-F are a set of 6 graphs that show the single agent activity on

differentiation and apoptosis in AML cell lines. 3 AML cell lines: HL-60 (Figures 4A and 4D), Kasumi-1 (Figures 4B and 4E), and NB4 (Figures 4C and 4F) were treated with the indicated concentrations of compounds. In Figures 4A-C, surface levels of the myeloid differentiation marker CDl lb were determined. In Figures 4D-F, apoptosis was assessed by flow cytometry by measuring Annexin V binding and cellular permeability to propidium iodide at 96 hours post-treatment. The relative fraction of cells that were alive, in early apoptosis, in late apoptosis, or dead was then determined.

Figures 5A-F are a set of 6 graphs that show the combination of HDAC inhibitors and azacitidine in the HL-60 cell line. Cells were treated with DMSO, Compound B, Compound A, or Compound E as a single agent or in combination with azacitidine for 96 hours. Surface levels of the myeloid differentiation marker CDl lb were determined

(Figures 5A, 5C, 5E). Apoptosis was assessed by flow cytometry by measuring Annexin V binding and cellular permeability to propidium iodide at 96 hours post-treatment (Figures 5B, 5D, 5F). The relative fraction of cells that were alive, in early apoptosis, in late apoptosis, or dead was then determined.

Figures 6A-F are a set of 6 graphs that show the combination of HDAC inhibitors and azacitidine in the Kasumi-1 cell line. Cells were treated with DMSO, Compound B, Compound A, or Compound E as a single agent or in combination with azacitidine at the indicated concentrations. Surface levels of the myeloid differentiation marker CDl lb were determined (Figures 6A, 6C, 6E). Apoptosis was assessed by flow cytometry by measuring Annexin V binding and cellular permeability to propidium iodide at 96 hours post-treatment (Figures 6B, 6D, 6F). The relative fraction of cells that were alive, in early apoptosis, in late apoptosis, or dead was then determined.

Figures 7A-F are a set of 6 graphs that show the combination of HDAC inhibitors and azacitidine in the NB4 cell line. Cells were treated with DMSO, Compound B,

Compound A, or Compound E as a single agent or in combination with azacitidine at the indicated concentrations. Surface levels of the myeloid differentiation marker CDl lb were determined (Figure 7A, 7C, 7E). Apoptosis was assessed by flow cytometry by measuring Annexin V binding and cellular permeability to propidium iodide at 96 hours post-treatment (Figure 7B, 7D, 7F). The relative fraction of cells that were alive, in early apoptosis, in late apoptosis, or dead was then determined.

Figures 8A-F show exposure of AML cell lines to increasing doses of Compound E (Figure 8A), Compound H (Figure 8B), Compound C (Figure 8C), Compound A (Figure 8D), Compound B (Figure 8E) and Compound G (Figure 8F) for 72h to confirm their sensitivity to HDAC inhibition. 6 AML cell lines were used in this study: HL-60 (large filled circles), NB4 (upright filled triangles), Kasumi-1 (small filled diamonds), MV4-11 (open squares), THP-1 (open upside-down triangles), and MOLM-13 (open diamonds).

Figures 9A-C show treatment of MV4-11 with the indicated doses of compounds. Figure 9A shows surface levels of myeloid differentiation marker CD1 lb, determined by FACS at 72h post-treatment. Compound E, Compound H, Compound A, and Compound G increased the percentage of CD1 lb positive cells. Compound C had no effect. Figure 9B shows assessment of the cell cycle by flow cytometry after incorporation of EdU and staining with Far Red at 72h post-treatment. The distribution of cells among G0/G1 phase, G2/M phase, S phase and subGl phase was determined. Figure 9C shows the assessment of apoptosis by flow cytometry via measuring Annexin V binding and cellular permeability to propidium iodide at 96h post-treatment. The relative fraction of cells that were live, in early apoptosis, in late apoptosis, or dead was then determined.

Figures 10A-F show the treatment of the following AML cell lines: Kasumi-1

(Figures 10A and 10B), HL-60 (Figures IOC and 10D) and NB4 (Figures 10E and 10F), with indicated doses of compounds. Figures 10A, IOC, and 10E show surface levels of myeloid differentiation marker CD1 lb determined by FACS at 72h post-treatment. Figures 10B, 10D, and 10F show the assessment of apoptosis by FACS at 96h post-treatment (see, e.g., Figure 9C).

Figures 11A-D show that combinations of HDACl/2 inhibition with azacitidine result in synergistic decreases in HL-60 cell viability. HL-60 cells were treated with increasing doses of azacitidine with Compound E (Figure 11 A) or with Compound A

(Figure 11B) or with Compound H (Figure 11C) or with Compound C (Figure 11D), and cell viability was assessed at 72 hr by cell titer glo assay. The combination index (CI) and relative fraction affected (Fa) was determined at each dose level using CalcuSyn software. The measurement of CI values less than 1 (shaded region) strongly support a synergistic interaction between drugs. Figures 12A-F show the treatment of MV4-11 cells with Compound E or with Compound A or with Compound B as single agent or in combination with azacitidine at indicated doses. Figures 12A, 12C, and 12E show surface levels of CDl lb determined by FACS at 72h post-treatment. Figures 12B, 12D, and 12F show assessment of apoptosis by FACS at 96h post-treatment.

Figure 13A shows that treatment with Compound A plus azacitidine reduced tumor growth in vivo as compared to treatment with azacitidine or vehicle alone.

Figure 13B shows that treatment with Compound A plus azacitidine reduced the fold tumor volume change as compared to treatment with azacitidine or vehicle alone.

Figure 13C shows that treatment with Compound A plus azacitidine increased survival in vivo as compared to treatment with azacitidine or vehicle alone.

Figure 14A shows the IC 50 values of Compound A, Compound J and azacitidine on inhibiting colony formation in 6 bone marrow samples derived from AML patients.

Figure 14B shows the effect of HD AC 1/2 inhibition alone and in combination with azacitidine on colony formation of the primary AML patient sample 4031113SH.

Figure 14C shows the effect of HD AC 1/2 inhibition alone and in combination with azacitidine on colony formation of the primary AML patient sample VMBM0007.

Figure 14D shows the effect of HD AC 1/2 inhibition alone and in combination with azacitidine on colony formation of the primary AML patient sample 184090514.

Figure 14E shows the effect of HD AC 1/2 inhibition alone and in combination with azacitidine on colony formation of the primary AML patient sample 103113SH.

Figure 15A shows the IC 50 values of azacitidine, Compound A and Compound J on inhibiting proliferation of AML blast freshly derived from bone marrow of AML patients.

Figure 15B shows the AUC (area under the curve) values for azacitidine, Compound A and Compound J on inhibiting proliferation of AML blast freshly derived from bone marrow of AML patients.

Figure 15C shows that the combination of azacitidine with Compound J results in a synergistic interaction between the two drugs on inhibiting proliferation of primary AML cells freshly derived from AML patients in 4 out of 5 bone marrow samples.

Figure 16 shows Compound E and azacitidine synergistically induce GATA2 expression in MV4-11 AML cells.

Figure 17A shows that various AML cell lines are sensitive to HDACl/2 inhibition. Figure 17B shows the surface levels of myloid differentiation marker CD1 lb in MV4-11 (AML) cells as determined by FACS after 72 hours of treatment with the indicated compound.

Figure 17C shows a cell cycle assessment in MV4-11 (AML) cells as determined by flow cytometry after 72 hours of treatment with the indicated compound.

Figure 17D shows the relative fraction of MV4-11 (AML) cells that were live, in early apoptosis, in late apoptosis or dead as assessed by flow cytometry after 72 hours of treatment with the indicated compound.

DETAILED DESCRIPTION

Provided herein are combinations comprising an HDAC inhibitor and azacitidine for the treatment of leukemia in a subject in need thereof. Also provided herein are combinations comprising an HDAC inhibitor and azacitidine for the treatment of acute myelogenous leukemia in a subject in need thereof. Also provided herein are methods for treating leukemia in a subject in need thereof, comprising administering to the subject an effective amount of an HDAC inhibitor, or alternatively administering the above combination comprising an HDAC inhibitor and azacitidine. Provided herein are methods for treating acute myelogenous leukemia in a subject in need thereof, comprising administering to the subject an effective amount of an HDAC inhibitor, or alternatively administering the above combination comprising an HDAC inhibitor and azacitidine.

Definitions

Listed below are definitions of various terms used herein. These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.

The term "about" generally indicates a possible variation of no more than 10%, 5%, or 1% of a value. For example, "about 25 mg/kg" will generally indicate, in its broadest sense, a value of 22.5-27.5 mg/kg, i.e., 25 ± 2.5 mg/kg.

The term "alkyl" refers to saturated, straight- or branched-chain hydrocarbon moieties containing, in certain embodiments, between one and six (Ci- 6 alkyl), or one and eight carbon atoms (Ci-8 alkyl), respectively. Examples of Ci- 6 alkyl moieties include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl moieties; and examples of Ci-s alkyl moieties include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, and octyl moieties.

The number of carbon atoms in an alkyl substituent can be indicated by the prefix "C x - y ," where x is the minimum and y is the maximum number of carbon atoms in the substituent. Likewise, a C x chain means an alkyl chain containing x carbon atoms.

The term "alkoxy" refers to an -O-alkyl moiety.

The terms "cycloalkyl" or "cycloalkylene" denote a monovalent group derived from a monocyclic or polycyclic saturated or partially unsaturated carbocyclic ring compound. Examples of C 3 -8-cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C 3 -Ci2-cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl. Also contemplated are groups derived from a monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond. Examples of such groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, and the like. In some embodiments, cycloalkyl groups have from three to six carbon atoms (C 3 -6

cyclcoalkyl). In some embodiments, cycloalkyl groups have from three to eight carbon atoms (C3-8 cyclcoalkyl).

The term "aryl" refers to a mono- or poly-cyclic carbocyclic ring system having one or more aromatic rings, fused or non-fused, including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like. In some embodiments, aryl groups have six carbon atoms. In some embodiments, aryl groups have from six to ten carbon atoms (C 6 -io- aryl). In some embodiments, aryl groups have from six to sixteen carbon atoms (C 6 -i6-aryl).

The term "heteroaryl" refers to a mono- or poly-cyclic (e.g., bi-, or tri-cyclic or more) fused or non-fused, moieties or ring system having at least one aromatic ring, having from five to ten ring atoms of which one ring atom is selected from S, O, N and Si; zero, one or two ring atoms are additional heteroatoms independently selected from S, O, N and Si; and the remaining ring atoms are carbon. Heteroaryl includes, but is not limited to pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like.

