Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMBINATION THERAPY
Document Type and Number:
WIPO Patent Application WO/2020/132563
Kind Code:
A1
Abstract:
Provided herein are methods of treating diseases, such as cancer, using a combination therapy. In certain embodiments, the methods comprise administering an effective amount of a phosphoinositide-3-kinase (PI3K) inhibitor and an effective amount of a bispecific antibody to a patient.

Inventors:
GOLD DANIEL P (US)
Application Number:
PCT/US2019/068050
Publication Date:
June 25, 2020
Filing Date:
December 20, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MEI PHARMA INC (US)
International Classes:
A61K31/4184; C07D235/04
Domestic Patent References:
WO2018053437A12018-03-22
WO2019036489A12019-02-21
Foreign References:
US20180250298A12018-09-06
US20120252802A12012-10-04
Attorney, Agent or Firm:
YVON, Brigitte (US)
Download PDF:
Claims:
CFAIMS

WHAT IS CLAIMED IS:

1. A method for treating or preventing cancer, comprising administering to a subject in need thereof an effective amount of:

a) a compound of Formula (I) :

Formula (I),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are

nitrogen atoms; where Rx is hydrogen or Ci-6 alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -

S(0)NRlbRlc, or -S(0)2NRlbRlc; wherein each Rla, Rlb, Rlc, and Rld is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rlb and Rlc together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or Ci-6 alkyl; or R3 and R4 are linked together to form a bond, Ci-6 alkylene, Ci-6 heteroalkylene, C2-6 alkenylene, or

C2-6 heteroalkenylene;

R5a is (a) hydrogen or halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -C(0)NRlbRlc, -C(NRla)NRlbRlc,

R5b is (a) halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl,

R5c is -(CR5fR5g)n-(C6 i4 aryl) or -(CR5fR5g)n-heteroaryl;

R5d and R5e are each independently (a) hydrogen or halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -

R5f and R5g are each independently (a) hydrogen or halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -

S(0)NRlbRlc; or -S(0)2NRlbRlc; or (d) when one occurrence of R5f and one occurrence of R5g are attached to the same carbon atom, the R5f and R5g together with the carbon atom to which they are attached form a C3-10 cycloalkyl or heterocyclyl;

R6 is hydrogen, Ci-6 alkyl, -S-Ci-6 alkyl, -S(0)-Ci-6 alkyl, or -SO2-C1-6 alkyl;

m is 0 or 1 ; and

n is 0, 1, 2, 3, or 4;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R1, R2, R3, R4, R6, Rx, Rla, Rlb, Rlc, Rld, R5a, R5b, R5c, R5d, R5e, R5f, and R5g is optionally substituted with one, two, three, four, or five substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Qa; and (c) -C(0)Ra, -C(0)0Ra, -C(0)NRbRc, -C(NRa)NRbRc, -ORa, -0C(0)Ra, -

S(0)NRbRc, and -S(0)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one, two, three, or four substituents Qa; wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)Re, -C(0)0Re, -C(0)NRfRg, -C(NRe)NRfRg, -ORe, -0C(0)Re, -

S(0)NRfRg, and -S(0)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl;

wherein two substituents Q that are adjacent to each other optionally form a C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one, two, three, or four substituents Qa; and

b) a T-cell activating bispecific antigen-binding molecule.

2. The method of claim 1, wherein R5b is (a) halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10

cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, or heteroaryl; or (c) -C(0)Rla, -C(0)0Rla, -C(0)NRlbRlc, -

3. The method of claim 1, wherein R5a and R5b are each independently (a) halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -

4. The method of claim 3, wherein R5a and R5b are each methyl, optionally substituted with one, two, or three halo(s).

5. The method of any one of claims 1-4, wherein n is 1.

6. The method of any one of claims 1-5, wherein R5f and R5g are each hydrogen.

7. The method of any one of claims 1-4, wherein n is 0.

8. The method of any one of claims 1-7, wherein m is 0.

9. The method of any one of claims 1-6, wherein the compound of Formula (I) is of Formula (XI):

Formula (XI),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

R7a, R7b. R7c, R7d, and R7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Qa; or (c) -C(0)Ra, -

o of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one, two, three, or four substituents Qa. 10. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A35:

Compound A35,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. 11. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A36:

Compound A36,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. 12. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A68:

Compound A68,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. 13. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A70:

Compound A70,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. 14. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A37:

Compound A37,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

15. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A38:

Compound A38,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

16. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A41:

Compound A41,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

17. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A42:

Compound A42, or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. 18. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A43:

Compound A43,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

19. The method of any one of claims 1-4, wherein the compound of Formula (I) is Compound A44:

Compound A44,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

20. The method of any one of claims 1-19, wherein the T-cell activating bispecific antigen-binding molecule comprises:

a first Fab molecule which specifically binds to a first antigen; and

a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an

activating T-cell antigen and the second antigen is a target cell antigen.

21. The method of claim 20, wherein the T-cell antigen is CD3.

22. The method of claim 20 or 21, wherein the target cell antigen is CD3, CD 19, CD20, CD 13, CD30,

CD33, CD123, HER1, HER2, CEA, disialoganglioside GD2, PSMA, gpA33, BCMA, ROR1, or CD40.

23. The method of any one of claims 1-22, wherein the T-cell activating bispecific antigen-binding molecule comprises an intact Fc region that can bind to cell surface receptors (Fc receptors).

24. The method of any one of claims 1-19, wherein the T-cell activating bispecific antigen-binding molecule is obinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, ertumaxomab, maxomab, AMV564, AFM13, REGN-1979, GEN-3013, or pasotuxizumab.

25. The method of any one of the preceding claims, wherein the cancer is a hematological malignancy.

26. The method of any one of the preceding claims, wherein the cancer is a B-cell malignancy.

27. The method of any one of the preceding claims, wherein the cancer is acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), chronic lymphocytic leukemia (CLL), high-risk chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high-risk small lymphocytic lymphoma (SLL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom’s macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt’s lymphoma, non-Burkitt’s high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B -lymphoblastic lymphoma, B cell prolymphocytic leukemia,

lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis.

28. The method of any one of the preceding claims, wherein the cancer is chronic lymphocytic

leukemia or non-Hodgkin’s lymphoma.

29. The method of any one of the preceding claims, wherein the cancer is non-Hodgkin’s lymphoma diffuse large B-cell lymphoma (DLBCL).

30. The method of any one of the preceding claims, wherein the cancer is relapsed-refractory diffuse large B-cell lymphoma (r/r DLBCL).

31. The method of claim 29 or 30, wherein the diffuse large B-cell lymphoma is of the activated B-cell (ABC DLBCL) or Germinal center B-cell (GCB DLBCL).

32. The method of any one of claims 1-27, wherein the cancer is follicular lymphoma (FL).

33. The method of any one of the preceding claims, wherein the compound of Formula (I), or an

enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered simultaneously, approximately simultaneously, or sequentially in any order.

34. The method of any one of claims 1-33, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered simultaneously or approximately simultaneously.

35. The method of any one of claims 1-33, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered sequentially.

36. The method of claim 35, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof, is administered before the T-cell activating bispecific antigen -binding molecule.

37. The method of claim 35, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof, is administered after the T-cell activating bispecific antigen-binding molecule.

38. The method of any one of claims 1-37, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is formulated for oral administration.

39. The method of claim 38, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is formulated as a tablet or capsule.

40. The method of any one of claims 1-39, wherein about 30 mg, about 60 mg, about 120 mg, or about 180 mg of a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject.

41. The method of any one claims 1 -40, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least one 28 -day cycle.

42. The method of any one claims 1-40, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for two 28-day cycles.

43. The method of any one of claims 1-40, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered daily to the subject on a continuous schedule until disease progression or intolerable toxicity occurs.

44. The method of any one of claims 1-43, wherein the method comprises at least three cycles, wherein:

(i) the first two cycles comprise a continuous daily dosing schedule (CS), comprising

administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for two cycles; and

(ii) the subsequent cycle(s) comprises an intermittent dosing schedule (IS), comprising

administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for only the first 7 consecutive days in each subsequent cycle.

45. The method of claim 44, wherein the CS and IS cycles are each 28-day cycles.

46. The method of claim 44 or 45, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) until disease progression occurs or the incidence of at least one toxicity is reduced.

47. The method of claim 46, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered daily to the subject on a continuous dosing schedule (CS) after disease progression occurs on an intermittent dosing schedule (IS).

48. The method of claim 1 or 2, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject daily.

49. The method of any one of the preceding claims, wherein the compound of Formula (I), or an

enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once per day, twice per day, or three times per day.

50. The method of any one of the preceding claims, wherein the compound of Formula (I), or an

enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once per day.

51. The method of any one of the preceding claims, wherein about 60 mg/day of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject.

52. The method of any one claims 1-37, wherein the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is co-formulated with the T- cell activating bispecific antigen-binding molecule.

53. The method of any one of claims 1-52, wherein the T-cell activating bispecific antigen-binding molecule is obinutuzumab.

54. The method of claim 53, wherein obinutuzumab is administered by intravenous infusion.

55. The method of claim 53 or 54, wherein from about 100 mg to about 1,000 mg of obinutuzumab is administered.

56. The method of any one of claims 53-55, wherein a loading dose of obinutuzumab is administered.

57. The method of any one of claims 53-56, wherein obinutuzumab is administered for at least six 28- day cycles.

58. The method of any one of claims 53-57, wherein 100 mg on day 1 of Cycle 1 and 900 mg on day 2 of obinutuzumab is administered.

59. The method of claim 58, wherein 1,000 mg of obinutuzumab is administered on day 8 and on day 15 of Cycle 1.

60. The method of claim 59, wherein 1,000 mg of obinutuzumab is administered on day 1 of Cycles 2- 6

61. The method of any one of claims 53-57, wherein 1,000 mg of obinutuzumab is administered on day 1, 8, and 15 of Cycle 1.

62. The method of claim 61, wherein 1,000 mg of obinutuzumab is administered on day 1 of Cycles 2- 6

63. The method of claim 62, wherein obinutuzumab is administered every 2 months on Cycle(s) >7.

64. A pharmaceutical composition, comprising:

a) a compound of Formula (I) :

Formula (I),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

X, Y, and Z are each independently N or CRX, with the proviso that at least two of X, Y, and Z are

nitrogen atoms; where Rx is hydrogen or Ci-e alkyl;

R1 and R2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci-e alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -

S(0)NRlbRlc, or -S(0)2NRlbRlc; wherein each Rla, Rlb, Rlc, and Rld is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rlb and Rlc together with the N atom to which they are attached form heterocyclyl;

R3 and R4 are each independently hydrogen or Ci-6 alkyl; or R3 and R4 are linked together to form a bond, Ci-6 alkylene, Ci-6 heteroalkylene, C2-6 alkenylene, or

C2-6 heteroalkenylene;

R5a is (a) hydrogen or halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CYu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)ORla, -C(0)NRlbRlc, -C(NRla)NRlbRlc,

R5b is (a) halo; (b) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl,

R5c is -(CR5fR5g)n-(C6-i4 aryl) or -(CR5fR5g)n-heteroaryl;

R5d and R5e are each independently (a) hydrogen or halo; (b) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -

R5f and R5g are each independently (a) hydrogen or halo; (b) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -

S(0)NRlbRlc; or -S(0)2NRlbRlc; or (d) when one occurrence of R5f and one occurrence of R5g are attached to the same carbon atom, the R5f and R5g together with the carbon atom to which they are attached form a C3-10 cycloalkyl or heterocyclyl;

R6 is hydrogen, Ci-6 alkyl, -S-Ci-6 alkyl, -S(0)-Ci-6 alkyl, or -S02-Ci-6 alkyl;

m is 0 or 1 ; and

n is 0, 1, 2, 3, or 4;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R1, R2, R3, R4, R6, Rx, Rla, Rlb, Rlc, Rld, R5a, R5b, R5c, R5d, R5e, R5f, and R5g is optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) Ci-6 alkyl, C2-e alkenyl, C2-e alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Qa; and (c) -C(0)Ra, -C(0)0Ra, -C(0)NRbRc, -C(NRa)NRbRc, -ORa, - 0C(0)Ra, -0C(0)0Ra, -0C(0)NRbRc, -OC(=NRa)NRbRc, -0S(0)Ra, -0S(0)2Ra, -0S(0)NRbRc, -0S(0)2NRbRc, -NRbRc, -NRaC(0)Rd, -NRaC(0)0Rd, -NRaC(0)NRbRc, -NRaC(=NRd)NRbRc, -

NRaS(0)Rd, -NRaS(0)2Rd, -NRaS(0)NRbRc, -NRaS(0)2NRbRc, -SRa, -S(0)Ra, -S(0)2Ra, - S(0)NRbRc, and -S(0)2NRbRc, wherein each Ra, Rb, Rc, and Rd is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Qa; or (iii) Rb and Rc together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one, two, three, or four substituents Qa; wherein each Qa is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)Re, -C(0)0Re, -C(0)NRfRg, -C(NRe)NRfRg, -ORe, -0C(0)Re, -

S(0)NRfRg, and -S(0)2NRfRg; wherein each Re, Rf, Rg, and Rh is independently (i) hydrogen; (ii) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) Rf and Rg together with the N atom to which they are attached form heterocyclyl;

wherein two substituents Q that are adjacent to each other optionally form a C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one, two, three, or four substituents Qa; and

b) a T-cell activating bispecific antigen-binding molecule; and

c) at least one pharmaceutically acceptable excipient.

65. The pharmaceutical composition of claim 64, wherein R5b is (a) halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C6-14 aryl, C7-15 aralkyl, or heteroaryl; or (c) -

66. The pharmaceutical composition of claim 64, wherein R5a and R5b are each independently (a) halo;

(b) Ci-6 alkyl, C2-r, alkenyl, C2-r, alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)Rla, -C(0)0Rla, -C(0)NRlbRlc, -C(NRla)NRlbRlc, -ORla, -0C(0)Rla, -

67. The pharmaceutical composition of claim 66, wherein R5a and R5b are each methyl, optionally

substituted with one, two, or three halo(s).

68. The pharmaceutical composition of any one of claims 64-67, wherein n is 1.

69. The pharmaceutical composition of any one of claims 64-68, wherein R5f and R5g are each

hydrogen.

70. The pharmaceutical composition of any one of claims 64-67, wherein n is 0.

71. The pharmaceutical composition of any one of claims 64-70, wherein m is 0.

72. The pharmaceutical composition of any one of claims 64-69, wherein the compound of Formula (I) is of Formula (XI):

Formula (XI),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; wherein:

R7a, R7b. R7c, R7d, and R7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Qa; or (c) -C(0)Ra, -

o of R7a, R7b, R7c, R7d, and R7e that are adjacent to each other form C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one, two, three, or four substituents Qa. 73. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A35:

Compound A35,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

74. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A36:

Compound A36,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

75. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A68:

Compound A68,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

76. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A70:

Compound A70,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

77. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A37:

Compound A37,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

78. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A38:

Compound A38,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

79. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A41 :

Compound A41,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

80. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A42:

Compound A42,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

81. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A43 :

Compound A43,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

82. The pharmaceutical composition of any one of claims 64-67, wherein the compound of Formula (I) is Compound A44:

Compound A44,

or an isotopic variant thereof, a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

83. The pharmaceutical composition of any one of claims 64-82, wherein the T-cell activating

bispecific antigen-binding molecule comprises:

a first Fab molecule which specifically binds to a first antigen; and

a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an

activating T-cell antigen and the second antigen is a target cell antigen.

84. The pharmaceutical composition of claim 83, wherein the T-cell antigen is CD3.

85. The pharmaceutical composition of claim 83 or 84, wherein the target cell antigen is CD 19, CD20, CD13, CD30, CD33, BCMA, ROR1, or CD40.

86. The pharmaceutical composition of any one of claims 64-85, wherein the T-cell activating

bispecific antigen-binding molecule comprises an intact Fc region that can bind to cell surface receptors (Fc receptors).

87. The pharmaceutical composition of any one of claims 64-82, wherein the T-cell activating

bispecific antigen-binding molecule is obinutuzumab, mosunetuzumab, selicrelumab,

blinatumomab, AMV564, AFM13, REGN-1979, GEN-3013, or pasotuxizumab.

88. A method for treating or preventing cancer, comprising administering to a subject in need thereof an effective amount of the pharmaceutical composition of any one of claims 64-87.

89. The method of claim 88, wherein the T-cell activating bispecific antigen-binding molecule

comprises:

a first Fab molecule which specifically binds to a first antigen; and

a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an

activating T-cell antigen and the second antigen is a target cell antigen.

90. The method of claim 89 wherein the T-cell antigen is CD3.

91. The method of claim 88 or 89, wherein the target cell antigen is CD19, CD20, CD13, CD30, CD33, BCMA, RORl, or CD40.

92. The method of any one of claims 88-91, wherein the T-cell activating bispecific antigen-binding molecule comprises an intact Fc region that can bind to cell surface receptors (Fc receptors).