The term "halo" refers to a halogen, such as fluorine, chlorine, bromine, and iodine. The term "alkenyl" denotes a monovalent group derived from a hydrocarbon moiety containing, in certain embodiments, from two to six (C 2 - 6 alkenyl), or two to eight carbon atoms having at least one carbon-carbon double bond (C 2 - 8 alkenyl). The double bond may or may not be the point of attachment to another group. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, heptenyl, octenyl and the like.

The term "cycloalkyl" denotes a monovalent group derived from a monocyclic or polycyclic saturated or partially unsaturated carbocyclic ring compound. Examples of C 3 _s- cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C 3 _i2-cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptyl, and bicyclo [2.2.2] octyl. Also contemplated are groups derived from a monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond. Examples of such groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, eye lo heptenyl, cyclooctenyl, and the like. In some embodiments, cycloalkyl groups have from three to six carbon atoms (C 3 _ 6 cyclcoalkyl). In some embodiments, cycloalkyl groups have from three to eight carbon atoms (C 3 _8 cyclcoalkyl).

The term "heterocycloalkyl" refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused or non- fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur, and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (iii) the nitrogen and sulfur heteroatoms may optionally be oxidized, (iv) the nitrogen heteroatom may optionally be quaternized, and (iv) any of the above rings may be fused to a benzene ring. Representative heterocycloalkyl groups include, but are not limited to, [l,3]dioxolane, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl,

isothiazolidinyl, and tetrahydrofuryl. In an embodiment, the heterocycloalkyl group is a 4-7, e.g., 4-6, membered ring.

The term "HDAC" refers to histone deacetylases, which are enzymes that remove the acetyl groups from the lysine residues in core histones, thus leading to the formation of a condensed and transcriptionally silenced chromatin. There are currently 18 known histone deacetylases, which are classified into four groups. Class I HDACs, which include HDACl, HDAC2, HDAC3, and HDAC 8, are related to the yeast RPD3 gene. Class II HDACs, which include HDAC4, HDAC5, HDAC6, HDAC7, HDAC9, and HDACIO, are related to the yeast Hdal gene. Class III HDACs, which are also known as the sirtuins are related to the Sir2 gene and include SIRTl-7. Class IV HDACs, which contains only HDACl 1, has features of both Class I and II HDACs. The term "HDAC" refers to any one or more of the 18 known histone deacetylases, unless otherwise specified.

The term "HDAC6-specific" means that the compound binds to HDAC6 to a substantially greater extent, such as 5X, 10X, 15X, 20X greater or more, than to any other type of HDAC enzyme, such as HDACl or HDAC2. That is, the compound is selective for HDAC6 over any other type of HDAC enzyme. For example, a compound that binds to HDAC6 with an IC 50 of 10 nM and to HDACl with an IC 50 of 50 nM is HDAC6-specific. On the other hand, a compound that binds to HDAC6 with an IC 50 of 50 nM and to HDACl with an IC 50 of 60 nM is not HDAC6-specific.

The term "HDACl/2-specific" means that the compound binds to HDACl and HDAC2 to a substantially greater extent, such as 5X, 10X, 15X, 20X greater or more, than to any other type of HDAC enzyme, such as HDAC3 or HDAC6. That is, the compound is selective for HDACl and HDAC2 over any other type of HDAC enzyme. For example, a compound that binds to HDACl and HDAC2 with an IC 50 of 10 nM and to HDAC3 with an IC 50 of 50 nM is HDACl/2-specific. On the other hand, a compound that binds to HDACl and HDAC2 with an IC 50 of 50 nM and to HDAC3 with an IC 50 of 60 nM is not HDAC 1/2- specific.

The term "HDACl/2/6-specific" means that the compound binds to HDACl, HDAC2 and HDAC6 to a substantially greater extent, such as 5X, 10X, 15X, 20X greater or more, than to any other type of HDAC enzyme, such as HDAC3. That is, the compound is selective for HDACl, HDAC2 and HDAC6 over any other type of HDAC enzyme. For example, a compound that binds to HDACl, HDAC2 and HDAC6 with an IC 50 of 10 nM and to HDAC3 with an IC 50 of 50 nM is HDACl/2-specific. On the other hand, a compound that binds to HDACl, HDAC2 and HDAC6 with an IC 50 of 50 nM and to HDAC3 with an IC 50 of 60 nM is not HDACl/2-specific.

The term "combination" refers to two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such combination of therapeutic agents may be in the form of a single pill, capsule, or intravenous solution. However, the term "combination" also encompasses the situation when the two or more therapeutic agents are in separate pills, capsules, or intravenous solutions. Likewise, the term "combination therapy" refers to the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, or in separate containers (e.g., capsules) for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner, either at approximately the same time or at different times. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.

The term "leukemia" refers to a hematologic malignancy. The term "leukemia" includes but is not limited to acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), acute monocytic leukemia (AML), biphenotypic acute leukemia (BAL), hairy cell leukemia (HCL), or acute promyelocytic leukemia (APL).

As used herein, the term "CD 1 lb-expressing" refers to the expression of Cluster of Differentiation Molecule 1 1B (CD l ib).

The term "inhibitor" is synonymous with the term antagonist.

Histone Deacetylase (HDAC) Inhibitors

Provided herein are methods for treating leukemia in a subject in need thereof. Also provided herein are pharmaceutical combinations for the treatment of leukemia (e.g., AML) in a subject in need thereof.

The combinations and methods provided herein comprise a histone deacetylase (HDAC) inhibitor. The HDAC inhibitor can be any HDAC inhibitor. Thus, the HDAC inhibitor may be selective or non-selective to a particular type of histone deacetylase enzyme. Preferably, the HDAC inhibitor is a selective HDAC inhibitor. More preferably, the HDAC inhibitor is an HDAC6-specific inhibitor, an HDACl/2-specific inhibitor, or an HDAC 1/2/6- specific inhibitor.

In some embodiments, the HDAC6-specific inhibitor is a compound of Formula I:

(I) a pharmaceutically acceptable salt thereof,

wherein,

ring B is aryl or heteroaryl;

Ri is an aryl or heteroaryl, each of which may be optionally substituted by OH, halo, Ci_6-alkyl; and

R is H or Ci_6-alkyl.

The preparation and properties of selective HDAC6 inhibitors according to Formula I are provided in International Patent Application No. PCT/US2011/021982, the entire contents of which are incorporated herein by reference.

In other embodiments, -specific inhibitor is a compound of Formula II:

(II)

or a pharmaceutically acceptable salt thereof,

wherein,

R x and R y , together with the carbon to which each is attached, form a cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl;

each R A is independently Ci_6-alkyl, Ci_6-alkoxy, halo, OH, -N0 2 , -CN, or -NH 2 ; and m is 0, 1, or 2.

Representative compounds of Formula II include, but are not limited to:

Compound C Compound D

IC 50 (nM) HDAC6 = 7 HDAC1 = 2123 IC 50 (nM) HDAC6 = 2 HDAC1 = 94

HDAC2 = 2570 HDAC3=11223 HDAC2 = 128 HDAC3=219

or pharmaceutically acceptable salts thereof.

The preparation and properties of selective HDAC6 inhibitors according to Formula II are provided in International Patent Application No. PCT/US2011/060791, the entire contents of which are incorporated herein by reference.

In some embodiments, the HDACl/2-specific inhibitor is a compound of Formula III:

(Ill)

pharmaceutically acceptable salt thereof,

wherein,

Pv 1 is aryl or heteroaryl;

Pv 2 and R 3 are each independently selected from C3_6-cycloalkyl, Ci_6-alkyl-OR 6 , Ci_ 6 - alkyl-C3_6-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, and C2_6-alkenyl;

R 6 is H or Ci_6-alkyl; and

Pv 7 is H or C3-6-cycloalkyl.

Compounds of Formula III are represented by, but not limited to, Compound E, or pharmaceutically acceptable salts thereof.

N-(2-amino-5-(thiophen-2-yl)phenyl)-2-cyclopropyl- 1 -(2- morpholinoethyl)-lH-indole-5-carboxamide

IC 50 (nM): HDAC1 = 6 HDAC2 = 36 HDAC3 = 445

C max = 2037 AUC = 9496

hERG IC 50 (μΜ) >30

In another embodiments, the HDACl/2-specific inhibitor is N-(2-amino-5-(thiophen- 2-yl)phenyl)-2-(piperazin-l-yl)quinoline-6-carboxamide (or a pharmaceutically acceptable salt thereof:

The preparation and properties of selective HDAC 1/2 inhibitors according to Formula III, as well as Compound J, are provided in U.S. Patent Application No. 14/069,741, the entire contents of which are incorporated herein by reference.

In another embodiment, the HDAC inhibitor is 4-acetamido-N-(2-amino-5- fluorophenyl)benzamide (Compound F), or a pharmaceutically acceptable salt thereof.

COMPOUND F

4-acetamido-N-(2-amino-5-fluorophenyl)benzamide

IC 50 (nM) HDAC1 = 153 HDAC2 = 479 HDAC3 = 106

The preparation and properties of the HDAC inhibitor Compound F are provided in International Patent Application No. PCT/US2013/052572, the entire contents of which are incorporated herein by reference.

In some embodiments, the HDACl/2/6-specific inhibitor is a compound of Formula

IV:

(IV)

or a pharmaceutically acceptable salt thereof,

wherein,

Pv x is independently selected from the group consisting of -CO 2 R 1 , and - C(0)N(R 1 ) 2 ;

R y is selected from the group consisting of H, Ci_6-alkyl, Ci_6-alkoxy, halo, -OH, - N0 2 , -CN, -NH 2 , -C(0)R 1 , -CO2R 1 , and -C(0)N(R 1 ) 2 ;

each R 1 is, independently for each occurrence, selected from the group consisting of H, Ci_6-alkyl, C3-8-cycloalkyl, C3_7-heterocycloalkyl, aryl, heteroaryl, Ci_6-alkyl-cycloalkyl, Ci_6-alkyl-heterocycloalkyl, Ci_6-alkyl-aryl, and Ci_6-alkyl-heteroaryl; and

R z is selected from the group consisting of Ci_6-alkyl, C3-8-cycloalkyl, C3-7- heterocycloalkyl, aryl, and heteroaryl. Compounds of Formula IV are represented by, but not limited to, Compound G, or a pharmaceutically acceptable salt thereof.