93. The method of any one of claims 88-92, wherein the T-cell activating bispecific antigen-binding molecule is obinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, AMV564, AFM13, REGN-1979, GEN-3013, or pasotuxizumab.

94. The method of any one of the preceding claims, wherein the cancer is a hematological malignancy.

95. The method of any one of the preceding claims, wherein the cancer is a B-cell malignancy.

96. The method of any one of the preceding claims, wherein the cancer is acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), chronic lymphocytic leukemia (CLL), high-risk chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high-risk small lymphocytic lymphoma (SLL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom’s macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt’s lymphoma, non-Burkitt’s high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B -lymphoblastic lymphoma, B cell prolymphocytic leukemia,

lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis.

- I l l -

97. The method of any one of the preceding claims, wherein the cancer is chronic lymphocytic leukemia or non-Hodgkin’s lymphoma.

98. The method of any one of the preceding claims, wherein the cancer is non -Hodgkin’s lymphoma diffuse large B-cell lymphoma (DLBCL).

99. The method of any one of the preceding claims, wherein the cancer is relapsed-refractory diffuse large B-cell lymphoma (r/r DLBCL).

100. The method of claim 98 or 99, wherein the diffuse large B-cell lymphoma is of the activated B-cell (ABC DLBCL) or Germinal center B-cell (GCB DLBCL).

101. The method of any one of claims 88-96, wherein the cancer is follicular lymphoma (FL).

Description:
COMBINATION THERAPY

CROSS REFERENCE TO RELATED APPLICATION

[0001] This application claims the benefit of U.S. Provisional Application No. 62/783,910, filed December 21, 2018, which is incorporated by reference in the disclosure of this application.

FIELD

[0002] Provided herein are methods of treating diseases using a combination therapy for treatment of a proliferative disease, including a cancer, an autoimmune disease and an inflammatory disease. In certain embodiments, the methods comprise administering an effective amount of a phosphoinositide -3 -kinase (PI3K) inhibitor and an effective amount of a T cell activating bispecific antigen-binding molecule to a patient.

BACKGROUND OF THE DISCLOSURE

[0003] Phosphoinositide-3-kinases (PI3Ks) play a variety of roles in normal tissue physiology, with pi 10a having a specific role in cancer growth, pi 10b in thrombus formation mediated by integrin apb3 and pi 10g in inflammation, rheumatoid arthritis, and other chronic inflammation states. Inhibitors of PI3K have therapeutic potential in the treatment of various proliferative diseases, including cancer.

[0004] Bi-specific T-cell engagers are a class of artificial bispecific monoclonal antibodies that cause T cells to exert cytotoxic activity on tumor cells. These bispecific antibodies have therapeutic potential for use as anti -cancer drugs.

SUMMARY OF THE DISCLOSURE DISCLOSURE

[0005] Disclosed herein is a method for treating or preventing cancer, comprising administering to a subject in need thereof an effective amount of:

a) a compound of F ormula (I) :

Formula (I),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;

wherein:

X, Y, and Z are each independently N or CR X , with the proviso that at least two of X, Y, and Z are

nitrogen atoms; where R x is hydrogen or Ci- 6 alkyl; R 1 and R 2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -

S(0)NR lb R lc , or -S(0) 2 NR lb R lc ; wherein each R la , R lb , R lc , and R ld is independently (i) hydrogen; (ii) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R lb and R lc together with the N atom to which they are attached form heterocyclyl;

R 3 and R 4 are each independently hydrogen or Ci- 6 alkyl; or R 3 and R 4 are linked together to form a bond, Ci-6 alkylene, Ci-6 heteroalkylene, C2-6 alkenylene, or

C2-6 heteroalkenylene;

R 5a is (a) hydrogen or halo; (b) Ci- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, Ce-u aryl, C 7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , -C(0)NR lb R lc , -C(NR la )NR lb R lc ,

R 5b is (a) halo; (b) Ci-6 alkyl, C 2 -6 alkenyl, C 2 -6 alkynyl, C 3-10 cycloalkyl, Ce-u aryl, C7- 15 aralkyl,

R 5c is -(CR 5f R 5g ) n -(C 6 -i 4 aryl) or -(CR 5f R 5g ) n -heteroaryl;

R 5d and R 5e are each independently (a) hydrogen or halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , -

R 5f and R 5g are each independently (a) hydrogen or halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , - NR la S(0) 2 R ld , -NR la S (0)NR lb R lc , -NR la S(0) 2 NR lb R lc , -SR la , -S(0)R la , -S(0) 2 R la , -

S(0)NR lb R lc ; or -S(0) 2 NR lb R lc ; or (d) when one occurrence of R 5f and one occurrence of R 5g are atached to the same carbon atom, the R 5f and R 5g together with the carbon atom to which they are atached form a C3-10 cycloalkyl or heterocyclyl;

R 6 is hydrogen, Ci- 6 alkyl, -S-Ci- 6 alkyl, -S(0)-Ci-r, alkyl, or -S0 2- Ci-r, alkyl;

m is 0 or 1 ; and

n is 0, 1, 2, 3, or 4;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R 1 , R 2 , R 3 , R 4 , R 6 , R x , R la , R lb , R lc , R ld , R 5a , R 5b , R 5c , R 5d , R 5e , R 5f , and R 5g is optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two,

S(0)NR b R c , and -S(0) 2 NR b R c , wherein each R a , R b , R c , and R d is independently (i) hydrogen; (ii) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; or (iii) R b and R c together with the N atom to which they are atached form heterocyclyl, which is further optionally substituted with one, two, three, or four substituents Q a ;

wherein each Q a is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)R e , -C(0)0R e , -C(0)NR f R g , -C(NR e )NR f R g , -OR e , -0C(0)R e , -

S(0)NR f R g , and -S(0) 2 NR f R g ; wherein each R e , R f , R g , and R h is independently (i) hydrogen; (ii) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R f and R g together with the N atom to which they are atached form

heterocyclyl;

wherein two substituents Q that are adjacent to each other optionally form a C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; and

b) a T-cell activating bispecific antigen-binding molecule. [0006] Disclosed herein is a method for treating or preventing cancer, comprising administering to a subject in need thereof an effective amount of a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;and bispecific antibody or a bispecific antigen -binding molecule.

[0007] In some embodiments, R 5b is (a) halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl,

[0008] In some embodiments, R 5a and R 5b are each independently (a) halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2- 6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -

[0009] In some embodiments, R 5a and R 5b are each methyl, optionally substituted with one, two, or three halo(s). In some embodiments, n is 1. In some embodiments, R 5f and R 5g are each hydrogen. In some embodiments, n is 0. In some embodiments, m is 0.

[0010] In some embodiments, the compound of Formula (I) is of Formula (XI):

Formula (XI)

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;

wherein:

R 7a , R 7b , R 7c , R 7d , and R 7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents

OC(0)OR a , OS(0) 2 NR b R c , -NR b R c , -NR a C(0)R d , -NR a C(0)OR d , -NR a C(0)NR b R c , -NR a C(=NR d )NR b R c , -

NR a S(0)R d , -NR a S(0) 2 R d , -NR a S(0)NR b R c , -NR a S(0) 2 NR b R c , -SR a , -S(0)R a , -S(0) 2 R a , - S(0)NR b R c , or -S(0) 2 NR b R c ; or two of R 7a , R 7b , R 7c , R 7d , and R 7e that are adjacent to each other form C3-10 cycloalkenyl, Ce-w aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a .

[0011] In some embodiments, the compound of Formula (I) is Compound A35:

Compound A35,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0012] In some embodiments, the compound of Formula (I) is Compound A36:

Compound A36,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0013] In some embodiments, the compound of Formula (I) is Compound A68:

Compound A68,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0014] In some embodiments, the compound of Formula (I) is Compound A70:

Compound A70,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0015] In some embodiments, the compound of Formula (I) is Compound A37:

Compound A37,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0016] In some embodiments, the compound of Formula (I) is Compound A38:

Compound A38,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0017] In some embodiments, the compound of Formula (I) is Compound A41 :

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0018] In some embodiments, the compound of Formula (I) is Compound A42:

Compound A42,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0019] In some embodiments, the compound of Formula (I) is Compound A43:

Compound A43,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0020] In some embodiments, the compound of Formula (I) is Compound A44:

Compound A44,

or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0021] In some embodiments, the T-cell activating bispecific antigen-binding molecule comprises: a first Fab molecule which specifically binds to a first antigen; and

a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen.

[0022] In some embodiments, the T-cell antigen is CD3. In some embodiments, the T-cell activating bispecific antigen-binding molecule comprises an intact Fc region that can bind to cell surface receptors (Fc receptors).

[0023] In some embodiments, the target cell antigen is CD 19, CD20, CD 13, CD30, CD33, or CD40. [0024] In some embodiments, the the T-cell activating bispecific antigen-binding molecule is obinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, AMV564, AFM13, REGN-1979, GEN- 3013, or pasotuxizumab.

[0025] In some embodiments, the cancer being treated is a hematological malignancy. In some embodiments, the cancer being treated is a B-cell malignancy. In some embodiments, the cancer being treated is acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), chronic lymphocytic leukemia (CLL), high-risk chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high-risk small lymphocytic lymphoma (SLL), follicular lymphoma (FL), including relapsed/refractory FL, diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom’s macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt’s lymphoma, non-Burkitt high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B -lymphoblastic lymphoma, B cell

prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis. In some embodiments, the cancer being treated is chronic lymphocytic leukemia or non-Hodgkin’s lymphoma. In some

embodiments, the cancer being treated is non-Hodgkin’s lymphoma and the non-Hodgkin’s lymphoma is diffuse large B-cell lymphoma (DLBCL). In some embodiments, the cancer being treated is relapsed- refractory diffuse large B-cell lymphoma (r/r DLBCL). In some embodiments, the diffuse large B-cell lymphoma is of the activated B-cell (ABC-DLBCL) or Germinal center B-cell (GCB-DLBCL). In some embodiments, the cancer is follicular lymphoma (FL).

[0026] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered simultaneously, approximately simultaneously, or sequentially in any order.

[0027] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered simultaneously or approximately simultaneously.

[0028] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof and the T-cell activating bispecific antigen-binding molecule are administered sequentially.

[0029] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof, is administered before the T-cell activating bispecific antigen-binding molecule.

[0030] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof, is administered after the T-cell activating bispecific antigen-binding molecule. [0031] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is formulated for oral administration.

[0032] In some embodiments, about 30 mg, about 60 mg, about 120 mg, or about 180 mg of a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject.

[0033] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is formulated as a tablet or capsule.

[0034] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is co-formulated with the T-cell activating bispecific antigen binding molecule.

[0035] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least one 28-day cycle.

[0036] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for two 28-day cycles.

[0037] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered daily to the subject on a continuous schedule until disease progression or intolerable toxicity occurs.

[0038] In some embodiments, the cycle is a 28-day cycle.

[0039] In some embodiments, the method comprises at least three cycles,

wherein:

(i) the first two cycles comprise a continuous daily dosing schedule (CS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for two cycles; and

(ii) the subsequent cycle(s) comprises an intermittent dosing schedule (IS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for only the first 7 consecutive days in each subsequent cycle.

[0040] In some embodiments, the CS and IS cycles are each 28-day cycles. [0041] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) until disease progression occurs or the incidence of at least one toxicity is reduced.

[0042] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered daily to the subject on a continuous dosing schedule (CS) after disease progression occurs on an intermittent dosing schedule (IS)..

[0043] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject daily.

[0044] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once per day, twice per day, or three times per day.

[0045] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once per day.

[0046] In some embodiments, about 60 mg/day of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject.

[0047] In some embodiments, the T-cell activating bispecific antigen -binding molecule is obinutuzumab. In some embodiments, obinutuzumab is administered by intravenous infusion. In some embodiments, from about 100 mg to about 1,000 mg of obinutuzumab is administered. In some embodiments, a loading dose of obinutuzumab is administered. In some embodiments, obinutuzumab is administered for at least six 28-day cycles. In some embodiments, 100 mg on day 1 of Cycle 1 and 900 mg on day 2 of obinutuzumab is administered. In some embodiments, 1,000 mg of obinutuzumab is administered on day 8 and on day 15 of Cycle 1. In some embodiments, 1,000 mg of obinutuzumab is administered on day 1 of Cycles 2-6. In some embodiments, 1,000 mg of obinutuzumab is administered on day 1, 8, and 15 of Cycle 1. In some embodiments, 1,000 mg of obinutuzumab is administered on day 1 of Cycles 2-6.

[0048] In some embodiments, obinutuzumab is administered every 2 months on Cycle(s) >7.

[0049] In an aspect, provided herein is a pharmaceutical composition, comprising:

(i) a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;

(ii) a T-cell activating bispecific antigen-binding molecule; and

(iii) at least one pharmaceutically acceptable excipient. [0050] In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is of Formula (XI), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A35. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A36. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A68. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A70. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A37. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A38. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A41. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A42. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A43. In some embodiments of the pharmaceutical compositions provided herein, the compound of Formula (I) is Compound A44.

[0051] In some embodiments of the pharmaceutical compositions provided herein, the T-cell activating bispecific antigen-binding molecule comprises:

a first Fab molecule which specifically binds to a first antigen; and

a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen.

[0052] In some embodiments of the pharmaceutical compositions provided herein, the T-cell antigen is CD3. In some embodiments of the pharmaceutical compositions provided herein, the target cell antigen is CD19, CD20, CD13, CD30, CD33, BCMA, ROR1, or CD40. In some embodiments of the pharmaceutical compositions provided herein, the T-cell activating bispecific antigen-binding molecule comprises an intact Fc region that can bind to cell surface receptors (Fc receptors). In some embodiments of the pharmaceutical compositions provided herein, the T-cell activating bispecific antigen-binding molecule is obinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, AMV564, AFM13, REGN-1979, GEN- 3013, or pasotuxizumab.

[0053] In an aspect, provided herein is a method for treating or preventing cancer, comprising administering to a subject in need thereof an effective amount of:

(i) a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;

(ii) a T-cell activating bispecific antigen-binding molecule; and

(iii) at least one pharmaceutically acceptable excipient. INCORPORATION BY REFERENCE

[0054] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

DETAILED DESCRIPTION OF THE DISCLOSURE

[0055] Some embodiments provided herein describe pharmaceutical compositions comprising a PI3K delta inhibitor and methods for treating patients with B cell malignancies with a PI3K inhbitor. In some embodiments, the dosing regimens and schedules described herein reduce toxicities associate with PI3K delta inhibitors.

[0056] Class I phosphatidylinositol 3-kinases (PI3Ks) regulate numerous cellular functions. PI3Ks are composed of regulatory (p85) and catalytic (pi 10) subunits, with the catalytic unit consisting of 4 distinct isoforms designated a, b, g, and d. PI3K5 is primarily expressed in lymphocytes where it plays a key role in normal lymphocyte biology including proliferation, homing and survival. RI3Kd is frequently active in B-cell malignancies and is central to multiple B-cell receptor (BCR) signaling pathways that drive proliferation, survival, homing and retention of malignant B-cells in lymphoid tissue and bone marrow.

[0057] Small molecule PI3K delta (or RI3Kd) inhibitors are effective for the treatment of B-cell malignancies, including chronic lymphocytic leukemia (CLL) , follicular lymphoma and other B- cell lymphomas. However, in some instances, toxicities associated with the RI3Kd are severe and have been fatal in some patients. Toxicities reported with RI3Kd inhibitors (e.g., idelalisib, parsaclisib

(INCB050465), copanlisib, duvelisib, umbralisib, etc.) include but are not limited to enterocolitis (manifested as diarrhea/colitis), cutaneous toxicities (e.g., rashes), liver toxicity (manifested as elevation of transaminases), pulmonary toxicity (manifested as non-infectious pneumonitis), and infections. These toxicities may be severe and have been fatal in some patients. The frequency, severity and time to onset of these adverse events (AEs) vary among RI3Kd inhibitors. Enterocolitis, rash, and transaminitis have been reported in certain clinical studies of RI3Kd in patients with B-cell malignancies. In certain instances, lymphocytic infdtrates have reported in biopsies obtained from subjects with colitis and/or severe skin rash with corticosteroid therapy being an effective treatment approach in patients who developed diarrhea and rash.

[0058] A better understanding of the pathogenesis of these toxicities may help in developing approaches to mitigate their risk. Several lines of investigations have suggested that some of these toxicities are related to dysfunction in immune homeostasis. An immune mechanism for RBKd-associated enterocolitis has been hypothesized based on observations that include mice with genetic inactivation of pi 10d develop an autoimmune -like colitis; histopathologic data from patients with diarrhea/colitis associated with RI3Kd inhibitors show an intraepithelial lymphocytosis, indicative of an immune reaction; and some patients with late onset RI3Kd inhibitors-associated diarrhea/colitis do not respond to antidiarrheal or empiric antimicrobial therapy but may respond to treatment with corticosteroids, supporting an immune mechanism for the diarrhea. [0059] There is also evidence pointing to a role for the PI3K pathway in T lymphocytes, which may explain these immune dysregulations. For example, in mice, genetic inactivation of pi 105 results in a decrease of the function of regulatory T-cells (TREGs), a subset population of T-cells. TREGs have been shown to have an important role in controlling auto-immunity. In mice, pi 105 was shown to be required for mounting and effective T-cell responses to viral and bacterial infections. In some instances, PI3K5 inhibition results in various immune-mediated toxicities such as enterocolitis and skin toxicity due to TREG suppression, as well as infections due to suppression of B-cells and effector T-cells. In some instances, treatment regimens with small molecule PI3K5 delta (or PI3K5) inhibitors on an intermittent dosing schedule (IS) are used. However, in certain instances, progression of disease is observed in subjects who are treated for B-cell malignancies, including chronic lymphocytic leukemia (CLL) and lymphomas with small molecule PI3K5 inhibitors (e.g., parsaclisib (INCB050465)) on an IS dosing regimen. It has been demonstrated that parsaclisib once weekly dosing resulted in plasma levels > IC90 for 1.5/7 days (i.e., 32%). For parsaclisib, plasma approximates tissue levels and off target ~5 days of 7 was not sufficient to hold response to treatment in most patients. In some embodiments, the methods of treatment and dosing regimens and schedules described herein provide an efficacious and tolerable treatment of cancer. In some embodiments, the methods of treatment and dosing regimens and schedules described herein improve the frequency, severity and time to onset of the adverse events (AEs) associated with PI3K delta inhibitors. In some embodiments, the methods of treatment and dosing regimens and schedules described herein, including IS dosing regimens, result in partial or complete remission. In some embodiments, the methods of treatment and dosing regimens and schedules described herein, including IS dosing regimens (e.g., one week on / three week off dosing), result in plasma levels of > IC90 for 9/28 days (i.e., 32%) for the compounds described herein. For the compounds described herein (e.g.,

Compound A35), plasma levels underestimate tissue levels, and higher levels of the compounds described herein are predicted in tumor versus plasma.

[0060] Described herein are pharmaceutical compositions comprising i) a PI3K inhibitor; and ii) a T-cell activating bispecific antigen-binding molecule. In some instances, the pharmaceutical compositions described herein may be used for treating diseases or disorders such as cancer. Also described herein are methods of treating the diseases and disorders such as cancer with a combination of i) a PI3K inhibitor, and; ii) a T-cell activating bispecific antigen -binding molecule.

[0061] To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.

[0062] Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well-known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.

DEFINITIONS

[0063] The term“subject” refers to an animal, including, but not limited to, a primate (e.g., human), cow, pig, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms“subject” and“patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject, in one embodiment, a human.

[0064] The terms“treat,”“treating,” and“treatment” are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself.

[0065] The terms“prevent,”“preventing,” and“prevention” are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject’s risk of acquiring a disorder, disease, or condition.

[0066] The terms“therapeutically effective amount” and“effective amount” are meant to include the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder, disease, or condition being treated. The terms “therapeutically effective amount” or“effective amount” also refer to the amount of a compound that is sufficient to elicit the biological or medical response of a biological molecule (e.g., a protein, enzyme, RNA, or DNA), cell, tissue, system, animal, or human, which is being sought by a researcher, veterinarian, medical doctor, or clinician.

[0067] The terms“pharmaceutically acceptable carrier,”“pharmaceutically acceptable excipient,” “physiologically acceptable carrier,” and“physiologically acceptable excipient” refer to a

pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid fdler, diluent, solvent, or encapsulating material. In one embodiment, each component is“pharmaceutically acceptable” in the sense of being compatible with other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition, Lippincott Williams & Wilkins: Philadelphia, PA, 2005; Handbook of Pharmaceutical Excipients, 5th Edition, Rowe et al., Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and

Handbook of Pharmaceutical Additives, 3rd Edition, Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd Edition, Gibson Ed., CRC Press LLC: Boca Raton, FL, 2009.

[0068] The terms“about” and“approximately” mean an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the terms“about” and“approximately” mean within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term“about” or“approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.

[0069] The terms“active ingredient” and“active substance” refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition. As used herein, “active ingredient” and“active substance” may be an optically active isomer of a compound described herein.

[0070] The terms“drug,”“therapeutic agent,” and“chemotherapeutic agent” refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder, disease, or condition.

[0071] The terms“naturally occurring” and“native” when used in connection with biological materials such as nucleic acid molecules, polypeptides, host cells, and the like, refer to materials which are found in nature and are not manipulated by man. Similarly,“non-naturally occurring” or“non-native” refers to a material that is not found in nature or that has been structurally modified or synthesized by man.

[0072] The term“PI3K” refers to a phosphoinositide 3-kinase or variant thereof, which is capable of phosphorylating the inositol ring of PI in the D-3 position. The term“PI3K variant” is intended to include proteins substantially homologous to a native PI3K, i.e., proteins having one or more naturally or non- naturally occurring amino acid deletions, insertions, or substitutions (e.g., PI3K derivatives, homologs, and fragments), as compared to the amino acid sequence of a native PI3K. The amino acid sequence of a PI3K variant is at least about 80% identical, at least about 90% identical, or at least about 95% identical to a native PI3K. Examples of PI3K include, but are not limited to, pi 10a, pi 10b, pi 105, pi IOg, PI3K-C2a, PI3K-C2P, PI3K-C2y, Vps34, mTOR, ATM, ATR, and DNA-PK. See, Fry, Biochem. Biophys. Acta 1994, 1226 , 237-268; Vanhaesebroeck and Waterfield, Exp. Cell. Res. 1999, 253, 239-254; and Fry, Breast Cancer Res. 2001, 3, 304-312. PI3Ks are classified into at least four classes. Class I includes pi 10a, pi 10p, pi 105, and pi IOg. Class II includes PI3K-C2a, PI3K-C2P, and PI3K-C2y. Class III includes Vps34. Class IV includes mTOR, ATM, ATR, and DNA-PK. In certain embodiments, the PI3K is a Class I kinase. In certain embodiments, the PI3K is pi 10a, pi 10p, pi 105, or pi 10y. In certain embodiments, the PI3K is a variant of a Class I kinase. In certain embodiments, the PI3K is a pi 10a mutant. Examples of pi 10a mutants include, but are not limited to, R38H, G106V, K11 IN, K227E, N345K, C420R, P539R, E542K, E545A, E545G, E545K, Q546K, Q546P, E453Q, H710P, I800F, T1025S, M10431, M1043V, H1047F, H1047R, and El 1047Y (Ikenoue et al, Cancer Res. 2005, 65, 4562-4567; Gymnopoulos et al, Proc. Natl. Acad Sci., 2007, 104, 5569-5574). In certain embodiments, the PI3K is a Class II kinase. In certain embodiments, the PI3K is PI3K-C2a, PI3K- C2p, or PI3K-C2y. In certain embodiments, the PI3K is a Class III kinase. In certain embodiments, the PI3K is Vps34. In certain embodiments, the PI3K is a Class IV kinase. In certain embodiments, the PI3K is mTOR, ATM, ATR, or DNA-PK.

[0073] As used herein, the terms“bispecific antibody (BsAb),”“bispecific monoclonal antibody

(BsMAb),”“T-cell activating bispecific antigen-binding molecule,” and“bispecific T-cell engager (BiTE)” are used interchangeably and refer to an antibody or monoclonal antibody with binding sites for two different antigens (e.g., a first Fab molecule which specifically binds to a first antigen and a second Fab molecule which specifically binds to a second antigen). In some embodiments, the T-cell activating bispecific antigen-binding molecules described herein comprise a first antigen and a second antigen, where the first antigen the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen. It is to be understood that the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen or the second antigen is an activating T-cell antigen and the first antigen is a target cell antigen. In some embodiments, the terms, for example, anti-CD20 x anti-CD3 antibody or CD20 x CD3 antibody and the like, are synonymous and refer to a bispecific antibody that binds to antigens CD3 and CD20.

[0074] The terms“synergy,”“synergism,” and“synergistic” as used herein refer to a combination of therapies (e.g., use of a PI3K inhibitor of Formula (I) and a T-cell activating bispecific antigen-binding molecule) that is more effective than the expected additive effects of any two or more single therapies. For example, a synergistic effect of a combination of therapies permits the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies to a subject. The ability to utilize lower dosages of therapies and/or to administer the therapies less frequently reduces the toxicity associated with the administration of the therapies to a subject without reducing the efficacy of said therapies in the prevention, management, treatment, or amelioration of a given disease, such as an autoimmune disease, inflammatory disease, or cancer including, but not limited to, chronic lymphocytic leukemia or non-Hodgkin’s lymphoma. In addition, a synergistic effect can result in improved efficacy of therapies in the prevention, management, treatment, or amelioration of a given disease, such an autoimmune disease, inflammatory disease, or cancer including, but not limited to, chronic lymphocytic leukemia or non-Hodgkin’s lymphoma. Finally, synergistic effects of a combination of therapies may avoid or reduce adverse or unwanted side effects associated with the use of any single therapy. The “synergy,”“synergism,” or“synergistic” effect of a combination may be determined herein by the methods of Chou et ah, and/or Clarke et al. See Ting-Chao Chou, Theoretical Basis, Experimental Design, and Computerized Simulation of Synergism and Antagonism in Drug Combination Studies, Pharmacol Rev 58:621-681 (2006), and Clarke et al., Issues in experimental design and endpoint analysis in the study of experimental cytotoxic agents in vivo in breast cancer and other models, Breast Cancer Research and Treatment 46:255-278 (1997), which are both incorporated by reference for the methods of determining the“synergy,” synergism,” or“synergistic” effect of a combination.

[0075] The term“isotopic variant” refers to a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such a compound. In certain embodiments, an“isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen ( Ή). deuterium ( 2 H), tritium ( 3 H), carbon-11 ( n C), carbon-12 ( 12 C), carbon-13 ( 13 C), carbon- 14 ( 14 C), nitrogen-13 ( 13 N), nitrogen-14 ( 14 N), nitrogen-15 ( 15 N), oxygen-14 ( 14 0), oxygen-15 ( 15 0), oxygen-16 ( 16 0), oxygen-17 ( 17 0), oxygen-18 ( 18 0), fluorine-17 ( 17 F), fluorine-18 ( 18 F), phosphorus-31 ( 31 P), phosphorus-32 ( 32 P), phosphorus-33 ( 33 P), sulfur-32 ( 32 S), sulfur-33 ( 33 S), sulfur-34 ( 34 S), sulfur-35 ( 35 S), sulfur-36 ( 36 S), chlorine-35 ( 35 C1), chlorine-36 ( 36 C1), chlorine-37 ( 37 C1), bromine-79 ( 79 Br), bromine-81 ( 81 Br), iodine-123 ( 123 I), iodine-125 ( 125 I), iodine-127 ( 127 I), iodine-129 ( 129 I), and iodine-131 ( 131 I). In certain embodiments, an“isotopic variant” of a compound is in a stable form, that is, non radioactive. In certain embodiments, an“isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (¾), deuterium ( 2 H), carbon-12 ( 12 C), carbon-13 ( 13 C), nitrogen-14 ( 14 N), nitrogen-15 ( 15 N), oxygen-16 ( 16 0), oxygen-17 ( 17 0), oxygen-18 ( 18 0), fluorine-17 ( 17 F), phosphorus-31 ( 31 P), sulfur-32 ( 32 S), sulfur-33 ( 33 S), sulfur-34 ( 34 S), sulfur-36 ( 36 S), chlorine-35 ( 35 C1), chlorine-37 ( 37 C1), bromine-79 ( 79 Br), bromine-81 ( 81 Br), and iodine-127 ( 127 I). In certain embodiments, an“isotopic variant” of a compound is in an unstable form, that is, radioactive. In certain embodiments, an“isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium ( 3 H), carbon-11 ( n C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), oxygen-14 ( 14 0), oxygen-15 ( 15 0), fluorine-18 ( 18 F), phosphorus-32 ( 32 P), phosphorus-33 ( 33 P), sulfur-35 ( 35 S), chlorine-36 ( 36 C1), iodine-123 ( 123 I), iodine-125 ( 125 I), iodine-129 ( 129 I), and iodine-131 ( 131 I). It will be understood that, in a compound as provided herein, any hydrogen can be 2 H, for example, or any carbon can be 13 C, for example, or any nitrogen can be 15 N, for example, or any oxygen can be 18 0, for example, where feasible according to the judgment of one of skill. In certain embodiments, an“isotopic variant” of a compound contains unnatural proportions of deuterium (D).

[0076] The term“alkyl” refers to a linear or branched saturated monovalent hydrocarbon radical, wherein the alkylene may optionally be substituted with one or more substituents Q as described herein. The term “alkyl” also encompasses both linear and branched alkyl, unless otherwise specified. In certain embodiments, the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (Ci-20), 1 to 15 (Ci-15), 1 to 10 (Ci-10), or 1 to 6 (Ci-b) carbon atoms, or branched saturated monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. As used herein, linear Ci- 6 and branched C3-6 alkyl groups are also referred as“lower alkyl.” Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), «-propyl, isopropyl, butyl (including all isomeric forms), «-butyl, isobutyl, sec-butyl, /-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms). For example, Ci- 6 alkyl refers to a linear saturated monovalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.

[0077] The term“alkylene” refers to a linear or branched saturated divalent hydrocarbon radical, wherein the alkylene may optionally be substituted with one or more substituents Q as described herein. The term “alkylene” encompasses both linear and branched alkylene, unless otherwise specified. In certain embodiments, the alkylene is a linear saturated divalent hydrocarbon radical that has 1 to 20 (C i-20), 1 to 15 (Ci-15), 1 to 10 (Ci-10), or 1 to 6 (Ci-e) carbon atoms, or branched saturated divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. As used herein, linear Ci-6 and branched C3-6 alkylene groups are also referred as“lower alkylene.” Examples of alkylene groups include, but are not limited to, methylene, ethylene, propylene (including all isomeric forms), «- propylene, isopropylene, butylene (including all isomeric forms), «-butylene, isobutylene, /-butylene, pentylene (including all isomeric forms), and hexylene (including all isomeric forms). For example, Ci- 6 alkylene refers to a linear saturated divalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical of 3 to 6 carbon atoms.

[0078] The term“heteroalkylene” refers to a linear or branched saturated divalent hydrocarbon radical that contains one or more heteroatoms each independently selected from O, S, and N in the hydrocarbon chain. For example, Ci- 6 heteroalkylene refers to a linear saturated divalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the heteroalkylene is a linear saturated divalent hydrocarbon radical that has 1 to 20 (Ci-20),

1 to 15 (Ci- 15 ), 1 to 10 (Ci- 10 ), or 1 to 6 (Ci-b) carbon atoms, or branched saturated divalent hydrocarbon radical of 3 to 20 (C3- 20 ), 3 to 15 (C3- 15 ), 3 to 10 (C3- 10 ), or 3 to 6 (C3-6) carbon atoms. As used herein, linear Ci- 6 and branched C 3-6 heteroalkylene groups are also referred as“lower heteroalkylene.” Examples of heteroalkylene groups include, but are not limited to, -CH 2 O-, -CH 2 OCH 2 -, -CH 2 CH 2 O-, -CH 2 NH-, -CH 2 NHCH 2 -, -CH 2 CH 2 NH-, -CH 2 S-, -CH 2 SCH 2 -, and -CH 2 CH 2 S-. In certain embodiments, heteroalkylene may also be optionally substituted with one or more substituents Q as described herein.

[0079] The term“alkenyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon- carbon double bond(s). The alkenyl may be optionally substituted with one or more substituents Q as described herein. The term“alkenyl” also embraces radicals having cis and Ira ns configurations, or alternatively,“Z” and“E” configurations, as appreciated by those of ordinary skill in the art. As used herein, the term“alkenyl” encompasses both linear and branched alkenyl, unless otherwise specified. For example, C 2-6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C 2-20 ), 2 to 15 (C 2-15 ), 2 to 10 (C 2-10 ), or 2 to 6 (C 2-6 ) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C 3-20 ),

3 to 15 (C3- 15 ), 3 to 10 (C3- 10 ), or 3 to 6 (C3-6) carbon atoms. Examples of alkenyl groups include, but are not limited to, ethenyl, propen-1 -yl, propen-2 -yl, allyl, butenyl, and 4-methylbutenyl.

[0080] The term“alkenylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon- carbon double bond(s). The alkenylene may be optionally substituted with one or more substituents Q as described herein. Similarly, the term“alkenylene” also embraces radicals having“cis” and Ira ns configurations, or alternatively,“E” and“Z” configurations. As used herein, the term“alkenylene” encompasses both linear and branched alkenylene, unless otherwise specified. For example, C 2-6 alkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C 2-20 ), 2 to 15 (C 2-15 ), 2 to 10 (C 2-10 ), or 2 to 6 (C 2 -6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3- 20 ), 3 to 15 (C3- 15 ), 3 to 10 (C3- 10 ), or 3 to 6 (C 3-6 ) carbon atoms. Examples of alkenylene groups include, but are not limited to, ethenylene, allylene, propenylene, butenylene, and 4-methylbutenylene.