The preparation and properties of HDACl/2/6 specific inhibitors according to Formula IV are provided in International Application No. PCT/US2014/059863, the entire contents of which are incorporated herein by reference.

In some embodiments, the compounds described herein are unsolvated. In other embodiments, one or more of the compounds are in solvated form. As known in the art, the solvate can be any of pharmaceutically acceptable solvent, such as water, ethanol, and the like.

Combinations/Pharmaceutical Combinations

Provided herein are combinations for the treatment of leukemia in a subject in need thereof. Provided in some embodiments are combinations comprising a histone deacetylase (HDAC) inhibitor and azacitidine for the treatment of leukemia (e.g., AML) in a subject in need thereof.

In some embodiments of the combinations, the HDAC inhibitor is an HDAC6- specific inhibitor. In specific embodiments, the HDAC6-specific inhibitor is a compound of Formula I:

(I)

or a pharmaceutically acceptable salt thereof. In preferred embodiments, the compound of Formula I is:

or a pharmaceutically acceptable salt thereof.

In other preferred embodiments, the compound of Formula I is:

or a pharmaceutically acceptable salt thereof.

In other specific embodiments, the HDAC6-specific inhibitor is a compound of Formula II:

(II)

or a pharmaceutically acceptable salt thereof.

In preferred embodiments, the compound of Formula II is:

or a pharmaceutically acceptable salt thereof.

In other preferred embodiments, the compound of Formula II is:

or a pharmaceutically acceptable salt thereof. In some embodiments of the combinations, the HDAC inhibitor is an HDAC 1/2- specific inhibitor. In specific embodiments, the HDACl/2-specific inhibitor is a compound of Formula III:

(III)

or a pharmaceutically acceptable salt thereof.

In preferred embodiments, the compound of Formula III is:

or a pharmaceutically acceptable salt thereof.

In another embodiment, the HDACl/2-specific inhibitor is the compound J:

or a pharmaceutically acceptable salt thereof.

In another embodiment, the H s the compound F:

or a pharmaceutically acceptable salt thereof.

In some embodiments of the combinations, the HDAC inhibitor is an HDAC 1/2/6- specific inhibitor. In other specific embodiments, the HDACl/2/6-specific inhibitor is a compound of Formula IV:

(IV)

or a pharmaceutically acceptable salt thereof.

In preferred embodiments, the compound of Formula IV is:

or a pharmaceutically acceptable salt thereof.

In some embodiments of the combinations, azacitidine may be the free base or a pharmaceutically acceptable salt thereof. See Cihak, "Biological effects of 5-azacytidine in eukaryotes", Oncology, vol.. 30(5), pp. 405-422 (1974). 5-azacytidine (also known as azacitidine and 4-amino-l- -D-ribofuranosyl-S-triazin-2(lH)-one; Nation Service Center designation NSC- 102816; CAS Registry Number 320-67-2) is sold under the trade name Vidaza for the treatment of myelodysplastic syndrome (MDS).

Although the compounds of Formulas I, II, III, IV, Compound F, and Compound J are depicted in their neutral forms, in some embodiments, these compounds are used in a pharmaceutically acceptable salt form. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.

Administration/Do se

In some embodiments, the HDAC inhibitor (a compound of Formulas I, II, III, IV, Compound F or Compound J) is administered simultaneously with azacitidine. Simultaneous administration typically means that both compounds enter the patient at precisely the same time. However, simultaneous administration also includes the possibility that the HDAC inhibitor and azacitidine enter the patient at different times, but the difference in time is sufficiently miniscule that the first administered compound is not provided the time to take effect on the patient before entry of the second administered compound. Such delayed times typically correspond to less than 1 minute, and more typically, less than 30 seconds. In one example, wherein the compounds are in solution, simultaneous administration can be achieved by administering a solution containing the combination of compounds. In another example, simultaneous administration of separate solutions, one of which contains the HDAC inhibitor and the other of which contains azacitidine, can be employed. In one example wherein the compounds are in solid form, simultaneous administration can be achieved by administering a composition containing the combination of compounds. Alternatively, simultaneous administration can be achieved by administering two separate compositions, one comprising the HDAC inhibitor and the other comprising azacitidine.

In other embodiments, the HDAC inhibitor and azacitidine are not administered simultaneously. In some embodiments, the HDAC inhibitor is administered before azacitidine. In other embodiments, azacitidine is administered before the HDAC inhibitor. In other embodiments, the first administered compound is provided time to take effect on the patient before the second administered compound is administered. Generally, the difference in time does not extend beyond the time for the first administered compound to complete its effect in the patient, or beyond the time the first administered compound is completely or substantially eliminated or deactivated in the patient.

In some embodiments, one or both of the HDAC inhibitor and azacitidine are administered in a therapeutically effective amount or dosage. A "therapeutically effective amount" is an amount of HDAC inhibitor (a compound of Formulas I, II, III, IV, Compound F or Compound J) or azacitidine that, when administered to a patient by itself, effectively treats leukemia. An amount that proves to be a "therapeutically effective amount" in a given instance, for a particular subject, may not be effective for 100% of subjects similarly treated for the disease or condition under consideration, even though such dosage is deemed a "therapeutically effective amount" by skilled practitioners. The amount of the compound that corresponds to a therapeutically effective amount is strongly dependent on the type of cancer, stage of the cancer, the age of the patient being treated, and other facts. In general, therapeutically effective amounts of these compounds are well-known in the art, such as provided in the supporting references cited above. In other embodiments, one or both of the HDAC inhibitor and azacitidine are administered in a sub-therapeutically effective amount or dosage. A sub-therapeutically effective amount is an amount of HDAC inhibitor (a compound of Formulas I, II, III, IV, Compound F or Compound J) or azacitidine that, when administered to a patient by itself, does not completely inhibit over time the biological activity of the intended target.

Whether administered in therapeutic or sub-therapeutic amounts, the combination of the HDAC inhibitor and azacitidine should be effective in treating a leukemia, e.g., AML. For example, a sub-therapeutic amount of a compound of azacitidine can be an effective amount if, when combined with a compound of Formulas I, II, III, IV, Compound F, or Compound J (HDAC inhibitor), the combination is effective in the treatment of leukemia. For example, a sub-therapeutic amount of a compound of azacitidine can be an effective amount if, when combined with a compound of Formulas I, II, III, Compound F, or

Compound J (HDAC inhibitor), the combination is effective in the treatment of leukemia, wherein the combination is administered at dosages that would not be effective when one or both of the compounds are administered alone, but which amounts are effective in

combination.

In some embodiments, the combination of compounds exhibits a synergistic effect (i.e., greater than additive effect) in the treatment of leukemia. In further embodiments, the combination of compounds exhibits a synergistic effect (i.e., greater than additive effect) in the treatment of acute myelogenous leukemia. The term "synergistic effect" refers to the action of two agents, such as, for example, an HDAC inhibitor and azacitidine, producing an effect, for example, slowing the symptomatic progression of cancer or symptoms thereof, which is greater than the simple addition of the effects of each drug administered alone. A synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid- Emax equation (Holford, N. H. G. and Scheiner, L. B., Clin. Pharmacokinet. 6: 429-453

(1981)), the equation of Loewe additivity (Loewe, S. and Muischnek, H., Arch. Exp. Pathol Pharmacol. 114: 313-326 (1926)) and the median-effect equation (Chou, T. C. and Talalay, P., Adv. Enzyme Regul. 22: 27-55 (1984)). Each equation referred to above can be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively. In preferred embodiments provided herein are combinations and methods that include an HDAC inhibitor of Formula I and azacitidine. Thus, in one embodiment, the

combinations and methods include Compound A and azacitidine. In another embodiment, the combinations and methods include Compound B and azacitidine. In other preferred embodiments provided herein, the combinations and methods include an HDAC inhibitor of Formula II and azacitidine. Thus, in one embodiment, the combinations and methods include Compound C and azacitidine. In another embodiment, the combinations and methods include Compound D and azacitidine. In other preferred embodiments provided herein, the combinations and methods include an HDAC inhibitor of Formula III and azacitidine. Thus, in one embodiment, the combinations and methods include Compound E and azacitidine. In another preferred embodiment provided herein, the combinations and methods include the HDAC inhibitor Compound J and azacitidine. In other preferred embodiments provided herein, the combinations and methods include the HDAC inhibitor Compound F and azacitidine. In other preferred embodiments provided herein, the combinations and methods include an HDAC inhibitor of Formula IV and azacitidine. Thus, in one embodiment, the combinations and methods include Compound G and azacitidine.

In different embodiments, depending on the combination and the effective amounts used, the combination of compounds can inhibit leukemia growth, achieve leukemia stasis, or even achieve substantial or complete leukemia regression.

While the amounts of an HDAC inhibitor and azacitidine should result in the effective treatment of leukemia, the amounts, when combined, are preferably not excessively toxic to the patient (i.e., the amounts are preferably within toxicity limits as established by medical guidelines). In some embodiments, either to prevent excessive toxicity and/or provide a more efficacious treatment of leukemia, a limitation on the total administered dosage is provided. Typically, the amounts considered herein are per day; however, half-day and two-day or three-day cycles also are considered herein.

Different dosage regimens may be used to treat leukemia. In some embodiments, a daily dosage, such as any of the exemplary dosages described above, is administered once, twice, three times, or four times a day for three, four, five, six, seven, eight, nine, or ten days. Depending on the stage and severity of the cancer, a shorter treatment time (e.g., up to five days) may be employed along with a high dosage, or a longer treatment time (e.g., ten or more days, or weeks, or a month, or longer) may be employed along with a low dosage. In some embodiments, a once- or twice-daily dosage is administered every other day. In some embodiments, each dosage contains both an HDAC inhibitor and azacitidine to be delivered as a single dosage, while in other embodiments each dosage contains an HDAC inhibitor or azacitidine to be delivered as separate dosages.

Compounds of Formulas I, II, III, IV, Compound F, or Compound J, or their pharmaceutically acceptable salts or solvate forms, in pure form or in an appropriate pharmaceutical composition, can be administered via any of the accepted modes of administration or agents known in the art. The compounds can be administered, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally. The dosage form can be, for example, a solid, semi- so lid, lyophilized powder, or liquid dosage forms, such as for example, tablets, pills, soft elastic or hard gelatin capsules, powders, solutions, suspensions, suppositories, aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages. A particular route of administration is oral, particularly one in which a convenient daily dosage regimen can be adjusted according to the degree of severity of the disease to be treated.