[0081] The term“heteroalkenylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon-carbon double bond(s), and which contains one or more heteroatoms each independently selected from O, S, and N in the hydrocarbon chain. The heteroalkenylene may be optionally substituted with one or more substituents Q as described herein. The term“heteroalkenylene” embraces radicals having a“cis” or“ trans” configuration or a mixture thereof, or alternatively, a“Z” or“E” configuration or a mixture thereof, as appreciated by those of ordinary skill in the art. For example, C2-6 heteroalkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the heteroalkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C 2-20 ), 2 to 15 (C 2-15 ), 2 to 10 (C 2-10 ), or 2 to 6 (C 2 -6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3- 20 ), 3 to 15 (C3- 15 ), 3 to 10 (C3- 10 ), or 3 to 6 (C3-6) carbon atoms. Examples of heteroalkenylene groups include, but are not limited to, -CH=CHO-, - CH=CH0CH 2 -, -CH=CHCH 2 0-, -CH=CHS-, -CH=CHSCH 2 -, -CH=CHCH 2 S-, or -CH=CHCH 2 NH-.

[0082] The term“alkynyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one, carbon- carbon triple bond(s). The alkynyl may be optionally substituted with one or more substituents Q as described herein. The term“alkynyl” also encompasses both linear and branched alkynyl, unless otherwise specified. In certain embodiments, the alkynyl is a linear monovalent hydrocarbon radical of 2 to 20 (C 2-20 ), 2 to 15 (C 2-15 ), 2 to 10 (C 2-10 ), or 2 to 6 (C 2 -6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3- 20 ), 3 to 15 (C3- 15 ), 3 to 10 (C3- 10 ), or 3 to 6 (C3-6) carbon atoms.

Examples of alkynyl groups include, but are not limited to, ethynyl ( - C º CH) and propargyl (-CH 2 C º CH). For example, C 2-6 alkynyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.

[0083] The term“cycloalkyl” refers to a cyclic saturated bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted with one or more substituents Q as described herein. In certain embodiments, the cycloalkyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, decalinyl, and adamantyl.

[0084] The term“cycloalkenyl” refers to a cyclic unsaturated, nonaromatic bridged and/or non-bridged monovalent hydrocarbon radical, which may be optionally substituted with one or more substituents Q as described herein. In certain embodiments, the cycloalkenyl has from 3 to 20 (C 3-20 ), from 3 to 15 (C 3-15 ), from 3 to 10 (C 3-10 ), or from 3 to 7 (C 3-7 ) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclobutenyl, cyclopentenyl, cyclohexenyl, or cycloheptenyl.

[0085] The term“aryl” refers to a monocyclic aromatic group and/or multicyclic monovalent aromatic group that contain at least one aromatic hydrocarbon ring. In certain embodiments, the aryl has from 6 to 20 (G,-2O). from 6 to 15 (Ce-is), or from 6 to 10 (CV ) ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). In certain embodiments, aryl may be optionally substituted with one or more substituents Q as described herein.

[0086] The terms“aralkyl” and“arylalkyl” refer to a monovalent alkyl group substituted with one or more aryl groups. In certain embodiments, the aralkyl has from 7 to 30 (C7-30), from 7 to 20 (C7-20), or from 7 to 16 (C7-16) carbon atoms. Examples of aralkyl groups include, but are not limited to, benzyl, 2- phenylethyl, and 3-phenylpropyl. In certain embodiments, the aralkyl are optionally substituted with one or more substituents Q as described herein.

[0087] The term“heteroaryl” refers to a monovalent monocyclic aromatic group or monovalent polycyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, N, and P in the ring. A heteroaryl group is bonded to the rest of a molecule through its aromatic ring. Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, one to four N atoms, and/or one or two P atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. In certain embodiments, the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. In certain embodiments, the heteroaryl may also be optionally substituted with one or more substituents Q as described herein as described herein.

[0088] The terms“heterocyclyl” and“heterocyclic” refer to a monovalent monocyclic non-aromatic ring system or monovalent polycyclic ring system that contains at least one non-aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, N, and P; and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms. A heterocyclyl group is bonded to the rest of a molecule through its non-aromatic ring. In certain embodiments, the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may be spiro, fused, or bridged, and in which nitrogen or sulfur atoms may be optionally oxidized, nitrogen atoms may be optionally quatemized, and some rings may be partially or fully saturated, or aromatic. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic groups include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl,

benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, b-carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl,

dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl,

octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4- piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl,

tetrahydroquinolinyl, and 1,3,5-trithianyl. In certain embodiments, the heterocyclyl may also be optionally substituted with one or more substituents Q as described herein.

[0089] The terms“halogen”,“halide” and“halo” refer to fluorine, chlorine, bromine, and/or iodine.

[0090] The term“optionally substituted” is intended to mean that a group or substituent, such as an alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, cycloalkenyl, aryl, aralkyl, heteroaryl, heteroaryl-Ci-e alkyl, and heterocyclyl group, may be substituted with one or more substituents Q, each of which is independently selected from, e.g., (a) oxo (=0), halo, cyano (-CN), and nitro (-NO2); (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment,

P(0)(OR a )R d , -P(0)(OR a )(OR d ), -SR a , -S(0)R a , -S(0) 2 R a , -S(0)NR b R c , and -S(0) 2 NR b R c , wherein each R a , R b , R c , and R d is independently (i) hydrogen; (ii) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, C 14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; or (iii) R b and R c together with the N atom to which they are attached form heteroaryl or heterocyclyl, optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a . As used herein, all groups that can be substituted are“optionally substituted,” unless otherwise specified.

[0091] In one embodiment, each substituent Q a is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; and (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)R e , -C(0)OR e , -C(0)NR f R g , -C(NR e )NR f R g , -OR e , - R g , - f , R g , and R h is independently (i) hydrogen, Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (ii) R f and R g together with the N atom to which they are attached form heteroaryl or heterocyclyl.

[0092] In certain embodiments,“optically active” and“enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the racemate in question.

[0093] In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The (+) and (-) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (-) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (-), is not related to the absolute configuration of the molecule, R and S.

[0094] The phrase“an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof’ has the same meaning as the phrase“an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of the compound referenced therein; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein.”

[0095] The term“solvate” refers to a complex or aggregate formed by one or more molecules of a solute, e.g., a compound provided herein, and one or more molecules of a solvent, which present in a

stoichiometric or non-stoichiometric amount. Suitable solvents include, but are not limited to, water, methanol, ethanol, «-propanol, isopropanol, and acetic acid. In certain embodiments, the solvent is pharmaceutically acceptable. In one embodiment, the complex or aggregate is in a crystalline form. In another embodiment, the complex or aggregate is in a noncrystalline form. Where the solvent is water, the solvate is a hydrate. Examples of hydrates include, but are not limited to, a hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate, and pentahydrate.

[0096] The terms“resistent,”“relapsed,” or“refractory” refer to a cancer that has a reduced

responsiveness to a treatment, e.g., the point at which the cancer does not respond to attempted forms of treatment. The cancer can be resistant at the beginning of treatment or it may become resistant during treatment. The term“refractory” can refer to a cancer for which treatment (e.g., chemotherapy drugs, biological agents, and/or radiation therapy) has proven to be ineffective. A refractory cancer tumor may shrink, but not to the point where the treatment is determined to be effective. Typically however, the tumor stays the same size as it was before treatment (stable disease), or it grows (progressive disease).

[0097] The terms“intermittent dosing schedule” or“IS” refer to drugs (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) dosed or administered less than once daily. In some embodiments herein, IS refers to dosing or administration of a drug (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) to a subject once daily for a period of about 7 days in a 28-day cycle. In other embodiments herein, IS refers to dosing or administration of a drug (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) daily for up to three 28-day cycles (e.g., two 28-day cycles) and, in the third cycle and subsequent cycles, dosing or administration of the drug to the subject once daily for a period of about 7 days in a 28-day cycle. In some embodiments, IS is continued until progression of disease occurs/is observed or until an incidence of at least one toxicity is reduced.

[0098] The terms“continuous dosing schedule” or“CS” refer to drugs (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) dosed or administered once daily. In some embodiments herein, CS refers to dosing or administration of a drug (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) to a subject daily in a 28-day cycle. In other embodiments herein, CS refers to dosing or administration of a drug (e.g., the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof) daily for > three 28-day cycles and, in the one or more subsequent cycles, the drug is dosed or administered to the subject once daily for a period of about 7 days in a 28-day cycle (i.e., a late switch to IS). In some embodiments, the subject on CS is never switched to IS. In some embodiments, the subject on CS is switched to IS after > three 28-day cycles (i.e., late switch to IS). In some embodiments, CS is continued until intolerable toxicity occurs/is observed.

[0099] “Responsiveness” or to“respond” to treatment, and other forms of this term, as used herein, refer to the reaction of a subject to treatment with a therapeutic, e.g., a PI3K inhibitor, alone or in combination, e.g., monotherapy or combination therapy. Responsiveness to a therapy, e.g., treatment with a PI3K inhibitor alone or in combination, can be evaluated by comparing a subject's response to the therapy using one or more clinical criteria, such as IWCLL 2008 (for CLL) described in, e.g., Hallek, M. et al. (2008) Blood 111 (12): 5446-5456; the Lugano Classification described in, e.g., Cheson, B.D. et al. Journal of Clinical Oncology, 32(27): 3059-3067; and the like. Additional classifications of responsiveness are provided by. These criteria provide a set of published rules that define when cancer patients improve (“respond”), stay the same (“stable”) or worsen (“progression”) during treatments.

[0100] For example, a subject having CLL can be determined to be in complete remission (CR) or partial remission (PR). For example, according to IWCLL 2008, a subject is considered to be in CR if at least all of the following criteria as assessed after completion of therapy are met: (i) Peripheral blood lymphocytes (evaluated by blood and different count) below 4 x 10 9 /L (4000 pi); (ii) no hepatomegaly or splenomegaly by physical examination; (iii) absence of constitutional symptoms; and (iv) blood counts (e.g., neutrophils, platelets, hemoglobin) above the values set forth in Hallek, M. et al. Partial remission (PR) for CLL is defined according to IWCLL 2008 as including one of: (i) a decrease in number of blood lymphocytes by 50% or more from the value before therapy; (ii) a reduction in lymphadenopathy, as detected by CT scan or palpation; or (iii) a reduction in pretreatment enlargement of spleen or liver by 50% or more, as detected by CT scan or palpation; and blood counts (e.g., neutrophils, platelets, hemoglobin) according to the values set forth in Hallek, M. et al. In other embodiments, a subject having CLL is determined to have progressive disease (PD) or stable disease (SD). For example, according to IWCLL 2008, a subject is considered to be in PD during therapy or after therapy if at least one of the following criteria is met: (i) progression on lymphadenopathy; (ii) an increase in pretreatment enlargement of spleen or liver by 50% or more, or de novo appearance of hepatomegaly or splenomegaly; (iii) an increase in the number of blood lymphocytes by 50% or more with at least 5000 B lymphocytes per microliter; (iv) transformation to a more aggressive histology (e.g., Richter syndrome); or (v) occurrence of cytopenia (neutropenia, anemia or thrombocytopenia) attributable to CLL. Stable disease (SD) for CLL is defined according to IWCLL 2008 as a patient who has not achieved CR or a PR, and who has not exhibited progressive disease.

[0101] For example, in some embodiments, a subject with CLL responds to treatment with a PI3K inhibitor, alone or in combination, if at least one of the criteria for disease progression according to IWCLL is retarded or reduced, e.g., by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more. In another example, a subject responds to treatment with a PI3K inhibitor, alone or in combination, if the subject experiences a life expectancy extension, e.g., extended by about 5%, 10%, 20%, 30%, 40%, 50% or more beyond the life expectancy predicted if no treatment is administered. In another example, a subject responds to treatment with a PI3K inhibitor, alone or in combination, if the subject has one or more of: an increased progression-free survival, overall survival or increased time to progression (TTP), e.g., as described in Hallek, M. et al.

PI3K Inhibitors

[0102] Some embodiments provided herein describe pharmaceutical compositions or methods for using the pharmaceutical compositions comprising a PI3K inhibitor described herein in combination with a T- cell activating bispecific antigen -binding molecule. In some embodiments, the PI3K inhibitor is a PI3K5 inhibitor.

[0103] In some embodiments, the PI3K inhibitor has structural Formula (I): (I),

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof;

wherein:

X, Y, and Z are each independently N or CR X , with the proviso that at least two of X, Y, and Z are

nitrogen atoms; where R x is hydrogen or Ci- 6 alkyl;

R 1 and R 2 are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -

S(0)NR lb R lc , or -S(0) 2 NR lb R lc ; wherein each R la , R lb , R lc , and R ld is independently (i) hydrogen; (ii) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R lb and R lc together with the N atom to which they are attached form heterocyclyl;

R 3 and R 4 are each independently hydrogen or Ci- 6 alkyl; or R 3 and R 4 are linked together to form a bond, Ci-6 alkylene, Ci-6 heteroalkylene, C2-6 alkenylene, or

C2-6 heteroalkenylene;

R 5a is (a) hydrogen or halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CY 14 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , -C(0)NR lb R lc , -C(NR la )NR lb R lc ,

R 5b is (a) halo; (b) Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl,

R 5c is -(CR 5f R 5g ) n -(C 6 i 4 aryl) or -(CR 5f R 5g ) n -heteroaryl;

R 5d and R 5e are each independently (a) hydrogen or halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , - NR la S(0) 2 R ld , -NR la S (0)NR lb R lc , -NR la S(0) 2 NR lb R lc , -SR la , -S(0)R la , -S(0) 2 R la , -

S(0)NR lb R lc , or -S(0) 2 NR lb R lc ;

R 5f and R 5g are each independently (a) hydrogen or halo; (b) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , -

S(0)NR lb R lc ; or -S(0) 2 NR lb R lc ; or (d) when one occurrence of R 5f and one occurrence of R 5g are attached to the same carbon atom, the R 5f and R 5g together with the carbon atom to which they are attached form a C3-10 cycloalkyl or heterocyclyl;

R 6 is hydrogen, Ci-6 alkyl, -S-Ci- 6 alkyl, -S(0)-Ci- 6 alkyl, or -S0 2- Ci- 6 alkyl;

m is 0 or 1 ; and

n is 0, 1, 2, 3, or 4;

wherein each alkyl, alkylene, heteroalkylene, alkenyl, alkenylene, heteroalkenylene, alkynyl, cycloalkyl, aryl, aralkyl, heteroaryl, and heterocyclyl in R 1 , R 2 , R 3 , R 4 , R 6 , R x , R la , R lb , R lc , R ld , R 5a , R 5b , R 5c , R 5d , R 5e , R 5f , and R 5g is optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q, wherein each substituent Q is independently selected from (a) oxo, cyano, halo, and nitro; (b) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two,

S(0)NR b R c , and -S(0) 2 NR b R c , wherein each R a , R b , R c , and R d is independently (i) hydrogen; (ii) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is further optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; or (iii) R b and R c together with the N atom to which they are attached form heterocyclyl, which is further optionally substituted with one, two, three, or four substituents Q a ;

wherein each Q a is independently selected from the group consisting of (a) oxo, cyano, halo, and nitro; (b) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, and heterocyclyl; and (c) -C(0)R e , -C(0)0R e , -C(0)NR f R g , -C(NR e )NR f R g , -OR e , -0C(0)R e , -

S(0)NR f R g , and -S(0) 2 NR f R g ; wherein each R e , R f , R g , and R h is independently (i) hydrogen; (ii) Ci- 6 alkyl, C 2-r , alkenyl, C 2-r , alkynyl, C3-10 cycloalkyl, Ce-w aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (iii) R f and R g together with the N atom to which they are attached form

heterocyclyl;

wherein two substituents Q that are adjacent to each other optionally form a

C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a .

[0104] In some embodiments, the compound of structural Formula (I) is not 4-(2-(difluoromethyl)- l//- benzo[r/]imidazol-l-yl)-6-morpliolino-/V-(2 -phenyl -2-(pyrrolidin-l-yl)ethyl)-l, 3, 5-triazin-2 -amine or 6-(2- (difluoromcthyl)- 1 //-bcnzo| £/|imidazol- 1 -yl)-A'-( 1 -(4-((/Z)-3-(methoxymethyl)morpholino)phenyl)ethyl)- 2-morpholinopyrimidin -4-amine.

[0105] In one embodiment of a compound of Formula (I), X, Y, and Z are each independently N or CR X ’ with the proviso that at least two of X, Y, and Z are nitrogen atoms; where R x is hydrogen or Ci- 6 alkyl. In another embodiment of a compound of Formula (I), X, Y, and Z are N.In some embodiments, R 5b is (a) halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C 3-10 cycloalkyl, CY aryl, C 7-15 aralkyl, or heteroaryl; or (c)

[0106] In some embodiments, R 5a and R 5b are each independently

(a) halo; (b) Ci- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-10 cycloalkyl, Ce-u aryl, C 7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)OR la , -C(0)NR lb R lc , -C(NR la )NR lb R lc ,

[0107] In some embodiments, R 5a and R 5b are each methyl, optionally substituted with one or more halo.