As discussed above, the HDAC inhibitor and azacitidine pharmaceutical combination can be administered in a single unit dose or separate dosage forms. Accordingly, the phrase "pharmaceutical combination" includes a combination of two drugs in either a single dosage form or separate dosage forms, i.e., the pharmaceutically acceptable carriers and excipients described throughout the application can be combined with an HDAC inhibitor and azacitidine in a single unit dose, as well as individually combined with an HDAC inhibitor and azacitidine when these compounds are administered separately.

Auxiliary and adjuvant agents may include, for example, preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents.

Prevention of the action of microorganisms is generally provided by various antibacterial and antifungal agents, such as, parabens, chlorobutanol, phenol, sorbic acid, and the like. Isotonic agents, such as sugars, sodium chloride, and the like, may also be included. Prolonged absorption of an injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. The auxiliary agents also can include wetting agents, emulsifying agents, pH buffering agents, and antioxidants, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylated hydroxytoluene, and the like. Solid dosage forms can be prepared with coatings and shells, such as enteric coatings and others well-known in the art. They can contain pacifying agents and can be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds also can be in

microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., the HDAC inhibitors or azacitidine described herein, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1 ,3- butyleneglycol, dimethyl formamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol,

polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.

Generally, depending on the intended mode of administration, the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of the compounds described herein, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a pharmaceutically acceptable excipient. In one example, the composition will be between about 5% and about 75% by weight of a compound described herein, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.

Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art. Reference is made, for example, to Remington's Pharmaceutical Sciences, 18th Ed. (Mack Publishing Company, Easton, Pa., 1990).

Methods

Provided herein are methods for treating leukemia in a subject in need thereof comprising administering to the subject a pharmaceutical combination provided herein.

Further provided herein are methods for treating leukemia in a subject in need thereof comprising administering to the subject an HDAC inhibitor. Thus, provided herein are methods for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a combination comprising an HDAC inhibitor and azacitidine, or alternatively administering to the subject a therapeutically effective amount of an HDAC inhibitor. In a preferred embodiment of the methods provided herein, the leukemia is acute myelogenous leukemia. In another preferred embodiment of the methods provided herein, the HDAC inhibitor is an HDAC6-specific, H D AC 1 2-spec i fie, or HDAC 1 /2/6-specific inhibitor. In another preferred embodiment of the methods provided herein, the HDAC inhibitor is a compound of Formula I, Formula I I, Formula I I I or Formula IV. In another preferred embodiment of the methods provided herein, the HDAC inhibitor is Compound A, Compound B, Compound C, Compound D, Compound E, Compound F, Compound G, Compound H or Compound J.

Also provided herein are methods for treating a CD 1 l b-expressing cancer in a subject in need thereof comprising administering to the subject a pharmaceutical combination provided herein. Further provided herein are methods for treating a CD 1 l b-expressing cancer in a subject in need thereof comprising administering to the subject an HDAC inhibitor. Thus, provided herein are methods for treating a CD 1 l b-expressing cancer in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a combination comprising an HDAC inhibitor and azacitidine, or alternatively administering to the subject a therapeutically effective amount of an HDAC inhibitor. In another preferred embodiment of the methods provided herein, the H DAC inhibitor is an HDAC6-specific, HDAC 1 /2-specific, or HDACl/2/6-specific inhibitor. In another preferred embodiment of the methods provided herein, the HDAC inhibitor is a compound of Formula I, Formula I I, Formula I I I or Formula IV. In another preferred embodiment of the methods provided herein, the HDAC inhibitor is Compound A, Compound B, Compound C,

Compound D, Compound E, Compound F, Compound G, Compound H or Compound J.

The subject considered herein is typically a human. However, the subject can be any mammal for which treatment is desired. Thus, the methods described herein can be applied to both human and veterinary applications.

The terms "treating" or "treatment" indicates that the method has, at the least, mitigated abnormal cellular proliferation. For example, the method can reduce the rate of leukemia growth in a patient, or prevent the continued growth or spread of the leukemia, or even reduce the overall reach of leukemia.

In one embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDAC6-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2/6-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula II, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula III, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula IV, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound A, or a pharmaceutically acceptable salt thereof, and azacitidine, or a

pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound B, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound C, or a pharmaceutically acceptable salt thereof, and azacitdine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound D, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound E, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound F, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound G, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound H, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound J, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In one embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDAC6-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of an HDACl/2-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2/6-specific inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula II, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula III, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula IV, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound A, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound B, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound C, or a pharmaceutically acceptable salt thereof, and azacitdine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound D, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound E, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound F, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound G, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound H, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound J, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

In one embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDAC6-specific inhibitor, or a pharmaceutically acceptable salt thereof. In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2-specific inhibitor, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2/6-specific inhibitor, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula II, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula III, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound of Formula IV, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound A, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound B, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound C, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound D, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound E or a pharmaceutically acceptable salt thereof. In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound F or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound G or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound H, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound J, or a pharmaceutically acceptable salt thereof.

In one embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of an HDAC6-specific inhibitor, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of an HDACl/2-specific inhibitor, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, provided herein is a method for treating acute

myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDACl/2/6-specific inhibitor, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula II, or a pharmaceutically acceptable salt thereof. In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula III, or a pharmaceutically acceptable salt thereof.

In another embodiment, provided herein is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a

therapeutically effective amount of a compound of Formula IV, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound A, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound B, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound C, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound D, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound E, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound F, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound G, or a pharmaceutically acceptable salt thereof.

In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound H, or a pharmaceutically acceptable salt thereof. In another embodiment is a method for treating acute myelogenous leukemia in a subject in need thereof comprising administering to the subject a therapeutically effective amount of Compound J, or a pharmaceutically acceptable salt thereof.

Also provided herein are methods for inhibiting migration and/or invasion of leukemia cells. In particular, provided herein are methods for inhibiting migration and/or invasion of leukemia cells in a subject in need thereof. Specifically, provided herein are methods for inhibiting migration and invasion of leukemia cells, or both, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of an HDAC inhibitor of Formulas I, II, III, IV, Compound A, Compound B, Compound C, Compound D, Compound E, Compound F, Compound G, Compound H, or Compound J. In an embodiment, the HDAC inhibitor is Compound J, or a pharmaceutically acceptable salt thereof.

Provided herein are methods for decreasing cell viability of cancer cells by administering a combination comprising an HDAC inhibitor and azacitidine. In an embodiment, the HDAC inhibitor is Compound J, or a pharmaceutically acceptable salt thereof.

Also provided herein are methods for inducing differentiation of cancer cells by administering a combination comprising an HDAC inhibitor and azacitidine. In an embodiment, the H D AC inhibitor is Compound J, or a pharmaceutically acceptable salt thereof.

Also provided herein are methods for inducing apoptosis of cancer cells by administering a combination comprising an HDAC inhibitor and azacitidine. In an embodiment, the HDAC inhibitor is Compound J, or a pharmaceutically acceptable salt thereof.

Kits

In other embodiments, kits are provided. Kits provided herein include package(s) comprising compounds or compositions provided herein. In some embodiments, kits comprise an HDAC inhibitor, or a pharmaceutically acceptable salt thereof, and azacitidine, or a pharmaceutically acceptable salt thereof.

The phrase "package" means any vessel containing compounds or compositions presented herein. In some embodiments, the package can be a box or wrapping. Packaging materials for use in packaging pharmaceutical products are well-known to those of skill in the art. Examples of pharmaceutical packaging materials include, but are not limited to, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.

The kit can also contain items that are not contained within the package, but are attached to the outside of the package, for example, pipettes.

Kits can further contain instructions for administering compounds or compositions provided herein to a patient. Kits also can comprise instructions for approved uses of compounds herein by regulatory agencies, such as the United States Food and Drug

Administration. Kits can also contain labeling or product inserts for the compounds. The package(s) and/or any product insert(s) may themselves be approved by regulatory agencies. The kits can include compounds in the solid phase or in a liquid phase (such as buffers provided) in a package. The kits can also include buffers for preparing solutions for conducting the methods, and pipettes for transferring liquids from one container to another.

EXAMPLES

Examples have been set forth below for the purpose of illustration and to describe certain specific embodiments provided herein. However, the scope of the claims is not to be in any way limited by the examples set forth herein. Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods provided herein may be made without departing from the spirit provided herein and the scope of the appended claims. Definitions of the variables in the structures in the schemes herein are commensurate with those of corresponding positions in the formulae presented herein.

The synthesis of the compounds of Formula I (Compounds A and B) is provided in PCT/US2011/021982, which is incorporated herein by reference in its entirety. The synthesis of compounds of Formula II (Compounds C and D) is provided in PCT/US2011/060791, which is incorporated herein by reference in its entirety. The synthesis of compounds of Formula III, as well as Compound J is provided in U.S. Application No. 14/069,741, which is incorporated herein by reference in its entirety. The synthesis of compounds of Formula IV (e.g., Compound G) is provided in International Application No. PCT/US2014/059863, which is incorporated herein by reference in its entirety. Example 1: Synthesis of 2-(diphenylamino)-N- -(hydroxyamino)-7-oxoheptyl)pyrimidine-5-carboxamide (Compound A)

Synthesis of Intermediate 2: A mixture of aniline (3.7 g, 40 mmol), compound 1 (7.5 g, 40 mmol), and K 2 C0 3 (11 g, 80 mmol) in DMF (100 ml) was degassed and stirred at 120 0 C under N 2 overnight. The reaction mixture was cooled to r.t. and diluted with EtOAc (200 ml), then washed with saturated brine (200 ml x 3). The organic layers were separated and dried over Na 2 S0 4 , evaporated to dryness and purified by silica gel chromatography

(petroleum ethers/EtOAc = 10/1) to give the desired product as a white solid (6.2 g, 64 %).

Synthesis of Intermediate 3: A mixture of compound 2 (6.2 g, 25 mmol), iodobenzene (6.12 g, 30 mmol), Cul (955 mg, 5.0 mmol), Cs 2 C0 3 (16.3 g, 50 mmol) in TEOS (200 ml) was degassed and purged with nitrogen. The resulting mixture was stirred at 140 0 C for 14 hrs. After cooling to r.t., the residue was diluted with EtOAc (200 ml). 95% EtOH (200 ml) and NH 4 F-H 2 0 on silica gel [50g, pre-prepared by the addition of NH 4 F (lOOg) in water (1500 ml) to silica gel (500g, 100-200 mesh)] was added, and the resulting mixture was kept at r.t. for 2 hrs. The solidified materials were filtered and washed with EtOAc. The filtrate was evaporated to dryness and the residue was purified by silica gel chromatography (petroleum ethers/EtOAc = 10/1) to give a yellow solid (3 g, 38%).