[0108] In some embodiments, R 5f and R 5g are each hydrogen.

[0109] In some embodiments of compounds of structural Formula (I):

X, Y, and Z are each N;

R 1 and R 2 are each hydrogen;

R 3 and R 4 are each hydrogen;

R 5a is Ci-6 alkyl;

R 5b is Ci-6 alkyl;

R 5c is -(CH2)-phenyl, wherein R 5c is optionally substituted with one, two, three, or four substituents Q;

R 5d and R 5e are each hydrogen;

R 6 is CHF2; and

m is 0; wherein each alkyl is optionally substituted with one, two, three, or four substituents Q, wherein each substituent Q is independently selected from Ce-u aryl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Q a , wherein the heteroaryl has from 5 to 10 ring atoms and one or more heteroatoms independently selected from O, S, and N, and the heterocyclyl has from 3 to 15 ring atoms and one or more heteroatoms independently selected from O, S, and N;

wherein each Q a is independently selected from the group consisting of halo, Ci- 6 alkyl, Ci- 6 alkylsulfonyl and -OR e , wherein R e is hydrogen or Ci- 6 alkyl.

[0110] In some embodiments of compounds of structural Formula (I):

X, Y, and Z are each N;

R 1 and R 2 are each hydrogen;

R 3 and R 4 are each hydrogen;

R 5a and R 5b are each methyl, optionally substituted with one or more halo;

R 5c is -(CH2)-phenyl, wherein R 5c is optionally substituted with one, two, three, or four substituents Q;

R 5d and R 5e are each hydrogen;

R 6 is CHF2; and

m is 0;

wherein each alkyl is optionally substituted with one, two, three, or four substituents Q, wherein each substituent Q is independently selected from Ce-u aryl, heteroaryl, and heterocyclyl, each of which is further optionally substituted with one, two, three, or four substituents Q a , wherein the heteroaryl has from 5 to 10 ring atoms and one or more heteroatoms independently selected from O, S, and N, and the heterocyclyl has from 3 to 15 ring atoms and one or more heteroatoms independently selected from O, S, and N;

wherein each Q a is independently selected from the group consisting of halo, Ci- 6 alkyl, Ci- 6 alkylsulfonyl and -OR e , wherein R e is hydrogen or Ci- 6 alkyl.

[0111] Provided herein is a compound of Formula (II):

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, R 5c is C 14 aryl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is phenyl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is naphthyl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is -(CR 5f R 5g ) n -(C6 i4 aryl), wherein the aryl is optionally substituted with one or more substituents Q. In some embodiments, R 5c is -(CH2)-phenyl, wherein the phenyl is optionally substituted with one or more substituents Q. In some embodiments, R 5c is -(CF^-naphthyl, wherein the naphthyl is optionally substituted with one or more substituents Q. In some embodiments, R 5c is heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is monocyclic heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is 5- or 6-membered heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is bicyclic heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 5c is -(CR 5f R 5g ) n -heteroaryl, wherein the heteroaryl is optionally substituted with one or more substituents Q. In some embodiments, R 5c is - (CR 5f R 5g ) n -(monocyclic heteroaryl), wherein the heteroaryl is optionally substituted with one or more substituents Q. R 5c is -(CR 5f R 5g ) n -(5- or 6-membered heteroaryl), wherein the heteroaryl is optionally substituted with one or more substituents Q. In some embodiments, R 5c is -(CR 5f R 5g ) n -(bicyclic heteroaryl), wherein the heteroaryl is optionally substituted with one or more substituents Q.

[0112] Also provided herein is a compound of Formula (VII):

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof,

wherein:

R 7a , R 715 , R 7c , R 7d , and R 7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, four, or five

,

R 7b , R 7c , R 7d , and R 7e that are adjacent to each other form C3-10 cycloalkenyl, Ce-i4 aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q.

[0113] Also provided herein is a compound of Formula (IX):

or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, wherein:

R 7a , R 715 , R 7c , R 7d , and R 7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; or (c) -C(0)R a , -C(0)0R a , -C(0)NR b R c , -C(NR a )NR b R c , -OR a , -0C(0)R a , -

S(0)NR b R c , or -S(0) 2 NR b R c ; or two of R 7a , R 7b , R 7c , R 7d , and R 7e that are adjacent to each other form C3-10 cycloalkenyl, CVu aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a .

[0114] In some embodiments, R 7a is hydrogen, halo, Ci- 6 alkyl optionally substituted with one or more substituents Q, or -OR la .

[0115] In some embodiments, R 7a is hydrogen. In some embodiments, R 7a is (a) cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, four, or five

-S(0) 2 NR lb R lc . In some embodiments, R 7a is (i) halo; (ii) Ci- 6 alkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q; or (iii) -OR la or -NR lb R lc .

[0116] In some embodiments, R 7b is hydrogen, halo, C M, alkyl optionally substituted with one or more substituents Q, or -OR la . In some embodiments, R 7b is hydrogen.

[0117] In some embodiments, R 7c is hydrogen, halo, C , alkyl optionally substituted with one or more substituents Q, or -OR la . In some embodiments, R 7c is hydrogen, halo, or -OR la . In some embodiments, R 7c is chloro. In some embodiments, R 7c is -O-Ci- 6 alkyl, optionally substituted with one or more substituents Q. [0118] In some embodiments, R 7d is hydrogen, halo, C , alkyl optionally substituted with one or more substituents Q, or -OR la . In some embodiments, R 7d is hydrogen.

[0119] In some embodiments, R 7e is hydrogen, halo, C , alkyl optionally substituted with one or more substituents Q, or -OR la . In some embodiments, R 7e is hydrogen. In some embodiments, two of R 7a , R 7b , R 7c , R 7d , and R 7e that are adjacent to each other form C3-10 cycloalkenyl, CVu aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q. In some embodiments, R 7a and FC b together with the carbon atoms to which they are attached form Ce-u aryl, optionally substituted with one or more substituents Q.

[0120] In some embodiments, R 5a is hydrogen. In some embodiments, R 5a is Ci- 6 alkyl, optionally substituted with one or more substituents Q. In some embodiments, R 5a is hydrogen, methyl, or ethyl.

[0121] In some embodiments, R 5b is Ci- 6 alkyl, optionally substituted with one or more substituents Q. In some embodiments, R 5b is methyl, ethyl, or propyl. In some embodiments, R 5b is -C(0)OR la . In some embodiments, R 5b is -C(0)0-Ci-r, alkyl. In some embodiments, R 5b is -C(0)OC]¾.

[0122] Also provided herein is a compound of Formula (X):

Formula (X), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0123] Provided herein is a compound of Formula (XI):

Formula (XI), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, wherein:

R 7a , R 7b . R 7c , R 7d , and R 7e are each independently (a) hydrogen, cyano, halo, or nitro; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, CVu aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; or (c) -C(0)R a , -C(0)0R a , -C(0)NR b R c , -C(NR a )NR b R c , -OR a , -0C(0)R a , -

S(0)NR b R c , or -S(0) 2 NR b R c ; or two of R 7a , R 7b , R 7c , R 7d , and R 7e that are adjacent to each other form C3-10 cycloalkenyl, Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a .

[0124] In certain embodiments, R 5a and R 5b are each independently (a) halo; (b) Ci- 6 alkyl, C 2-r , alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-u aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -

S(0) 2 NR lb R lc . In certain embodiments, one of R 7a , R 7b , R 7c , R 7d , and R 7e is Ce-u aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; in certain embodiments, one of R 7a , R 7b , R 7c , R 7d , and R 7e is CVu aryl, e.g., phenyl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, one of R 7a , R b . R 7c , R 7d , and R 7e is heteroaryl, e.g., 5-membered or 6-membered heteroaryl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, one of R 7a , R 7b , R 7c , R 7d , and R 7e is heterocyclyl, e.g., 5-membered or 6-membered heterocyclyl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, one of R 7a , R b . R 7c , R 7d , and R 7e is phenyl, imidazolyl, pyrozolyl, pyridinyl, piperidinyl, or piperazinyl, each optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, one of R 7a , R b . R 7c , R 7d , and R 7e is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each optionally substituted with one or more substituents Q a ; in certain embodiments, one of R 7a , R 7b , R 7c , R 7d , and R 7e is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3- methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-methoxyphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2- methylpyridin-4-yl, 2-methoxypyridin-4-yl, l-methylpiperidin-4-yl, or 4-methylpiperazin-l-yl; and in certain embodiments, one of R 7a , R 7b , R 7c , R 7d , and R 7e is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2- bromophenyl, 2-methylphenyl, 2-(3-dimethylaminopropyl)phenyl, 2-methoxyphenyl, 3 -fluorophenyl, 3- chlorophenyl, 3-methylphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4- methoxyphenyl, 2,4-difluorophenyl, 2,6-difluorophenyl, 4-fluoro-3-methoxyphenyl, 3-methoxyphenyl, 4- methoxyphenyl, 3-morpholin-4-ylmethylphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2- methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2-fluoropyridin-3-yl, 2-methylpyridin-4-yl, 2- (4-methylpiperazin-l-yl)pyridin-4-yl, 2-methoxypyridin-4-yl, pyrimidin-5-yl, pyrrolidin-3-yl, 1- methylpyrrolidin-3-yl, piperidin-4-yl, 1 -methylpiperidin-4-yl, l-ethylpiperidin-4-yl, 1-isopropylpiperidin- 4-yl, l-acetylpiperidin-4-yl, l-methylsulfonylpiperidin-4-yl, or 4-methylpiperazin-l-yl. [0125] In certain embodiments , R 7a is CVu aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more, in one embodiment, one, two, three, or four substituents Q a ; in certain embodiments, R 7a is CVu aryl, e.g., phenyl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, R 7a is heteroaryl, e.g., 5-membered or 6-membered heteroaryl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, R 7a is heterocyclyl, e.g., 5-membered or 6-membered heterocyclyl, optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, piperidinyl, or piperazinyl, each optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each optionally substituted with one, two, three, or four substituents Q a ; in certain embodiments, R 7a is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2- methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4- bromophenyl, 4-methoxyphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3- yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2-methylpyridin-4-yl, 2-methoxypyridin-4-yl, 1- methylpiperidin-4-yl, or 4-methylpiperazin-l-yl; and in certain embodiments, R 7a is phenyl, 2- fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-(3-dimethylaminopropyl)phenyl, 2- methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3-methylphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4- chlorophenyl, 4-bromophenyl, 4-methoxyphenyl, 2,4-difluorophenyl, 2,6-difluorophenyl, 4-fluoro-3- methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 3-morpholin-4-ylmethylphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2- fluoropyri din-3 -yl, 2-methylpyridin-4-yl, 2-(4-methylpiperazin- 1 -yl)pyridin-4-yl, 2-methoxypyridin-4-yl, pyrimidin-5-yl, pyrrolidin-3-yl, l-methylpyrrolidin-3-yl, piperidin-4-yl, l-methylpiperidin-4-yl, 1- ethylpiperidin-4-yl, l-isopropylpiperidin-4-yl, l-acetylpiperidin-4-yl, l-methylsulfonylpiperidin-4-yl, or 4-methylpiperazin- 1 -yl .

[0126] In certain embodiments:

R 1 is hydrogen or -OR la , where R la is Ci-e alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 5a and R 5b are each independently Ci-e alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 5f and R 5g are each independently hydrogen, halo, Ci-e alkyl, optionally substituted with one, two, three, four, or five substituents Q; or R 5f and R 5g together with the carbon atom to which they are attached form C HO cycloalkyl or heterocyclyl, each of which is optionally substituted with one, two, three, four, or five substituents Q;

R 7a is C aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ; R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CR X , with the proviso that at least two of X, Y, and Z are N; where R x is a hydrogen or Ci- 6 alkyl, optionally substituted with one, two, three, or four substituents Q a .

[0127] In certain embodiments:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one or more halo;

R 5a and R 5b are each independently Ci- 6 alkyl;

R 5f and R 5g are each independently hydrogen or Ci- 6 alkyl; or R 5f and R 5g together with the carbon atom to which they are attached form Ci-io cycloalkyl;

R 7a is Ce-i aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0128] In certain embodiments:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are methyl;

R 5f and R 5g are hydrogen; or R 5f and R 5g together with the carbon atom to which they are attached form cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;

R 7a is C aryl, monocyclic heteroaryl, or monocyclic heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0129] In certain embodiments:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are methyl;

R 5f and R 5g are hydrogen; or R 5f and R 5g together with the carbon atom to which they are attached form cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;

R 7a is phenyl, 5- or 6-membered heteroaryl, or 5- or 6-membered heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ; R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0130] In certain embodiments:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are methyl;

R 5f and R 5g are hydrogen; or R 5f and R 5g together with the carbon atom to which they are attached form cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl;

R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0131] In certain embodiments, R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, piperidinyl, or piperazinyl, each of which is optionally substituted with one, two, three, or four substituents Q a .

[0132] Provided herein is a compound of Formula (XVI):

Formula (XVI), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0133] In some embodiments, R 5a is Ci- 6 alkyl, optionally substituted with one or more substituents Q. In some embodiments, R 5a is methyl.

[0134] In some embodiments, R 5b is Ci- 6 alkyl, optionally substituted with one or more substituents Q. In some embodiments, R 5b is methyl.

[0135] In some embodiments, R 5a and R 5b are methyl.

[0136] In some embodiments, R 7a is hydrogen, halo, Ci- 6 alkyl, Ce-u aryl, heteroaryl, or heterocyclyl, where the alkyl, aryl, heteroaryl, and heterocyclyl are each optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q. In some embodiments, R 7a is Ce-w aryl, optionally substituted with one or more substituents Q. In some embodiments, R 7a is phenyl, optionally substituted with one or more substituents Q In some embodiments, R 7a is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-(3-dimethylaminopropyl)phenyl,

2-methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3-methylphenyl, 3-methoxyphenyl, 4-florophenyl, 4-chlorophenyl, 4-bromophenyl, 4-methoxyphenyl, 2,4-difluorophenyl,

2,6-difluorophenyl, 4-fluoro-3-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, or

3-morpholin-4-ylmethylphenyl. In some embodiments, R 7a is heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 7a is monocyclic heteroaryl, optionally substituted with one or more substituents Q. In some embodiments, R 7a is 5- or 6-membered heteroaryl, each optionally substituted with one or more substituents Q. In some embodiments, R 7a is imidazolyl, pyrozolyl, pyridinyl, or pyrimidinyl, each optionally substituted with one or more substituents Q. In some embodiments, R 7a is imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2- yl, pyridin-3-yl, pyridin-4-yl, 2-fluoropyridin-3-yl, 2-methylpyridin-4-yl, 2-(4-methylpiperazin-l- yl)pyridin-4-yl, 2-methoxypyridin-4-yl, pyrimidin-5-yl. In some embodiments, R 7a is heterocyclyl, optionally substituted with one or more substituents Q. In some embodiments, R 7a is monocyclic heterocyclyl, optionally substituted with one or more substituents Q. In some embodiments, R 7a is 5- or 6- membered heterocyclyl, each optionally substituted with one or more substituents Q. In some

embodiments, R 7a is pyrrolidinyl, piperidinyl, or piperazinyl, each optionally substituted with one or more substituents Q. In some embodiments, R 7a is pyrrolidin-3-yl, l-methylpyrrolidin-3-yl, piperidin-4-yl, 1- methylpiperidin-4-yl, l-ethylpiperidin-4-yl, l-isopropylpiperidin-4-yl, 1 -acetylpiperidin-4-yl, 1- methylsulfonylpiperidin-4-yl, or 4-methylpiperazin-l-yl.

[0137] In some embodiments, R 7b is hydrogen, halo, or Ci- 6 alkyl optionally substituted with one or more substituents Q. In some embodiments, R 7b is hydrogen.

[0138] In some embodiments, R 7c is hydrogen, halo, or Ci- 6 alkyl optionally substituted with one or more substituents Q. In some embodiments, R 7c is hydrogen.

[0139] In some embodiments, R 7d is hydrogen, halo, or C M, alkyl optionally substituted with one or more substituents Q. In some embodiments, R 7d is hydrogen.

[0140] In some embodiments, R 7e is hydrogen, halo, or C , alkyl optionally substituted with one or more substituents Q. In some embodiments, R 7e is hydrogen.

[0141] In some embodiments, R 7a is Ce-u aryl, heteroaryl, or heterocyclyl, each optionally substituted with one or more, in one embodiment, one, two, three, four, or five substituents Q; and R 7b , R 7c , R 7d , and R 7e are hydrogen.

[0142] In one embodiment of a compound of Formula (XVI), one of R 7a , R 7b . R 7c , R 7d , and R 7e is CYu aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 7b , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0143] In another embodiment of a compound Formula (XVI), one of R 7a , R 7b , R 7c , R 7d , and R 7e is Ce-u aryl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , |;] qc remaining of R 7a , R 715 , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0144] In yet another embodiment of a compound of Formula (XVI), one of R 7a , R 71 ’, R 7c , R 7d , and R 7e is heteroaryl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 7b , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein. [0145] In yet another embodiment of a compound of Formulae (XVI), one of R 7a , R 7b . R 7c , R 7d , and R 7e is heterocyclyl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 7b , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0146] In yet another embodiment of a compound of Formula (XVI), one of R 7a , R 71 ’, R 7c , R 7d , and R 7e is 5-membered or 6-membered heterocyclyl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 7b , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0147] In yet another embodiment of a compound of Formula (XVI), one of R 7a , R 715 , R 7c , R 7d , and R 7e is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 7b , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0148] In yet another embodiment of a compound of Formula (XVI), one of R 7a , R 715 , R 7c , R 7d , and R 7e is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-(3- dimethylaminopropyl)phenyl, 2-methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3-methylphenyl, 3- methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-methoxyphenyl, 2,4-difluorophenyl, 2,6-difluorophenyl, 4-fluoro-3-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 3-morpholin-4- ylmethylphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2-fluoropyridin-3-yl, 2-methylpyridin-4-yl, 2-(4-methylpiperazin-l-yl)pyridin- 4-yl, 2-methoxypyridin-4-yl, pyrimidin-5-yl, pyrrolidin-3-yl, l-methylpyrrolidin-3-yl, piperidin-4-yl, 1- methylpiperidin-4-yl, l-ethylpiperidin-4-yl, l-isopropylpiperidin-4-yl, 1 -acetylpiperidin-4-yl, 1- methylsulfonylpiperidin-4-yl, or 4-methylpiperazin-l-yl.

[0149] In still another embodiment of a compound of Formula (XVI), one of R 7a , R 71 ’, R 7c , R 7d , and R 7e is phenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-methoxyphenyl, 3- fluorophenyl, 3-chlorophenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4- methoxyphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2-methylpyridin-4-yl, 2-methoxypyridin-4-yl, l-methylpiperidin-4-yl, or 4- methylpiperazin-l-yl; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , the remaining of R 7a , R 715 , R 7c , R 7d , and R 7e , X, Y, and Z are each as defined herein.

[0150] In one embodiment of a compound of Formula (XVI), R 7a is Ce-u aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5S , RY Y RY RY R Y X. Y. and Z are each as defined herein.

[0151] In yet another embodiment of a compound of Formula (XVI), R 7a is heterocyclyl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , R 7b ,

R 7c , R 7d , R 7e , X, Y, and Z are each as defined herein.

[0152] In yet another embodiment of a compound of Formula (XVI), R 7a is 5-membered or 6-membered heterocyclyl, which is optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , R 7b , R 7c , R 7d , R 7e , X, Y, and Z are each as defined herein. [0153] In yet another embodiment of a compound of Formula (XVI), R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each optionally substituted with one, two, three, or four substituents Q a ; and R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , R 7b . R 7c , R 7d , R 7e , X, Y, and Z are each as defined herein.

[0154] In yet another embodiment of a compound of Formula (XVI), R 7a is phenyl, 2-fluorophenyl, 2- chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-(3-dimethylaminopropyl)phenyl, 2-methoxyphenyl, 3- fluorophenyl, 3-chlorophenyl, 3-methylphenyl, 3-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4- bromophenyl, 4-methoxyphenyl, 2,4-difluorophenyl, 2,6-difluorophenyl, 4-fluoro-3-methoxyphenyl, 3- methoxyphenyl, 4-methoxyphenyl, 3-morpholin-4-ylmethylphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl - pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2-fluoropyridin-3-yl, 2- methylpyridin-4-yl, 2-(4-methylpiperazin-l -yl)pyridin-4-yl, 2-methoxypyridin-4-yl, pyrimidin-5-yl, pyrrolidin-3-yl, l-methylpyrrolidin-3-yl, piperidin-4-yl, l-methylpiperidin-4-yl, l-ethylpiperidin-4-yl, 1- isopropylpiperidin-4-yl, l-acetylpiperidin-4-yl, l-methylsulfonylpiperidin-4-yl, or 4-methylpiperazin-l-yl.

[0155] In yet another embodiment of a compound of Formula (XVI), R 7a is phenyl, 2-fluorophenyl, 2- chlorophenyl, 2-bromophenyl, 2-methylphenyl, 2-methoxyphenyl, 3 -fluorophenyl, 3-chlorophenyl, 3- methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-methoxyphenyl, imidazol-l-yl, pyrozol-4-yl, 1 -methyl -pyrozol-4-yl, 2-methylpyrozol-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 2- methylpyridin-4-yl, 2-methoxypyridin-4-yl, l-methylpiperidin-4-yl, or 4-methylpiperazin-l-yl; and R 1 ,

R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , R 7b , R 7c , R 7d , R 7e , X, Y, and Z are each as defined herein.

[0156] In one embodiment of a compound of Formula (XVI),

R 1 is hydrogen or -OR la , where R la is Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 5a and R 5b are each independently Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 7a is C aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one or more substituents Q a ; and

R 7b , R 7c , R 7d , and R 7e are hydrogen.

[0157] In one embodiment of a compound of Formula (XVI):

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one or more halo;

R 5a and R 5b are each independently Ci- 6 alkyl;

R 7a is C aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ; and

R 7b , R 7c , R 7d , and R 7e are hydrogen.

[0158] In one embodiment of a compound of Formula (XVI):

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are methyl;

R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each of which is optionally substituted with one, two, three, four, or five substituents Q; and R 7b , R 7c , R 7d , and R 7e are hydrogen.

[0159] In one embodiment of a compound of Formula (XVI), R 5a and R 5b are each independently (a) halo; (b) Ci- 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 cycloalkyl, Ce-i4 aryl, C7-15 aralkyl, heteroaryl, or heterocyclyl; or (c) -C(0)R la , -C(0)0R la , -C(0)NR lb R lc , -C(NR la )NR lb R lc , -OR la , -0C(0)R la , -

R 7d , R 7e , R la , R lb , R lc , and R ld are defined herein elsewhere.

[0160] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or -OR la , where R la is Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one, two, three, four, or five substituents Q;

R 5a and R 5b are each independently hydrogen or Ci- 6 alkyl optionally substituted with one, two, three, four, or five substituents Q;

R 7a is C 14 aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CR X , with the proviso that at least two of X, Y, and Z are N; where R x is a hydrogen or Ci- 6 alkyl, optionally substituted with one, two, three, or four substituents Q a .

[0161] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is Ci- 6 alkyl, optionally substituted with one or more halo; R 5a and R 5b are each independently hydrogen or Ci-6 alkyl;

R 7a is Ce-i4 aryl, heteroaryl, or heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0162] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are each independently hydrogen or Ci-6 alkyl;

R 7a is C 14 aryl, monocyclic heteroaryl, or monocyclic heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0163] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are each independently hydrogen or Ci-6 alkyl;

R 7a is phenyl, 5- or 6-membered heteroaryl, or 5- or 6-membered heterocyclyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0164] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or methoxy;

R 2 is hydrogen;

R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are each independently hydrogen or Ci-6 alkyl;

R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, piperidinyl, or piperazinyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0165] In one embodiment of any of the formulae provided herein:

R 1 is hydrogen or methoxy;

R 2 is hydrogen; R 3 and R 4 are hydrogen;

R 6 is difluoromethyl;

R 5a and R 5b are each independently hydrogen or Ci- 6 alkyl;

R 7a is phenyl, imidazolyl, pyrozolyl, pyridinyl, piperidinyl, or piperazinyl, each of which is optionally substituted with one, two, three, or four substituents Q a ;

R 7b , R 7c , R 7d , and R 7e are hydrogen; and

X, Y, and Z are each independently N or CH.

[0166] In one embodiment of any of the formulae provided herein, R 1 is hydrogen. In one embodiment of any of the formulae provided herein, R 1 is -OR la . In one embodiment of any of the formulae provided herein, R 1 is -O-Ci- 6 alkyl. In one embodiment of any of the formulae provided herein, R 1 is methoxy.

[0167] In one embodiment of any of the formulae provided herein, R 2 is hydrogen. In one embodiment of any of the formulae provided herein, R 2 is -NR lb R lc . In one embodiment of any of the formulae provided herein, R 2 is amino.

[0168] In one embodiment of any of the formulae provided herein, R 3 is hydrogen.

[0169] In one embodiment of any of the formulae provided herein, R 4 is hydrogen.

[0170] In one embodiment of any of the formulae provided herein, R 6 is Ci- 6 alkyl, optionally substituted with one or more substituents Q.

[0171] In one embodiment of any of the formulae provided herein, R 6 is methyl, fluoromethyl, difluoromethyl, or trifluoromethyl. In one embodiment of any of the formulae provided herein, R 6 is difluoromethyl.

[0172] The groups or variables, R 1 , R 2 , R 3 , R 4 , R 6 , R 5a , R 5b , R 5c , R 5d , R 5e , R 5f , R 5g , R 7a , R 7b , R 7c , R 7d , R 7e , m, n, X, Y, and Z in Formulae provided herein, e.g., Formulae (I), (II), (VII), (IX), (X), (XI), (XVI), are further defined in the embodiments described herein. All combinations of the embodiments provided herein for such groups and/or variables are within the scope of this disclosure.

[0173] In certain embodiments, m is 0. In certain embodiments, m is 1.

[0174] In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2. In certain embodiments, n is 3. In certain embodiments, n is 4. In certain embodiments, n is 0, 1, or 2. In certain embodiments, n is 0, 1, 2, or 3. In certain embodiments, n is 1, 2, or 3. In certain embodiments, n is 1 or 2.

[0175] In certain embodiments, m is 0, and n is 0, 1, 2, or 3. In certain embodiments, m is 0, n is 0, 1, or 2. In certain embodiments, m is 0, n is 0 or 1. In certain embodiments, m is 0, n is 0. In certain embodiments, m is 0 and n is 1. In certain embodiments, m is 1, n is 0, 1, 2, or 3. In certain embodiments, m is 1, n is 0, 1, or 2. In certain embodiments, m is 1, n is 0 or 1. In certain embodiments, m is 1, n is 0. In certain embodiments, m is 1, n is 1.

[0176] In specific embodiments, m is 0, n is 1, and R 5a and R 5b are each methyl.

[0177] In certain embodiments, X is N. In certain embodiments, X is CR X , wherein R x is as defined herein. In certain embodiments, X is CH. [0178] In certain embodiments, Y is N. In certain embodiments, Y is CR X , wherein R x is as defined herein. In certain embodiments, Y is CH.

[0179] In certain embodiments, Z is N. In certain embodiments, Z is CR X , wherein R x is as defined herein. In certain embodiments, Z is CH.

[0180] In certain embodiments, X, Y, and Z are N. In certain embodiments, X and Y are N, and Z is CH. In certain embodiments, X and Z are N, and Y is CH. In certain embodiments, Y and Z are N, and X is CH.

[0181] In certain embodiments, the compound provided herein is not 4-(2-(difluoromethyl)- l//- benzo[r/]imidazol-l-yl)-6-morpliolino-/V-(2 -phenyl -2-(pyrrolidin-l-yl)ethyl)-l, 3, 5-triazin-2 -amine. In certain embodiments, the compound provided herein is not 6-(2-(difluoromethyl)- l//-benzo|<:/|imidazol- 1 - yl)-/V-(l-(4-((R)-3-(methoxymethyl)morpholino)phenyl)ethyl)- 2-morpholinopyrimidin-4-amine.

[0182] In certain embodiments, when X, Y, and Z are N, and R 5a is hydrogen, R 5b is not heterocyclyl. In certain embodiments, when X, Y, and Z are N, and R 5a is hydrogen, R 5b is not 5-membered heterocyclyl. In certain embodiments, when X, Y, and Z are N, and R 5a is hydrogen, R 5b is not pyrrolidinyl. In certain embodiments, when X, Y, and Z are N, and R 5a is hydrogen, R 5b is not pyrrolidin-l-yl.

[0183] In certain embodiments, when X and Z are N, Y is CH, and R 5a is hydrogen, R 5b is morpholino- substituted phenyl. In certain embodiments, when X and Z are N, Y is CH, and R 5a is hydrogen, R 5b is not 4-((R)-3 -(methoxymethyl)morpholino)phenyl .

[0184] In one embodiment, provided herein is a compound selected from:

A13 A14

A23 A24

A39

A40

A62 A63

A68 A70

or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0185] In one embodiment, the PI3K inhibitor is Compound A35, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A36, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A68, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A70, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A37, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A38, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A41, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A42, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A43, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A44, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A62, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A63, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A64, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A65, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A66, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof. In one embodiment, the PI3K inhibitor is Compound A67, or isotopic variants, pharmaceutically acceptable salts, solvates, hydrates, or prodrugs thereof.

[0186] Synthesis of compounds of any of the Formulae provided herein e.g., Formulae (I), (II), (VII), (IX), (X), (XI), (XVI), is described in US Patent No. 9,056,852 B2, which is incorporated by reference for such disclosure.

Bispecific antigen-binding molecules

[0187] Some embodiments provided herein describe pharmaceutical compositions or methods for using the pharmaceutical compositions comprising a PI3K inhibitor described herein in combination with a bispecific antibody or a bispecific antigen -binding molecule.

[0188] Some embodiments provided herein describe pharmaceutical compositions or methods for using the pharmaceutical compositions comprising a PI3K inhibitor described herein in combination with a T- cell activating bispecific antigen -binding molecule.

[0189] Some embodiments provided herein describe pharmaceutical compositions or methods for using the pharmaceutical compositions comprising a PI3K inhibitor described herein in combination with a bispecific antigen-binding molecule or bispecific antibody including, but not limited to, a bispecific antigen-binding molecule or bispecific antibody that is a bispecific T-cell engager or bispecific antibody that disrupts tumor cell proliferation. In some embodiments, the bispecific antibodies disclosed herein disrupt tumor cell proliferation by bispecific antibody -dependent T-cell mediated lysis, cell-mediated cytotoxicity, and/or phagocytosis of tumor cells. In some embodiments, the bispecific antibodies disclosed herein disrupt tumor cell proliferation by, e.g., selectively blocking CD47 on tumor cells.

[0190] Any suitable T-cell activating bispecific antigen-binding molecule, or other bispecific antigen binding molecule (e.g., anti-CD47/anti-CD19 antibody), may be used in combination with a PI3K inhibitor described herein. In some embodiments, the T-cell activating bispecific antigen-binding molecule is obinutuzumab, or pharmaceutically acceptable salts thereof.

[0191] In some embodiments, the T-cell activating bispecific antigen-binding molecule isobinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, AMV564, AFM13, REGN-1979, mosunetuzumab, GEN- 3013, or pasotuxizumab. In some embodiments, the T-cell antigen is CD3.

[0192] In some embodiments, the T-cell activating bispecific antigen-binding molecule isobinutuzumab, mosunetuzumab, selicrelumab, blinatumomab, AMV564, AFM13, REGN-1979, mosunetuzumab, GEN- 3013, pasotuxizumab, catumaxomab, cemiplimab, RG-7802, MCLA-128, MGD-007, MGD-007, MCLA- 117, REGN-4018, AMG-330, XmAb-14045, ABL-001, MGD-013, XmAb-18087, ABBV-428, RO- 7121661, JNJ-9178, GBR-1302, PF-06863135, CC-93269, MEDI-5752, AMG-757, AMG-427, GEM- 333, MBS-301, PF-06671008, orlotamab, KN-046, ERY-974, JNJ-7957, navicixizumab, ZW-25, JNJ- 6372, flotetuzumab, BI-836880, RO-7082859, XmAb-20717, AMG-424, AMG-701, AMG-596, AK-104, AMG-673, RG-7992, RG-7992, GBR-1342, AMG-570, FS-118, BTRC-4017A, JNJ-7564, XmAb-13676, AMG-420, MCLA-158, A-337, KN-026, vanucizumab, AMG-562, UCB-0159, M-701, APVO-436, ATOR-1015, AK-112, MGD-019, A-319, EMB-01, M-802, XmAb-23104, MPE-831959, TB-535H, VB- 601, XmAb-14484, XmAb-19722, ND-007, El-3s, IMM-0306, IMM-03, AP-505, SIB-003, HMBD- 004A, HMBD-003, ABP-100, ABP-120, BH-2941, CB-307, FIT-012, BA-002, ABL-503, KY-1055, NIBX-2101, BH-2950, GBR-1372, COVA-4231, MT-6194, JNJ-0819, BH-2922, ABP-200, ABP-110, ABL-501, IBI-318, IBI-322, ALLI-0143, SBT-200, GNR-022, AFM-24I, FIT-1, GO-2, SMET-1, KY- 1049, EM-801, TRIO-201, JS-003, AFM-24T, ABL-301, AMG-160, GMA-104, AGEN-1423, AP-201, IBI-323, INBRX-105, BA-003, MPEV-201959, ALG.APV-527, TG-1801, PMC-201, CKD-702, JNJ- 9383, PMC-001, BS-027125, KN-052, VIS-FNG, PMC-002, IPH-61, Fsn-1006, XmAb-22841, AFM-26, KN-043, NI-1801, REGN-5458, BH-2954, A-329, SIB-001, KY-1043, VIS-RSV, TF-12, TMB-365, AMG-910, AGEN-1223, SMET-2, ATOR-1144, BMX-002, BMX-101, SL-634, TSR-075, ALX-0141, IMM-02, APVO-437, TNB-383B, GO-6, FS-120, FS-222, TACSYN, MCLA-145, Novotarg, IBI-319, IBI-322, PC-101, PT-217, RG6026, DSP-107(KAHR-107), or ABP-201.