Synthesis of Intermediate 4: 2N NaOH (200 ml) was added to a solution of compound 3 (3.0 g, 9.4 mmol) in EtOH (200 ml). The mixture was stirred at 60 0 C for 30min. After evaporation of the solvent, the solution was neutralized with 2N HC1 to give a white precipitate. The suspension was extracted with EtOAc (2 x 200 ml), and the organic layers were separated, washed with water (2 x 100 ml), brine (2 x 100 ml), and dried over Na 2 S0 4 . Removal of the solvent gave a brown solid (2.5 g, 92 %).

Synthesis of Intermediate 6: A mixture of compound 4 (2.5 g, 8.58 mmol), compound 5 (2.52 g, 12.87 mmol), HATU (3.91 g, 10.30 mmol), and DIPEA (4.43 g, 34.32 mmol) was stirred at r.t. overnight. After the reaction mixture was filtered, the filtrate was evaporated to dryness and the residue was purified by silica gel chromatography (petroleum ethers/EtOAc = 2/1) to give a brown solid (2 g, 54 %).

Synthesis of 2-(diphenylamino)-N-(7-(hydroxyamino)-7-oxoheptyl)pyrimidine -5- carboxamide (Compound A): A mixture of the compound 6 (2.0 g, 4.6 mmol), sodium hydroxide (2N, 20 mL) in MeOH (50 ml) and DCM (25 ml) was stirred at 0 0 C for 10 min. Hydroxylamine (50%) (10 ml) was cooled to 0 0 C and added to the mixture. The resulting mixture was stirred at r.t. for 20 min. After removal of the solvent, the mixture was neutralized with 1M HC1 to give a white precipitate. The crude product was filtered and purified by pre-HPLC to give a white solid (950 mg, 48%).

Example 2: Synthesis of 2-((2-chlorophenyl)(phenyl)amino)- -(7-(hydroxyamino)-7-oxoheptyl)pyrimidine-5-carboxamide (Compound B)

Synthesis of Intermediate 2: See synthesis of intermediate 2 in Example 1.

Synthesis of Intermediate 3: A mixture of compound 2 (69.2 g, 1 equiv.), l-chloro-2- iodobenzene (135.7 g, 2 equiv.), Li 2 C0 3 (42.04 g, 2 equiv.), K 2 C0 3 (39.32 g, 1 equiv.), Cu (1 equiv. 45 μιη) in DMSO (690 ml) was degassed and purged with nitrogen. The resulting mixture was stirred at 140 0 C. Work-up of the reaction gave compound 3 at 93 % yield.

Synthesis of Intermediate 4: See synthesis of intermediate 4 in Example 1.

Synthesis of Intermediate 6: See synthesis of intermediate 6 in Example 1. Synthesis of 2-((2-chlorophenyl)(phenyl)amino)-N-(7-(hydroxyamino)-7- oxoheptyl)pyrimidine-5-carboxamide (Compound B): See synthesis of Compound A in Example 1.

Example 3: Synthesis of 2-((l-(3-fluorophenyl) cyclohexyl)amino)-N-hydroxypyrimidine-5-carboxamide (Compound C)

Synthesis of Intermediate 2: To a solution of compound 1 (100 g, 0.74 mol) in dry DMF (1000 ml) was added 1,5-dibromopentane (170 g, 0.74 mol). NaH (65 g, 2.2 eq) was added dropwise while the reaction was cooled in an ice bath. The resulting mixture was vigorously stirred overnight at 50 °C. The suspension was carefully quenched with ice water and extracted with ethyl acetate (3 x 500 ml). The combined organic layers were concentrated to afford the crude product, which was purified by flash column chromatography to give compound 2 as pale solid (100 g, 67%).

Synthesis of Intermediate 3: A solution of compound 2 (100 g, 0.49 mol) in PPA (500 ml) was heated at 110 °C for about 5-6 hours. After completion, the resulting mixture was carefully adjusted to a pH of about 8-9 with sat.NaHC0 3 solution. The resulting precipitate was collected and washed with water (1000 ml) to afford compound 3 as white solid (95 g, 87%).

Synthesis of Intermediate 4: To a solution of compound 3 (95 g, 0.43 mol) in n- BuOH (800 ml) was added NaCIO (260 ml, 1.4 eq). 3N NaOH (400 ml, 2.8 equiv.) was then added at 0 °C and the reaction was stirred overnight at r.t. The resulting mixture was extracted with EA (2 x 500 ml), and the combined organic layers washed with brine. The solvent was removed in vacuo to afford the crude product which was further purified by treatment with HC1 salt to yield compound 4 as a white powder (72 g, 73%). Synthesis of Intermediate 6: To a solution of compound 4 (2.29 g 10 mmol) in dioxane (50 ml) was added compound 5 (1.87 g, 1.0 equiv.) and DIPEA (2.58 g, 2.0 equiv.). The mixture was heated overnight at 110-120 °C. The resulting mixture was directly purified on silica gel column to afford the coupled product, compound 6, as a white solid (1.37 g, 40%).

Synthesis of 2-((l-(3-fluorophenyl)cyclohexyl)amino)-N-hydroxypyrimidine- 5- carboxamide (Compound C):

To a solution of compound 6 (100 mg, 0.29 mmol) in MeOH/DCM(10 ml, 1 : 1) was added 50% NH 2 OH in water (2 ml, excess). Sat. NaOH in MeOH (2 ml, excess) was then added at 0 °C and the reaction was stirred for 3-4 hours. After completion, the resulting mixture was concentrated and acidified with 2N HC1 to reach a pH of 4-5. The precipitate was collected and washed with water (10 ml) to remove excess NH 2 OH. Drying the precipitate afforded 2-((l-(3-fluorophenyl)cyclohexyl)amino)-N-hydroxypyrimidine- 5- carboxamide as a white powder (70 mg, 73%).

Example 4: Synthesis of N-hydroxy-2- ((l-phenylcyclopropyl)amino)pyrimidine-5-carboxamide (Compound D)

Synthesis of Intermediate 2: A solution of compound 1, benzonitrile, (250 g, 1.0 equiv.), and Ti(OiPr) 4 (1330 ml, 1.5 equiv.) in MBTE (3750 ml) was cooled to about -10 to - 5 °C under a nitrogen atmosphere. EtMgBr (1610 ml, 3.0M, 2.3 equiv.) was added dropwise over a period of 60 min., during which the inner temperature of the reaction was kept below 5 °C. The reaction mixture was allowed to warm to 15-20 °C for 1 hr. BF 3 -ether (1300 ml, 2.0 equiv.) was added dropwise over a period of 60 min., while the inner temperature was maintained below 15 °C. The reaction mixture was stirred at 15-20 °C for 1-2 hr. and stopped when a low level of benzonitrile remained. IN HC1 (2500 ml) was added dropwise while maintaining the inner temperature below 30 °C. NaOH (20%, 3000 ml) was added dropwise to bring the pH to about 9.0, while still maintaining a temperature below 30 °C. The reaction mixture was extracted with MTBE (3 L x 2) and EtOAc (3 L x 2), and the combined organic layers were dried with anhydrous Na 2 S0 4 and concentrated under reduced pressure (below 45 °C) to yield a red oil. MTBE (2500 ml) was added to the oil to give a clear solution, and upon bubbling with dry HCl gas, a solid precipitated. This solid was filtered and dried in vacuum yielding 143 g of compound 2.

Synthesis of Intermediate 4: Compound 2 (620 g, 1.0 equiv) and DIPEA (1080 g, 2.2 equiv. were dissolved in NMP (3100 ml) and stirred for 20 min. Compound 3 (680 g, 1.02 equiv.) was added and the reaction mixture was heated to about 85-95 °C for 4 firs. The solution was allowed to slowly cool to r.t. This solution was poured onto H 2 0 (20 L) and much of the solid was precipitated out from the solution with strong stirring. The mixture was filtered and the cake was dried under reduced pressure at 50 °C for 24 hr., yielding 896 g of compound 4 (solid, 86.8%).

Synthesis of N-hydroxy-2-((l-phenylcyclopropyl)amino)pyrimidine-5-carboxa mide (Compound D): A solution of MeOH(1000 ml) was cooled to about 0-5 °C with stirring.

NH 2 OH HCl (1107 g, 10 equiv.) was added, followed by careful addition of NaOCH 3 (1000 g, 12.0 equiv.) The resulting mixture was stirred at 0-5 °C for one hr, and was filtered to remove the solid. Compound 4 (450 g, 1.0 equiv.) was added to the reaction mixture in one portion, and stirred at 10 °C for two hours until compound 4 was consumed. The reaction mixture was adjusted to a pH of about 8.5-9 through addition of HCl (6N), resulting in precipitation. The mixture was concentrated under reduced pressure. Water (3000 ml) was added to the residue with intense stirring and the precipitate was collected by filtration. The product was dried in an oven at 45 °C overnight (340 g, 79% yield).

Example 5: Synthesis of N-(2-amino-5-(thiophen-2-yl)phenyl)- -cyclopropyl-l-(2-morpholinoethyl)-lH-indole-5-carboxamide (Compound E)

Experimental Procedure

Step 1: To a solution of compound 1 in DCE was added POBr 3 and imidazole. The reaction was stirred at 80°C overnight. Water and DCM were added to the reaction, and the organic layer was separated, washed with brine, and dried under reduced pressure to give compound 2.

Step 2: To a solution of compound 2 in DMSO was added compound a and KOH.

The resulting reaction mixture was stirred at 45 °C for 4 h, quenched with H 2 0, and extracted with EA. The combined organic layers were purified by gel chromatography to yield the desired product, compound 3.

Step 3: A mixture of compound 3, cyclopropyl boronic acid, Pd(OAc) 2 ,

tricyclohexylphosphine, and K 3 P0 4 in toluene and water was stirred at 100°C under N 2 atmosphere overnight. The mixture was cooled, filtered, and concentrated to obtain a residue, which was purified by Prep-TLC to get compound 4.

Step 4: A mixture of compound 4 and NaOH in EtOH and THF was stirred at 60 °C for 5 h. The mixture was concentrated to obtain a residue, to which was added aq. sat. citric acid and extracted with EA. The organic layers were separated, dried, filtered and concentrated to obtain compound 5.