[0193] In some embodiments, the T-cell activating bispecific antigen-binding molecule comprises a first Fab molecule which specifically binds to a first antigen; and a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen where the target is selected from one or more of: CD37, CD 123, PDL1, PD1, VEGFR1, CD98hc, HER-2, HER-3, FLT3, CD19, CD20, 5T4, CD27, CD279, cd73, CD47, CD276, CD340, CD4, GD2, TfR, CD40, 4-1BB, BCMA, ROR1, PSMA, CD137, LAG3, EGFR, HLA-DR, CD5, CD 16, HBsAg, HBV, CD3E, CD133, IL13RA2, IL-8, CD274, TNFRSF9, CLL-1, Gpl20, TNF, TAFI, PAI-1, CD33, CEACAM6, TNFRSF9, Ang2, EphA2, EpCAM, EGFRvIII, MUC-1, RG6026, 41BBL, mesothelin, and CXCL-12. In some embodiments of the T-cell activating bispecific antigen-binding molecule comprising a first Fab molecule which specifically binds to a first antigen; and a second Fab molecule, the second Fab molecule is capable of binding to more than one target antigen on targets selected from: CD37, CD123, PDL1, VEGFR1, CD98hc, HER-2, HER-3, FLT3, CD19, CD20, 5T4, CD27, CD47, CD276, CD340, CD4, GD2, TfR, CD40, 4-1BB, BCMA, ROR1, PSMA, CD137, LAG3, EGFR, HLA-DR, CD5, CD16, HBsAg, HBV, CD3E, CD133, IL13RA2, IL-8, CD274, TNFRSF9, CLL- 1, Gpl20, TNF, TAFI, PAI-1, CD33, CEACAM6, TNFRSF9, Ang2, EphA2, EpCAM, EGFRvIII, MUC- 1, RG6026, 41BBL, mesothelin, and CXCL-12.

[0194] Provided herein are monovalent antibodies and/or bispecific antibodies that include at least a first arm that is specific for CD47. Provided herein are bispecific antibodies that recognize CD47 and a second target. In some embodiments, provided herein are bispecific antibodies where one of the binding sites is specific for CD47 and the second binding site is specific for another target, for example a tumor- associated antigen (TAA). In some embodiments, the TAA is an antigen that is expressed on the cell surface of a cancer cell. In some embodiments, the TAA is CD 19. In some embodiments, the TAA is 41BBL. In some embodiments, the TAA is CD33. [0195] In an aspect, provided herein is a bispecific antibody that selectively blocks targets on tumor cells and elicits an anti-tumor immune response. In some embodiments of the bispecific antibody provided herein, the antibody comprises three arms. In some embodiments of the bispecific antibody provided herein, the antibody comprises a targeting arm, an effector arm, and an Fc arm. In some embodiments of the bispecific antibody provided herein, the antibody comprises a targeting arm that binds to CD 19 on tumor cells, an effector arm, and an Fc arm. hi some embodiments of the bispecific antibody provided herein, the antibody comprises a targeting arm that binds to CD 19 on tumor cells, an effector arm that has CD 47 blocking activity, and an Fc arm. hi some embodiments of the bispecific antibody provided herein, the antibody comprises a targeting arm that binds to CD 19 on tumor cells, an effector arm that has selective tumor cell CD47 blocking activity, and an Fc arm that serves as a bridge to recruit macrophages and other innate immune killer cells to the tumor cell.

[0196] In an aspect, provided herein is a trifunctional antibody (triAb) that can bind to three different cell types. In some embodiments, the trifunctional antibodies of the present disclosure are T-cell activating bispecific antibodies that can bind to tumor cells, T-cells, and accessory cells. In some embodiments, the T-cell activating bispecific antigen-binding molecule is a trifunctional antibody that comprises a first Fab molecule which specifically binds to a first antigen; a second Fab molecule which specifically binds to a second antigen, wherein the first antigen is an activating T-cell antigen and the second antigen is a target cell antigen; and an intact Fc region that can bind to cell surface receptors (Fc receptor) on accessory cells or other Fc receptor expressing cells. In some embodiments, accessory cells include but are not limited to monocytes/macrophages, natural killer cells, dendritic cells. Examples of trifiinctional antibodies include but are not limited to catumaxomab, ertumaxomab, FBTA05 (lymphomun), and TRBS07 (ektomab). In some embodiments of the T-cell activating bispecific antigen-binding molecules of the present disclosure, the first Fab molecule of the T-cell activating bispecific antigen-binding molecule binds to a first antigen, wherein the first antigen is an activating T-cell antigen. In some embodiments, the activating T-cell antigen is CD3. In some embodiments, the trifiinctional antibodies of the present disclosure elicit immune responses or cytotoxic T-cell responses from CD4 T-cells and/or CD8 T-cells.

Methods of Use

[0197] In certain embodiments, provided herein are methods for treating or preventing a disease, comprising administering an effective amount of a compound of Formula (I), or an isotopic variant thereof or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and an effective amount of a T-cell activating bispecific antigen -binding molecule.

[0198] Some embodiments provided herein describe a method for treating a patient with B cell maligancies, the method comprising administering an effective amount of a compound of Formula (I), or an isotopic variant thereof or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and an effective amount of a T-cell activating bispecific antigen -binding molecule. In some embodiments, the T-cell activating bispecific antigen -binding molecule is obinutuzumab.

[0199] In some embodiments, the methods described herein avoid and/or reduce adverse or unwanted side effects associated with the use of the PI3K inhibitor and/or the T-cell activating bispecific antigen- binding molecule. In some embodiments, the methods described herein avoid, reduce, or minimize the risk of death due to infections associated with PI3K inhibitor and/or the T-cell activating bispecific antigen-binding molecule. In some embodiments, the methods described herein avoid, reduce, or minimize infections, neutropenia, diarrhea/colitis, elevated liver transaminases (alanine

aminotransferase/aspartate aminotransferase > 5x upper limit of normal), pneumonitis, rash, hepatic impairment, renal impairment, pyrexia, or increased triglycerides, or a combination thereof in patients receiving the treatment described herein. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of infection. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of neutropenia. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of diarrhea/colitis. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of elevated liver transaminases. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of pneumonitis. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of a rash. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of hepatic impairment or renal impairment. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of pyrexia. In certain embodiments, the methods described herein avoid, reduce, or minimize the incidence of increased triglycerides. In certain embodiments, the methods described herein avoid, reduce, or minimize enterocolitis (manifested as diarrhea), cutaneous toxicities, liver toxicity (manifested as elevation of transaminases), pulmonary toxicity (manifested as non-infectious

pneumonitis), infections, or combinations thereof.

[0200] In some embodiments, the methods described herein provides a high objective response rate (ORR) as determined by tumor assessment from radiological tests and/or physical examination. In some embodiments, the methods described herein provide a durable response (DR) and/or increased durable response rate (DRR; a continuous response [complete or partial objective response] beginning within 12 months of treatment and lasting >6 months) in the subject or patient. In some embodiments, the methods described herein provide complete remission. In some embodiments, the methods described herein provide a better response compared to the monotherapy treatment of a compound of Formula (I) and/or the T-cell activating bispecific antigen-binding molecule. In some embodiments, the methods described herein provide complete remission beginning within 12 months of treatment and lasting >6 months. In some embodiments, the methods described herein provide a complete response (CR) and/or no evidence of disease (NED) beginning within 12 months of treatment and lasting >6 months.

[0201] In some embodiments of a method of treating follicular lymphoma (FL) including relapsed or refractory FL, the discontinuation rate due to adverse events is less than 25%, less than 20%, less than 15%, less than 10%, less than 8%, less than 5%. In some embodiments of a method of treating a T-cell malignancy including a relapsed or refractory T-cell malignancy, the discontinuation rate due to adverse events is less than 25%, less than 20%, less than 15%, less than 10%, less than 8%, less than 5%. In some embodiments of a method of treating a B-cell malignancy including a relapsed or refractory B-cell malignancy, the discontinuation rate due to adverse events is less than 25%, less than 20%, less than 15%, less than 10%, less than 8%, less than 5%.

[0202] The“discontinuation rate” is defined as the number of subjects who discontinue the study drugs prior to the study completion divided by the number of subjects treated.

[0203] In some embodiments, the discontinuation rate due to adverse events is less than 25%, less than 20%, less than 15%, less than 10%, less than 8%, less than 5%. In some embodiments, the discontinuation rate due to adverse events is less than 25%. In some embodiments, the discontinuation rate due to adverse events is less than 20%. In some embodiments, the discontinuation rate due to adverse events is less than 15%. In some embodiments, the discontinuation rate due to adverse events is less than 10%. In some embodiments, the discontinuation rate due to adverse events is less than 8%. In some embodiments, the discontinuation rate due to adverse events is about 4%.

[0204] In some embodiments, the discontinuation rate due to adverse events when the subjects are administered a compound of Formula (I), or an isotopic variant thereof or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug is less for subjects on an intermittent dosing schedule (IS) than the discontinuation rate observed for subjects on a continuous dosing schedule (CS).

[0205] In certain embodiments, provided herein are methods for treating or preventing a disease, comprising administering an effective amount of: a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule to a subject in need thereof. In some embodiments, the compound of Formula (I) is Compound A35 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A36 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A68 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A70 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A37 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A38 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A41 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A42 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A43 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A44 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A62 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A63 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A64 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A65 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A66 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A67 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (II) is Compound XVII or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0206] In one embodiment, provided herein are methods for treating or preventing a proliferative disease, comprising administering a compound of Formula (I), or an isotopic variant thereof; or a

pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and an effective amount of a T-cell activating bispecific antigen-binding molecule to a subject in need thereof. In one embodiment, the T-cell activating bispecific antigen-binding molecule is obinutuzumab.

[0207] In certain embodiments, provided herein are methods for treating or preventing a proliferative disease, comprising administering an effective amount of: a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule to a subject in need thereof. In some embodiments, the compound of Formula (I) is Compound A35 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A36 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A68 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A70 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A37 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A38 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A41 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A42 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A43 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A44 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A62 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A63 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A64 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A65 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A66 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A67 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (II) is Compound XVII or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0208] In one embodiment, provided herein are methods for treating or preventing cancer, comprising administering a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and an effective amount of a T-cell activating bispecific antigen binding molecule to a subject in need thereof. In one embodiment, the T-cell activating bispecific antigen binding molecule is obinutuzumab.

[0209] In one embodiment, provided herein are methods for treating or preventing cancer, comprising administering an effective amount of: a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule to a subject in need thereof. In some embodiments, the compound of Formula (I) is Compound A35. In some embodiments, the compound of Formula (I) is Compound A36. In some embodiments, the compound of Formula (I) is Compound A68. In some embodiments, the compound of Formula (I) is Compound A70. In some embodiments, the compound of Formula (I) is Compound A37. In some embodiments, the compound of Formula (I) is Compound A38. In some embodiments, the compound of Formula (I) is Compound A41. In some embodiments, the compound of Formula (I) is Compound A42. In some embodiments, the compound of Formula (I) is Compound A43. In some embodiments, the compound of Formula (I) is Compound A44. In some embodiments, the compound of Formula (I) is Compound A62. In some embodiments, the compound of Formula (I) is Compound A63. In some embodiments, the compound of Formula (I) is Compound A64. In some embodiments, the compound of Formula (I) is Compound A65. In some embodiments, the compound of Formula (I) is Compound A66. In some embodiments, the compound of Formula (I) is Compound A67.

[0210] In certain embodiments, the proliferative disease or the cancer is a hematological cancer or malignancy.

[0211] In certain embodiments, the proliferative disease or the cancer is a cancer of the breast, skin, prostate, cervix, uterus, ovary, testes, bladder, lung, liver, larynx, oral cavity, colon and gastrointestinal tract (e.g., esophagus, stomach, pancreas), brain, thyroid, blood, and lymphatic system,

[0212] In certain embodiments, the cancers treatable with the methods provided herein include, but are not limited to, (1) leukemias, including, but not limited to, acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemias such as myeloblastic, promyelocytic, myelomonocytic, monocytic, erythroleukemia leukemias and myelodysplastic syndrome or a symptom thereof (such as anemia, thrombocytopenia, neutropenia, bicytopenia or pancytopenia), refractory anemia (RA), RA with ringed sideroblasts (RARS), RA with excess blasts (RAEB), RAEB in transformation (RAEB-T), preleukemia, and chronic myelomonocytic leukemia (CMML), (2) chronic leukemias, including, but not limited to, chronic myelocytic (granulocytic) leukemia, chronic lymphocytic leukemia, and hairy cell leukemia; (3) polycythemia vera; (4) lymphomas, including, but not limited to, follicular lymphoma (FL), Hodgkin’s disease and non-Hodgkin’s disease; (5) multiple myelomas, including, but not limited to, smoldering multiple myeloma, nonsecretory myeloma, osteosclerotic myeloma, plasma cell leukemia, solitary plasmacytoma, and extramedullary plasmacytoma; (6) Waldenstrom’s macroglobulinemia; (7) monoclonal gammopathy of undetermined significance; (8) benign monoclonal gammopathy; (9) heavy chain disease; (10) bone and connective tissue sarcomas, including, but not limited to, bone sarcoma, osteosarcoma, chondrosarcoma, Ewing’s sarcoma, malignant giant cell tumor, fibrosarcoma of bone, chordoma, periosteal sarcoma, soft-tissue sarcomas, angiosarcoma (hemangiosarcoma), fibrosarcoma, Kaposi’s sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, metastatic cancers,

neurilemmoma, rhabdomyosarcoma, and synovial sarcoma; (11) brain tumors, including, but not limited to, glioma, astrocytoma, brain stem glioma, ependymoma, aligodendrogboma, nonglial tumor, acoustic neurinoma, craniopharyngioma, medulloblastoma, meningioma, pineocytoma, pineoblastoma, and primary brain lymphoma; (12) breast cancer, including, but not limited to, adenocarcinoma, lobular (small cell) carcinoma, intraductal carcinoma, medullary breast cancer, mutinous breast cancer, tubular breast cancer, papillary breast cancer, primary cancers, Paget’s disease, and inflammatory breast cancer; (13) adrenal cancer, including, but not limited to, pheochromocytom and adrenocortical carcinoma; (14) thyroid cancer, including, but not limited to, papillary or follicular thyroid cancer, medullary thyroid cancer, and anaplastic thyroid cancer; (15) pancreatic cancer, including, but not limited to, insulinoma, gastrinoma, glucagonoma, vipoma, somatostatin-secreting tumor, and carcinoid or islet cell tumor; (16) pituitary cancer, including, but limited to, Cushing’s disease, prol actin-secreting tumor, acromegaly, and diabetes insipius; (17) eye cancer, including, but not limited, to ocular melanoma such as iris melanoma, choroidal melanoma, and cilbary body melanoma, and retinoblastoma; (18) vaginal cancer, including, but not limited to, squamous cell carcinoma, adenocarcinoma, and melanoma; (19) vulvar cancer, including, but not limited to, squamous cell carcinoma, melanoma, adenocarcinoma, basal cell carcinoma, sarcoma, and Paget’s disease; (20) cervical cancers, including, but not limited to, squamous cell carcinoma, and adenocarcinoma; (21) uterine cancer, including, but not limited to, endometrial carcinoma and uterine sarcoma; (22) ovarian cancer, including, but not limited to, ovarian epithelial carcinoma, borderline tumor, germ cell tumor, and stromal tumor; (23) esophageal cancer, including, but not limited to, squamous cancer, adenocarcinoma, adenoid cystic carcinoma, mucoepidermoid carcinoma,

adenosquamous carcinoma, sarcoma, melanoma, plasmacytoma, verrucous carcinoma, and oat cell (small cell) carcinoma; (24) stomach cancer, including, but not limited to, adenocarcinoma, fungating (polypoid), ulcerating, superficial spreading, diffusely spreading, malignant lymphoma, liposarcoma, fibrosarcoma, and carcinosarcoma; (25) colon cancer; (26) rectal cancer; (27) liver cancer, including, but not limited to, hepatocellular carcinoma and hepatoblastoma; (28) gallbladder cancer, including, but not limited to, adenocarcinoma; (29) cholangiocarcinomas, including, but not limited to, pappillary, nodular, and diffuse; (30) lung cancer, including, but not limited to, non -small cell lung cancer, squamous cell carcinoma (epidermoid carcinoma), adenocarcinoma, large-cell carcinoma, and small-cell lung cancer; (31) testicular cancer, including, but not limited to, germinal tumor, seminoma, anaplastic, classic (typical), spermatocytic, nonseminoma, embryonal carcinoma, teratoma carcinoma, and choriocarcinoma (yolk-sac tumor); (32) prostate cancer, including, but not limited to, adenocarcinoma, leiomyosarcoma, and rhabdomyosarcoma; (33) penal cancer; (34) oral cancer, including, but not limited to, squamous cell carcinoma; (35) basal cancer; (36) salivary gland cancer, including, but not limited to, adenocarcinoma, mucoepidermoid carcinoma, and adenoidcystic carcinoma; (37) pharynx cancer, including, but not limited to, squamous cell cancer and verrucous; (38) skin cancer, including, but not limited to, basal cell carcinoma, squamous cell carcinoma and melanoma, superficial spreading melanoma, nodular melanoma, lentigo malignant melanoma, and acral lentiginous melanoma; (39) kidney cancer, including, but not limited to, renal cell cancer, adenocarcinoma, hypernephroma, fibrosarcoma, and transitional cell cancer (renal pelvis and/or uterer); (40) Wilms’ tumor; (41) bladder cancer, including, but not limited to, transitional cell carcinoma, squamous cell cancer, adenocarcinoma, and carcinosarcoma; and other cancer, including, not limited to, myxosarcoma, osteogenic sarcoma, endotheliosarcoma, lymphangio- endotheliosarcoma, mesothelioma, synovioma, hemangioblastoma, epithelial carcinoma,

cystadenocarcinoma, bronchogenic carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, and papillary adenocarcinomas (See Fishman el ctl.. 1985. Medicine. 2d Ed., J.B. Lippincott Co., Philadelphia and Murphy et al , 1997, Informed Decisions: The Complete Book of Cancer Diagnosis, Treatment, and Recovery, Viking Penguin, Penguin Books U.S.A., Inc., United States of America).