Step 5: A mixture of compound 5, tert-butyl 2-amino-4-(thiophen-2- yl)phenylcarbamate, HO AT, EDCI, and DIPEA in DMF was stirred at 55°C for overnight.

Water was added to the mixture, and extracted with EA. The organic layers were separated, dried, filtered, and concentrated to get a residue, which was purified by Prep-TLC to afford compound 6.

Step 6: To a solution of compound 6 in DCM was added TFA and stirred at r.t. for 1 h. The mixture was concentrated to obtain a residue, which was purified by Prep-HPLC to afford compound 7. 1H NMR (500 MHz, DMSO) δ 9.63 (s, 1H), 8.16 (s, 1H), 7.79 - 7.73 (m, 1H), 7.51 (d, J = 2.1 Hz, 2H), 7.36 (d, J = 5.1 Hz, 1H), 7.29 (dd, J = 8.3, 2.1 Hz, 1H), 7.25 (d, J = 3.5 Hz, 1H), 7.05 (dd, J = 5.0, 3.6 Hz, 1H), 6.82 (d, J = 8.3 Hz, 1H), 6.24 (s, 1H), 5.12 (s, 2H), 4.43 (s, 2H), 3.57 (s, 5H), 2.77 - 2.58 (m, 2H), 2.09 (s, 1H), 1.02 (d, J = 8.0 Hz, 2H), 0.76 (d, J = 4.4 Hz, 2H).LCMS: m/z = 487.2 (M+H)+.

Example 6: Synthesis of N-hydroxy-2-((4-phenyl-l- (phenylcarbamoyl)piperidin-4-yl)amino)pyrimidine-5-carboxami de (Compound G)

Compound G

Step 1: To a solution of compound 8 (85 mg, 0.26 mmol) in THF (4 mL) was added isocyanatobenzene (46 mg, 0.39 mmol), DIPEA (0.2 ml) at r.t. The reaction was stirred for 2 firs, and subsequently concentrated in vacuo to give compound 9 (80 g, yield: 69%).

Step 2: To a solution of compound 9 (80 mg, 0.18 mmol) in MeOH (3 mL) and DCM (1 ml) at 0 C was added NH 2 OH (0.2 ml). The reaction was stirred for 10 mins, at which time NaOH/MeOH (0.4 ml) was added. The reaction was stirred for 2 firs. The resulting reaction mixture was concentrated, adjusted to pH=5 using 2N HCl, extracted with EA (10 ml), and purified by Pre-HPLC to afford N-hydroxy-2-((4-phenyl-l-

(phenylcarbamoyl)piperidin-4-yl)amino)pyrimidine-5-carbox amide (14 mg, 17%). 1H NMR (500 MHz, DMSO) δ 10.83 (s, 1H), 8.96 (s, 1H), 8.60 (s, 1H), 8.49 (s, 2H), 8.37 (s, 1H), 8.20 (s, 1H), 7.47-7.46 (d, J= 7.6 Hz, 2H), 7.41-7.39 (d, J= 7.4 Hz, 2H), 7.29-7.26 (t, J= 7.7 Hz, 2H), 7.23-7.20 (m, J= 7.7 Hz, 2H), 7.18-7.15 (t, J= 7.3 Hz, 1H), 6.92 (t, J= 7.3 Hz, 1H), 4.03 (d, J= 13.2 Hz, 2H), 3.13 (t, J= 12.1 Hz, 2H), 2.64 (d, J= 13.0 Hz, 2H), 1.90 (t, J = 11.0 Hz, 2H). LCMS: m/z = 433 (M+H) + . Example 7: Synthesis of N-(2-amino-5- (thiophen-2-yl)phenyl)-2-(piperazin-l-yl)quinoline-6-carboxa mide Compound J

The preparation of Compound J is provided in U.S. Patent Application No.

14/069,741, which is summarized below.

Reaction Scheme:

Experimental Procedure

Step 1: A mixture of compound 1 (10 g, 0.53 mol) and m-CPBA (18.4 g, 0.106 mol) in DCM (50 ml) is stirred at r.t. overnight. Aq. NaHC0 3 (40 ml, saturated) is added to the reaction mixture and stirred for 30 min. The organic layer is separated, dried, filtered and concentrated to obtain a residue, which can be re-crystallized in ethyl acetate (5 ml) to afford compound 2 as a light yellow solid.

Step 2: To a solution of compound 2 (4.0 g, 0.020) and DMF (8 ml) in DCM is added SOCl 2 (8 ml) slowly at 0°C and stirred at r.t. for 5 h. The resulting mixture is concentrated to obtain a residue, and DCM (50 ml) with Aq. NaHC0 3 (saturated, 20 ml) is added and stirred for 30 min. The organic layer is separated and concentrated to obtain a residue, which is purified by silica gel chromatography to afford compound 3 as a white solid.

Step 3: A mixture of compound 3 (10 g, 0.045 mol), Cul (10 g, 0.53 mol), N-boc- piperazine (25 g, 0.135 mol) and K 2 C0 3 (18.6g, 0.135 mol) in DMSO (120 ml) is stirred at 100°C overnight. Upon completion, as monitored by TLC (thin- layer chromatography), 300 ml of EA (ethyl acetate) is added, followed by filtration. Concentration of the mixture yields a residue, to which water (300 ml) and Aq. Citric acid (saturated, 30 ml) are added. Stirring at r.t. for 30 min., followed by filtration yields compound 4 as a yellow solid that can be used in the next step without purification.

Step 4: A mixture of compound 4 (18 g, crude) and 2M NaOH (50 ml) in EtOH (100 ml) and THF (100 ml) is stirred at 70°C for 4 h. TLC can be used to monitor the reaction. The reaction mixture is concentrated to a residue, to which water (300 ml) and aq. sat. citric acid (40 ml) are added. Subsequent filtration yields compound 5 as a yellow solid.

Step 5: A mixture of compound 5 (1 equiv.), tert-butyl 2-amino-4-(thiophen-2- yl)phenylcarbamate (1 equiv.), HO AT (1.5 equiv.), EDCI (2 equiv.), and DIPEA (4 equiv.) in DMF is stirred at 55°C overnight. Water is added to the mixture, and extracted with EA. The organic layers are separated, dried, filtered, and concentrated to yield a residue, which can be purified by by Prep-TLC to afford compound 7.

Step 6: A mixture of compound 7 (95 mg 0.15 mmol) and TFA (2 ml) in 2 ml DCM is stirred at r.t. for 2 h. Evaporation of the solvent yields crude product which can be purified by HPLC to afford the white product, Compound J (19 mg, 30%). 1H NMR (500 MHz, DMSO) δ 9.79 (s, 1H), 8.42 (d, J= 1.8 Hz, 1H), 8.17 - 8.09 (m, 2H), 7.60 (d, J= 8.8 Hz, 1H), 7.51 (d, J= 2.0 Hz, 1H), 7.36 (dd, J= 5.1, 0.8 Hz, 1H), 7.33 - 7.28 (m, 2H), 7.25 (d, J = 3.5 Hz, 1H), 7.06 (dd, J= 5.0, 3.6 Hz, 1H), 6.83 (d, J= 8.3 Hz, 1H), 5.18 (s, 2H), 3.73 (s, 4H), 2.89 (s, 4H). LCMS: m/z = 430 (M+H) +

Example 8: HDAC enzyme assays

Compounds for testing are diluted in DMSO to 50 fold the final concentration and a ten point three fold dilution series is made. The compounds are diluted in assay buffer (50 mM HEPES, pH 7.4, 100 mM Kill, 0.001% Tween-20, 0.05% BASE, 20 μΜ TEC) to 6 fold their final concentration. The HDAC enzymes (purchased from BPS Biosciences) are diluted to 1.5 fold their final concentration in assay buffer. The dipeptide substrate and trypsin at 0.05 μΜ final concentration are diluted in assay buffer at 6 fold their final concentration. The final enzyme concentrations to use in these assays are 3.3 ng/ml (HDACl), 0.2 ng/ml (HDAC2), 0.08 ng/ml (HDAC3) and 2 ng/ml (HDAC6). The final substrate concentrations to use are 16 μΜ (HDACl), 10 μΜ (HDAC2), 17 μΜ (HDAC3) and 14 μΜ (HDAC6). Five μΐ of compound and 20 μΐ of enzyme are added to wells of a black, opaque 384 well plate in duplicate. Enzyme and compound are incubated together at room temperature for 10 min. Five μΐ of substrate is added to each well, the plate is shaken for 60 seconds and placed into a Victor 2 microliter plate reader. The development of fluorescence is monitored for 60 min. and the linear rate of the reaction is calculated. The IC 50 is determined using Graph Pad Prism by a four parameter curve fit. Example 9: Synergy of HDAC inhibitors and Azacitidine on AML cells

Inhibition of HDAC or DNMT (DNA methyltransferase) has been shown to be cytotoxic to AML cells. Different HDAC inhibitors (Compound A and Compound C are HDAC6 selective, Compound E is HDAC 1/2 selective, and Compound F is a control) and the DNMT inhibitor, azacitidine, were combined in an AML cell viability assay measured by a Cell Titer Glo Assay™. With the same amount of cultured AML cells in each well, serial dilutions of one compound were added into each row of the wells from left to right, and serial dilutions of the second compound were mixed into each column of these wells from top to bottom. Therefore, those AML cells on the testing plate were exposed to various

combinations of the two compounds at different concentrations. The viable cells in each well were measured after 72 hours incubation at 37°C, and the percentage of unviable cells was calculated and normalized to the total cells. These values were reported as Fa (Fractional Activity), in the range of 0 - 1.0, to reflect cytotoxicity of the testing compounds, alone or in combinations. Combination Index (CI) values were calculated using the software CalcuSyn to determine whether a combination was synergistic (CI<1.0), additive (CI=1.0), or antagonistic (CI>1.0). The CI values were plotted as a function of Fa, as shown in Figures 1A-D. In order to avoid any possible false positives due to experimental data variability, a combination was determined "synergistic" only when CI<0.7, as shown in the shaded area of each graph. From these results, it was concluded that the tested combinations have a synergistic cytotoxic effect on AML cells, based on the synergy data from the three tested AML cell lines: HL-60, Kasumi-3 and THP-1. HDAC 1/2 inhibition appears to have a more predominant effect than HDAC3 or HDAC6 because the most synergy was observed from the Compound E/azacitidine combination. These results are presented in Figures 1A-D, where the graph in the top left (Figure 1 A) shows data for azacitidine and Compound A in HL-60 cells, the graph in the top right (Figure IB) shows data for azacitidine and Compound C in HL-60 cells, the graph in the lower left (Figure 1C) shows data for azacitidine and Compound E in HL-60 cells, and the graph in the lower right (Figure ID) shows data for azacitidine and Compound F in HL-60 cells. Thus, the data in Figures 1A-D show that azacitidine shows significant synergistic cell killing with Compound A and other HDAC isoform inhibitors in AML cell lines (Kasumi-3, HL-60, and THP-1). The synergism is driven predominantly by HDAC 1/2 inhibition. Example 10: HDAC inhibition increases apoptosis and suppresses AML/ETO in AML

HDAC inhibition causing AML cell death (apoptosis) was measured by staining the AML cells with propidium iodide after the cells were exposed to different concentrations of the HDAC inhibitors. Four distinct cell populations, Gl, S, G2, and sub-Gl, were separated and quantified based on their propidium iodide staining patterns. The sub-Gl cells are those dying or dead cells, and the percentage of this population was a reflection of the tested compound cytotoxicity. Increased amount of sub-Gl cells as the function of increased concentrations of HDAC inhibitors, especially the HDACl/2 selective inhibitor Compound E, suggested HDAC 1/2 mediated AML cytotoxicity. These results are presented in Figures 2A-C, which shows data for the Kasumi-1 cell cycle at 72 hours. Figure 2 A shows data for Compound B, Figure 2B shows data for Compound G, and Figure 2C shows data for Compound E.

One type of AML has a signature chromosome translocation t(8:21) and therefore expression of a unique fusion protein AML1/ETO. This fusion protein has been reported as critical for AML cell growth, and the pan-HDAC inhibitor panobinostat is able to cause its loss in the AML cell line Kasumi-1, which has the t(8:21) translocation. In this study, Kasumi-1 cells were exposed to different concentrations of the HDAC inhibitor Compound A, or another HDAC 1/2/6 selective inhibitor Compound G (data not shown), for 24 hours. The whole cell lysates were separated by SDS-PAGE (SDS-polyacrylamide gel

electrophoresis) and transferred to a membrane (Western blot). The AML1/ETO fusion protein was detected on the membrane using an AML1 specific antibody. The results showed that Compound A and Compound G both decrease the amount of this fusion protein in a concentration dependent manner. These results are presented in Figure 2D. Example 11: Isoform Selective Histone Deacetylase (HDAC) Inhibitors

Synergize in Combination with Azacitidine in Acute Myeloid Leukemia (AML)

AML is a heterogeneous group of hematopoietic stem cell disorders characterized by defects in myeloid differentiation and increased proliferation of neoplastic hematopoietic precursor cells. Aberrant epigenetic regulation plays an important role in the pathogenesis of AML. The DNA methyltransferase inhibitor azacitidine was approved for the treatment of myelodysplasia syndrome, which frequently progresses to AML.

HDAC inhibitors are emerging as promising agents for the treatment of AML.

Isoform selective HDAC inhibitors have the potential to reduce the combination of drug toxicity and other side effects observed with non-selective inhibitors, while also realizing beneficial therapeutic effects. One example is ricolinostat (Compound A), a first-in-class orally available HDAC inhibitor that is 11 -fold selective for HDAC6, synergizes with bortezomib (Blood, 20[210]: 4061) and immunomodulatory agents (Quayle, et al, ASH, 2013) in preclinical models of multiple myeloma, and has thus far demonstrated an improved safety and tolerability profile in Phase I trials (Raje, et al, EHA, 2014).

This work evaluated the combinatorial efficacy of azacitidine and HDAC inhibitors selective for either HDAC6 or HDAC 1/2 on AML cells.

Time course studies demonstrated induction of differentiation, accumulation of cell cycle arrest, and initiation of apoptosis after prolonged exposure to HDAC inhibitors (see Figures 3-4).

Figures 3A-D show the single agent activity on viability in AML cell lines. Briefly, each of the following cell lines: HL-60, THP-1, MV-4-11, Kasumi-1, NB4, and MOLM-13 were exposed to increasing concentrations of either Compound B (Figure 3A), Compound A (Figure 3B), Compound E (Figure 3C), or azacitidine (Figure 3D) to determine their response to drug treatment. Compound B is about 10 times selective for HDAC6.

Compound A is about 10 times selective for HDAC6. Compound E is selective for

HDAC 1/2. The panel of cell lines was also treated with azacitidine to measure their sensitivity. Thus, the data in Figures 3A-D show that AML cell lines are sensitive to HDAC inhibition.

Figures 4A-F show the single agent activity on differentiation and apoptosis in AML cell lines. Briefly, the AML cell lines HL-60 (Figures 4A and 4D), Kasumi-1 (Figures 4B and 4E) and NB4 (Figures 4C and 4F) were treated with the indicated concentrations of compounds. In Figures 4A-C, surface levels of myeloid differentiation marker CDl lb were determined by FACS at 72 hours post-treatment. Compound B, Compound A, and

Compound E increased the percentage of CDl lb positive cells in all three cell lines.

Azacitidine increased CDl lb positive cells in HL-60 (Figure 4A) and Kasumi-1 cells

(Figure 4B) and had minimal effects in NB4 cells (Figure 4C). In Figures 4D-F, apoptosis was assessed by flow cytometry by measuring Annexin V binding and cellular permeability to propidium iodide at 96 hours post-treatment. The relative fraction of cells that were alive, in early apoptosis, in late apoptosis, or dead was then determined. Treatment with

Compound B, Compound A, and azacitidine resulted in an increase in apoptosis relative to control cells. Compound E induced apoptosis in HL-60 (Figure 4D) and Kasumi-1 (Figure 4E) cells, but had minimal effects in NB4 cells (Figure 4F). Thus, the data in Figures 4A-F show that treatment of AML cells with Compound B, Compound A, Compound E, and azacitidine induced differentiation and apoptosis.

Combining HDAC inhibitors with azacitidine led to synergistic induction of differentiation and apoptosis in AML cells in vitro (see Figures 5-7).

Figures 5A-F show the combination of HDAC inhibitors and azacitidine in the HL- 60 cell line. Briefly, cells were treated with DMSO, Compound B, Compound A, or

Compound E as a single agent or in combination with azacitidine for 96 hours. Surface levels of CD1 lb (Figures 5A, 5C, 5E) and apoptosis (Figures 5B, 5D, 5F) was assessed by flow cytometry, as in Figure 4. The combination of Compound B with azacitidine,

Compound A with azacitidine, and Compound E with azacitidine resulted in synergistic increases of CD1 lb positive cells (Figures 5A, 5C, 5E) and apoptotic cells (Figures 5B, 5D, 5F) compared to single agent treatment. Thus, the data in Figures 5A-F show that the treatment of HL-60 cells with Compound B, Compound A, or Compound E plus azacitidine significantly induced differentiation and apoptosis.

Figures 6A-F shows the combination of HDAC inhibitors and azacitidine in the Kasumi-1 cell line. Briefly, cells were treated with DMSO, Compound B, Compound A, or Compound E as a single agent or in combination with azacitidine at the indicated

concentrations. Surface levels of CD1 lb were determined 72 hours post-treatment (Figures 6A, 6C, 6E), and apoptosis was assessed 96 hours post-treatment (Figures 6B, 6D, 6F). The combination of Compound B, Compound A, or Compound E with azacitidine resulted in synergistic increases of CD1 lb positive cells (Figures 6A, 6C, 6E) and apoptotic cells

(Figures 6B, 6D, 6F) compared to single agent treatment. Thus, the data in Figures 6A-F show that the treatment of Kasumi-1 cells with Compound B, Compound A, or Compound E plus azacitidine significantly induced differentiation and apoptosis.

Figures 7A-F show the combination of HDAC inhibitors and azacitidine in the NB4 cell line. Briefly, cells were treated with DMSO, Compound B, Compound A, or Compound E as single agent or in combination with azacitidine at the indicated concentrations. Surface levels of CD1 lb were determined 72 hours post-treatment (Figures 7A, 7C, 7E), and apoptosis was assessed 96 hours post-treatment (Figures 7B, 7D, 7F). The combination of Compound B or Compound A with azacitidine resulted in synergistic increases of CD1 lb positive cells (Figures 7A, 7C). The combination of Compound B, Compound A, or Compound E with azacitidine resulted in synergistic increases of apoptotic cells compared to single agent treatment (Figures 7B, 7D, 7F). Thus, the data in Figures 7A-F show that the treatment of NB4 cells with Compound B, Compound A, or Compound E plus azacitidine significantly induced differentiation and apoptosis.

HDAC inhibitors selective for HDAC 1/2 showed the strongest cellular activities. Furthermore, HDAC inhibitors reduced the level of AMLl-ETO fusion protein, which is essential for the survival of cell lines carrying this fusion protein. The potential of the drug combination is being explored in animal models of AML and in primary AML cells.

Together, these findings provide support for the clinical evaluation of selective HDAC inhibitors in combination with azacitidine in AML patients.

Example 12: HDAC 1/2 Inhibition Reduces Cell Viability

The following HDAC inhibitors were used to evaluate the relationship between HDAC selectivity and the viability of AML cell lines upon exposure to the HDAC inhibitor:

The above panel includes an HDAC3 selective inhibitor, Compound H, which is described in U.S. App. No. 14/169,732, and is incorporated herein in its entirety:

Figures 8A-F show the single agent activity on viability in AML cell lines. Briefly, each of the following cell lines: HL-60, NB4, MV4-11, Kasumi-1, THP-1, and MOLM-13 were exposed to increasing doses of either Compound E (Figure 8A), Compound H (Figure 8B), Compound C (Figure 8C), Compound A (Figure 8D), Compound B (Figure 8E), and Compound G (Figure 8F) for 72 hours to confirm their sensitivity to HDAC inhibition. Viability was calculated as a percentage of control (DMSO treated cells). Growth inhibition curves were generated using GraphPad Prism 6. The IC 50 of Compound E is within its HDAC 1/2 selective range (Figure 8 A). The IC 50 of Compound A and Compound G has some inhibitory effects on HDAC 1/2 at their IC 50 values (Figures 8D and 8F). The IC 50 of Compound C and Compound H is beyond its selective range for HDAC6 and HDAC3, respectively, and likely has an inhibitory effect on HDACl/2 (Figures 8C and 8B). Together, these data indicate HDACl/2 inhibition reduces cell viability.

Example 13: HDAC 1/2 Inhibition is Sufficient to Induce Differentiation, Cell Cycle Arrest, and Apoptosis

Figures 9A-C shows treatment of MV4-11 with the indicated doses of compounds. Figure 9A shows surface levels of myeloid differentiation marker CDl lb, determined by FACS at 72h post-treatment. Compound E, Compound H, Compound A, and Compound G increased the percentage of CDl lb positive cells. Compound C had no effect on CDl lb positive cells. Figure 9B shows assessment of the cell cycle by flow cytometry after incorporation of EdU and staining with Far Red at 72h post-treatment. The distribution of cells among G0/G1 phase, G2/M phase, S phase and subGl phase was determined.

Compound E, Compound A, and Compound G induced cell cycle arrest. Figure 9C shows the assessment of apoptosis by flow cytometry via measuring Annexin V binding and cellular permeability to propidium iodide at 96h post-treatment. The relative fraction of cells that were live, in early apoptosis, in late apoptosis, or dead was then determined. Treatment with Compound E, Compound A, and Compound G resulted in an increase in apoptosis relative to control cells.

In summary, HDACl/2 inhibition is sufficient to induce differentiation, cell cycle arrest and apoptosis in AML cell lines. HDAC3 inhibition induces differentiation marker

CDl lb only (i.e., had no effect on cell cycle and apoptosis). Selective HDAC6 inhibition has no obvious impact or effect.

Example 14: HDAC 1/2 Inhibition Induces Differentiation and Apoptosis in AML cells Figures 10A-F show the treatment of the following AML cell lines: Kasumi-1

(Figures 10A and 10B), HL-60 (Figures IOC and 10D) and NB4 (Figures 10E and 10F), with indicated doses of compounds. Figures 10A, IOC, and 10E show surface levels of myeloid differentiation marker CDl lb determined by FACS at 72h post-treatment. Compound E and Compound A increased percentage of CDl lb positive cells in all three cell lines. Figures 10B, 10D, and 10F show the assessment of apoptosis by FACS (see, e.g., Figure 9C).

Treatment with Compound E and Compound A resulted in increased apoptosis relative to control cells. Further, Compound E and Compound A induced differentiation and apoptosis in a dose-dependent manner in all three cell lines described. Example 15: HDACl/2 Inhibition Synergizes with Azacitidine in HL-60 Cells

Figures 11 A-D show that combinations of HD AC 1/2 inhibition with azacitidine result in synergistic decreases in HL-60 cell viability. HL-60 cells were treated with increasing doses of azacitidine with Compound E (Figure 11 A) or with Compound A (Figure 1 IB) or with Compound H (Figure 11C) or with Compound C (Figure 1 ID), and cell viability was assessed at 72 hr by cell titer glo assay. The combination index (CI) and relative fraction affected (Fa) was determined at each dose level using CalcuSyn software. The measurement of CI values less than 1 (shaded region) strongly support a synergistic interaction between drugs.

Significant enhancement of azacitidine activity is observed in combination with HDACl/2 inhibition. Compound E showed the strongest synergistic interaction with azacitidine.

Example 16: HDACl/2 Inhibition Enhances Activity of Azacitidine

Figure 12A-F show the treatment of MV4-11 cells with azacitidine plus Compound E or plus Compound A or plus Compound B significantly induced differentiation and apoptosis. MV4-11 cells were treated with Compound E or with Compound A or with Compound B as single agent or in combination with azacitidine at indicated doses. Figures 12A, 12C, and 12E show surface levels of CD1 lb determined by FACS at 72h post- treatment. Figures 12B, 12D, and 12F show assessment of apoptosis by FACS (as in, e.g., Figure 9C) at 96h post-treatment.

Combination of Compound E with azacitidine, Compound A with azacitidine and Compound B with azacitidine resulted in further increase of percentage of CD1 lb positive cells and enhanced induction of apoptosis greater than either single agent. As described above, this example shows significant enhancement of azacitidine activity in combination with HD AC 112 inhibition.

Example 17: Compound A Enhanced Tumor Growth Inhibition by Azacitidine

Figures 13A-C show that treatment with Compound A plus azacitidine reduces tumor growth in vivo. Ncr nu/nu mice implanted with MV4-11 cells were treated with vehicle, azacitidine (5 mg/kg IV q3d), or azacitidine (5 mg/kg IV q3d) plus Compound A (50 mg/kg IP 5/2/5/2/5/2/5) for up to 4 weeks. (A) Tumor volume was measured twice weekly and the mean tumor volume ± SD is plotted. (B) Fold tumor volume change on day 19 relative to day 1 is plotted. (C) Survival curve was plotted. Single agent azacitidine reduced tumor growth and increased survival of MV4-11. This effect was further enhanced by addition of Compound A.

Example 18: HDACl/2 Inhibition Enhanced the Activity of Azacitidine in pPrimary AML Sample Colony Formation Assay

Figure 14A-E, shows that HDACl/2 inhibition alone and in combination with azacitidine reduces colony formation of primary AML patient samples. (A) 6 bone marrow samples derived from AML patients were cultured in methylcellulose-based medium and treated with increasing concentrations of Compound A, Compound J and azacitidine for 14 days when the colonies reach reasonable size. IC50 values for each drug are plotted. The median IC50 values for Compound A, Compound J and azacitidine are 9.76uM, 2.95uM and 8.1 luM, respectively. The relative potency of the three drugs are Compound J > azacitidine > Compound A. (B-E) Each bone marrow sample from AML patient was treated with increasing concentrations of azacitidine alone or in the presence of Compound J at luM or 3uM or Compound A at luM, 3uM or lOuM. IC50 values were plotted for each patient sample. For sample 4031113SH (B) and sample VMBM0007 (C), Compound J and

Compound A decreased azacitidine IC50 value, indicating a good combination effect of HDACl/2 inhibition with azacitidine on these primary AML cell growth. For sample 184090514 (D), Compound J at 3uM and Compound A at lOuM, the concentrations close to their IC50 values, significantly reduced azacitidine IC50 value, indicating a good

combination effect. For sample 103113SH (E), only Compound J at 3uM reduced azacitidine IC50. Together, Compound J is more potent on inhibiting primary AML cell growth than Compound A and azacytidine. Compound J at the concentrations close to or below its own IC50 value significantly reduced IC50 value of azacitidine on all 4 primary AML cell colony formation.

Example 19: Ex vivo Pharmacological

Profiling of Azacitidine, Compound A and Compound J

Figure 15A-C, shows that HDACl/2 inhibition alone and in combination with azacitidine inhibit proliferation of AML blast freshly derived from bone marrow of AML patients. (A-B), 5 bone marrow samples derived from AML patients were treated with increasing concentrations of azacitidine, Compound A and Compound J and live AML cells were quantified by flow cytometry at 96h. IC50 values (A) and AUC values (B) were plotted. The median IC50 values for Compound A, Compound J and azacitidine are 7.2uM, 0.6uM and 2. luM, respectively. The relative potency of the three drugs are Compound J > azacitidine > Compound A, consistent with the result in Figure 2. (C) 5 bone marrow samples derived from AML patients were treated with increasing doses of azacitidine with Compound J and live AML cells were quantified by flow cytometry at 96h. The combination index (Comb IDX) were calculated and median Comb IDX values were plotted. In 4 out of 5 samples, the Comb IDX value is less than 1, supporting a synergistic interaction between the two drugs on inhibiting proliferation of primary AML cells freshly derived from AML patients.

Example 20: Gene Expression Profiling

MV4-11 cells were plated at 2 x 10 5 cells/ml and treated with azacitidne at ΙμΜ, Compound E at ΙμΜ, Compound E at 2μΜ, azacitidine at ΙμΜ plus Compound E at ΙμΜ, azacitidine at ΙμΜ plus Compound E at 2μΜ for 24h and 48h. Cells were collected and R A isolated. R A samples were subjected to Affymetrix PrimeView Gene Expression profiling. Azacitidine at ΙμΜ and Compound E at 2μΜ at 48h were the focus of the initial data analysis. Molecular signatures were analyzed by GSEA

(http://www.broadinstitute.org/gsea/index.jsp). The genes and signatures that were upregulated by the single and combination treatment are significantly more than those that were downregulated, consistent with the mechanisms of the compounds. In order to identify pathways and/or genes that mediate the combinatorial effects of azacitidine with Compound E, signatures and genes that were upregulated by single agent and further upregulated by combination treatment were identified. Signatures including apoptosis and CEBPA pathway, a major transcription factor driving differentiation, are among the top pathways and/or genes identified. More than 60 genes including GATA2 and CD86 follow this expression pattern.

Example 21: Induction of GATA2 Expression in MV4-11 AML Cell Line

Figure 16. Treatment of Compound E plus azacitidine significantly induced Gata2 in MV4-11 cells. (A-B) MV4-11 cells were plated at 2 x 10 5 cells/ml at indicated doses for 48h and 72h. RNA was prepared and analyzed for GATA2 and GAPDH as internal control. Azacitidine at luM and Compound E at luM induced GATA2 level as single agent at 48h and 72h. Combination of azacitidine and Compound E further induced GATA2 expression at both time points. Example 22: Single Agent Activity in AML Cell Lines

Figure 17. Compound J reduces cell viability, induces CD1 lb and apoptosis in AML cells. (A) Indicated AML cell lines were exposed to increasing concentrations of Compound J to confirm their sensitivity to HDACl/2 inhibition. (B-D) MV4-11 cells were treated with indicated concentrations of compounds. (B) Surface levels of myeloid differentiation marker CD1 lb were determined by FACS at 72h post-treatment. Compound J showed the highest potency increasing percentage of CD1 lb positive cells. (C) Cell cycle was assessed by flow cytometry after incorporation of EdU and staining with Far Red at 72h post-treatment. The distribution of cells among G0/G1 phase, G2/M phase, S phase and subGl phase was determined. Compound J, Compound E and Compound A induced cell cycle arrest as well as apoptosis. (D) Apoptosis was assessed by flow cytometry via measuring Annexin V binding and cellular permeability to propidium iodide at 96h post-treatment. The relative fraction of cells that were live, in early apoptosis, in late apoptosis or dead was then determined.

Treatment with Compound J, Compound E, and Compound A resulted in increase in apoptosis relative to control cells.

Incorporation by Reference

The contents of all references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art.

Equivalents

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents of the specific embodiments provided herein described herein. Such equivalents are intended to be encompassed by the following claims.