[0213] In certain embodiments, provided herein are methods of treating a hematological malignancy with a combination of an effective amount of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and an effective amount of a T-cell activating bispecific antigen-binding molecule in a patient. In one embodiment, the T-cell activating bispecific antigen-binding molecule is obinutuzumab.

[0214] In certain embodiments, provided herein are methods of treating a hematological malignancy with a combination of an effective amount of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule to a subject in need thereof. In certain embodiments, the hematological malignancy is a leukemia, a lymphoma, a myeloma, a non-Hodgkin’s lymphoma, a Hodgkin’s lymphoma, T-cell malignancy, or a B-cell malignancy. In some embodiments, the hematological malignancy is chronic lymphocytic leukemia, follicular lymphoma, diffuse large B-cell lymphoma, or non-Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is chronic lymphocytic leukemia or non- Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is chronic lymphocytic leukemia. In other embodiments, the hematological malignancy is non -Hodgkin’s lymphoma. In some embodiments, the hematological malignancy is follicular lymphoma. In other embodiments, the hematological malignancy is diffuse large B-cell lymphoma. In some embodiments, the compound of Formula (I) is Compound A35 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A36 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A68 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A70 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A37 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A38 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A41 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A42 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A43 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A44 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A62 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A63 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A64 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A65 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A66 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A67 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0215] In certain embodiments, the hematological malignancy is a T-cell malignancy. In certain embodiments, T-cell malignancies include peripheral T-cell lymphoma not otherwise specified (PTCL- NOS), anaplastic large cell lymphoma, angioimmunoblastic lymphoma, cutaneous T-cell lymphoma, adult T-cell leukemia/lymphoma (ATLL), blastic NK-cell lymphoma, enteropathy-type T-cell lymphoma, hematosplenic gamma-delta T-cell lymphoma, lymphoblastic lymphoma, nasal NK/T-cell lymphomas, or treatment-related T-cell lymphomas.

[0216] In certain embodiments, the hematological malignancy is a B-cell malignancy. In certain embodiments, B-cell malignancies include acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), acute monocytic leukemia (AMoL), chronic lymphocytic leukemia (CLL), high-risk chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), high-risk small lymphocytic lymphoma (SLL), follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Waldenstrom’s macroglobulinemia, multiple myeloma, extranodal marginal zone B cell lymphoma, nodal marginal zone B cell lymphoma, Burkitt’s lymphoma, non-Burkitt high grade B cell lymphoma, primary mediastinal B-cell lymphoma (PMBL), immunoblastic large cell lymphoma, precursor B -lymphoblastic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic marginal zone lymphoma, plasma cell myeloma, plasmacytoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, or lymphomatoid granulomatosis. In certain embodiments, the B-cell malignancy is diffuse large B-cell lymphoma (DLBCL). In certain embodiments, the hematological malignancy is diffuse large B-cell lymphoma (DLBCL). In certain embodiments, the DLBCL is an activated B-cell DLBCL (ABC-DLBCL), a germinal center B-cell like DLBCL (GBC-DLBCL), a double hit DLBCL (DH-DLBCL), or a triple hit DLBCL (TH-DLBCL). In certain embodiments, the

hematological malignancy is relapsed-refractory diffuse large B-cell lymphoma (r/r DLBCL). In some embodiments, the B cell malignancy is B-cell non-Hodgkin’s lymphoma (NHL). In certain embodiments, the B-cell malignancy is selected from chronic lymphocytic leukemia (CLL), small lymphocytic lymphoma (SLL), follicular lymphoma (FL), marginal zone B cell lymphoma (MZL), diffuse large B-cell lymphoma (DLBCL), and high-grade non-Hodgkin's lymphoma. In certain embodiments, the B-cell malignancy is selected from chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), marginal zone B cell lymphoma (MZL), or diffuse large B-cell lymphoma (DLBCL). In certain embodiments, the B cell malignancy is a relapsed or refractory B cell malignancy. In certain embodiments of the methods provided herein, the FL is a relapsed or refractory FL (R/R FL).

[0217] In certain embodiments, the hematological malignancy is a relapsed or refractory hematological malignancy. In certain embodiments, the relapsed or refractory hematological malignancy is a relapsed or refractory T-cell malignancy. In certain embodiments, the relapsed or refractory hematological malignancy is a relapsed or refractory B-cell malignancy.

[0218] Some embodiments provided herein describe a method for treating or preventing a proliferative disease or disorder comprising administering a PI3K inhibitor in combination with a T-cell activating bispecific antigen-binding molecule. In some embodiments, the combination therapy of a PI3K inhibitor described herein (e.g., a compound of Formula (I)) and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) provides a synergistic effect. In some embodiments, the combination therapy of a PI3K inhibitor described herein (e.g., a compound of Formula (I)) and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) provides a synergistic antitumor or anti -cancer activity. In certain embodiments, the combination therapy described herein permits the use of lower dosages of the PI3K inhibitor and/or the T-cell activating bispecific antigen-binding molecule. In some embodiments, the combination therapy described herein permits less frequent administration of the PI3K inhibitor and/or the T-cell activating bispecific antigen-binding molecule to a subject. In some embodiments, the combination therapy described herein reduces the toxicity associated with the administration of the PI3K inhibitor and/or the T-cell activating bispecific antigen-binding molecule to a subject without reducing the efficacy in the prevention, management, treatment, or amelioration of cancer, such as chronic lymphocytic leukemia. In some embodiments, the synergistic effect observed with the combination therapy described herein results in improved efficacy of therapies in the prevention, management, treatment, or amelioration of cancer, such as chronic lymphocytic leukemia. [0219] In some embodiments, the combination therapy described herein avoids or reduces adverse or unwanted side effects associated with the use of the PI3K inhibitor and/or the T-cell activating bispecific antigen-binding molecule. In some embodiments, the combination therapy described herein avoids, reduces, or minimizes infections, neutropenia, diarrhea, pneumonia, anemia, thrombocytopenia, nausea, vomiting, swelling in extremities, or a combination thereof in patients receiving the combination therapy. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of infection. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of neutropenia. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of diarrhea. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of pneumonia. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of anemia. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of thrombocytopenia. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of nausea. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of vomiting. In certain embodiments, the combination therapy described herein avoids, reduces, or minimizes the incidence of swelling in the extremities.

[0220] Depending on the disorder, disease, or condition to be treated, and the subject’s condition, the compounds or pharmaceutical compositions provided herein can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration and can be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants, and vehicles appropriate for each route of administration as described elsewhere herein.

DOSAGES AND DOSING REGIMENS

[0221] Depending on the disorder, disease, or condition to be treated, and the subject’s condition, the compounds or pharmaceutical compositions provided herein can be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration and can be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants, and vehicles appropriate for each route of administration as described elsewhere herein.

[0222] In certain embodiments, the methods provided herein comprise administering a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule to a patient simultaneously or sequentially by the same or different routes of administration. [0223] The suitability of a particular route of administration employed for a particular active agent will depend on the active agent itself (e.g., whether it can be administered orally without decomposing prior to entering the blood stream) and the disease being treated.

[0224] In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule is administered simultaneously, at essentially the same time, or sequentially. If administration takes place sequentially, the T-cell activating bispecific antigen-binding molecule may be administered before or after administration of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T- cell activating bispecific antigen-binding molecule is administered before administration of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T-cell activating bispecific antigen-binding molecule is administered simultaneously with administration of a compound of Formula (I), an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T-cell activating bispecific antigen-binding molecule is administered after the administration of a compound of Formula (I), an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0225] A compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule need not be administered by means of the same vehicle. In some embodiments, the T-cell activating bispecific antigen-binding molecule and a compound of Formula (I), or an isotopic variant thereof; or a

pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof are administered in different vehicles. The T-cell activating bispecific antigen-binding molecule may be administered one or more times, and the number of administrations of each component of the combination may be the same or different. In addition, a compound of Formula (I), or an isotopic variant thereof, or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule need not be administered at the same site.

[0226] In some instances, the methods described herein further comprise administering the PI3K inhibitor in combination with T-cell activating bispecific antigen -binding molecule to the subject or patient in need thereof in multiple cycles repeated on a regular schedule with periods of rest in between each cycle. For example, in some instances, treatment is given for one week followed by three weeks of rest is one treatment cycle.

[0227] In some instances, a cycle comprises administration of the PI3K inhibitor at the same time as administration of the T-cell activating bispecific antigen -binding molecule. In some instances, the PI3K inhibitor and the T-cell activating bispecific antigen-binding molecule are administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 21 days, about 22 days, about 23 days, about 24 days, about 25 days, about 26 days, about 27 days, or about 28 days.

[0228] In some instances, a cycle comprises administration of the PI3K inhibitor first followed by administration of the T-cell activating bispecific antigen-binding molecule second. In some instances, the PI3K inhibitor is administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days followed by administration of the T-cell activating bispecific antigen-binding molecule for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days.

[0229] In some instances, a cycle comprises administration of the PI3K inhibitor first followed by concurrent administration of the T-cell activating bispecific antigen -binding molecule. In some instances, the PI3K inhibitor is first administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days followed by the concurrent administration of the T-cell activating bispecific antigen-binding molecule for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days. In some instances, the PI3K inhibitor is first administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days followed by the concurrent administration of the T-cell activating bispecific antigen-binding molecule for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days. In some instances, the PI3K inhibitor is first administered for about 7 days followed by the concurrent administration of the T- cell activating bispecific antigen-binding molecule for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days. In some instances, the PI3K inhibitor is first administered for about 7 days followed by the concurrent administration of the T-cell activating bispecific antigen binding molecule for about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days.

[0230] In some instances, a cycle comprises administration of the PI3K inhibitor only. In some instances, the PI3K inhibitor is administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 21 days, about 22 days, about 23 days, about 24 days, about 25 days, about 26 days, about 27 days, or about 28 days.

[0231] In some instances, a cycle comprises administration of the the T-cell activating bispecific antigen binding molecule only. In some instances, the T-cell activating bispecific antigen-binding molecule is administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 21 days, about 22 days, about 23 days, about 24 days, about 25 days, about 26 days, about 27 days, or about 28 days.

[0232] In some instances, the method for multiple cycle chemotherapy comprises the administration of a second cycle within about 60 days or about 3 months. In some instances, the method for multiple cycle chemotherapy comprises the administration of a second cycle within 50 days. In another instance, the second cycle is administered within 45, 40, 35, 30, 28, 25, 21, 20, 15, 14, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 day(s) of the first cycle. In some embodiments, the administration of any additional cycles is within 50 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 10 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 9 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 8 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 7 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 6 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 5 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 4 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 3 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 2 days of the previous cycle. In some embodiments, the administration of any additional cycles is within 1 day of the previous cycle. In another embodiment, the additional cycle is administered within 45, 40, 35, 30, 28, 25, 21, 20, 15, 14, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 days of the previous cycle.

[0233] The length of a treatment cycle depends on the treatment being given. In some embodiments, the length of a treatment cycle ranges from two to six weeks. In some embodiments, the length of a treatment cycle ranges from four to six weeks. In some embodiments, the length of a treatment cycle is 28 days. In some embodiments, the length of a treatment cycle is 56 days. In some embodiments, a treatment cycle lasts one, two, three, or four weeks. In some embodiments, a treatment cycle lasts four weeks. The number of treatment doses scheduled within each cycle also varies depending on the drugs being given.

[0234] In certain instances, the compound of Formula (I), or an isotopic variant thereof; or a

pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on a 28-day cycle. In some embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least one 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a

pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least two 28-day cycles.

[0235] In certain embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for a period of up to about 7 days. In some embodiments, the days over which the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof are intermittent. In some embodiments, administering to subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof for about 7 consecutive days in a 28-day cycle.

[0236] In some embodiments, the method comprises an intermittent dosing schedule (IS), comprising administering to subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for 7 consecutive days followed by 21 days without treatment in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least one 28-day cycle. In some embodiments, the IS avoids or reduces adverse or unwanted side effects associated with the use of the PI3K inhibitor, such as enterocolitis (manifested as diarrhea), cutaneous toxicities, liver toxicity (manifested as elevation of transaminases), pulmonary toxicity (manifested as non-infectious pneumonitis), and infections. In some embodiments, the IS avoids or reduces enterocolitis, rash, transaminitis, or combinations thereof.

[0237] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for 28 consecutive days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on continuous dosing schedule (CS). In some embodiments, the continuous dosing schedule (CS), comprises once daily administration of compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof to the subject for 28 consecutive days in a 28-day cycle. In some

embodiments, the continuous dosing schedule (CS), comprises once daily administration of compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof to the subject for 28 consecutive days in a 28-day cycle until disease progression or intolerable toxicity occur. In some instances, patients on CS report of delayed onset of cases of enterocolitis and rash.

[0238] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of up to about 7 days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of up to about 7 intermittent days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of up to about 7 consecutive days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of up to about 7 consecutive days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of 7 consecutive days in a 28-day cycle. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS). In some embodiments, the intermittent dosing schedule (IS), comprises once daily administration of compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof to the subject for 7 consecutive days followed by 21 days without treatment in a 28-day cycle.

[0239] In some embodiments of the methods provided herein, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least three 28-day cycles, wherein: the first two 28-day cycles comprise a continuous daily dosing schedule (CS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, once daily for two 28- day cycles; and the third28-day cyclecomprises an intermittent dosing schedule (IS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, once daily for the first 7 consecutive days of the 28 -day cycle.

In some embodiments of the methods provided herein, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for at least three cycles, wherein: the first two cycles comprise a continuous daily dosing schedule (CS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for two cycles; and the subsequent cycle(s) comprises an intermittent dosing schedule (IS), comprising administering to subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for only the first 7 consecutive days in each subsequent cycle. In some embodiments of the methods provided herein, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject for four or more 28-day cycles, wherein: the first two or three 28-day cycles comprise a continuous daily dosing schedule (CS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, once daily for three or more 28-day cycles; and the subsequent 28-day cycle(s) comprise(s) an intermittent dosing schedule (IS), comprising administering to the subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, once daily for the first 7 consecutive days of the 28-day cycle.

[0240] In certain instances, CS refers to continuous daily dosing to a subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily on a 28-day schedule with no switch to IS. In certain instances, CS refers to continuous daily dosing to a subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily on a 28-day schedule for four or more cycles followed by a switch to IS (i.e., late switch to IS). In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a

pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a subject on an intermittent dosing schedule (IS) until progression of disease. In some embodiments, upon progression of disease, the subject resumes continuous daily dosing (CS) of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0241] In certain instances of the treatment regimen comprising administration of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof for two cycles of continuous daily administration (CS) followed by daily administration for only the first seven days of each subsequent cycle, the CS and IS cycles are 28-day cycles. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) to reduce or mitigate adverse side effects associated with PI3K5 inhibitors (e.g., enterocolitis, rash, and/or transaminitis). In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) resulting in mitigation or reduction of the incidence of immune -mediated toxicities by allowing recovery of TREG during treatment-free intervals.

[0242] In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) resulting in disease stabilization. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) resulting in disease regression. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) resulting in an objective rseponse. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) until disease stabilization is no longer observed. In some embodiments, the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject on an intermittent dosing schedule (IS) until disease progression is observed.

[0243] In certain instances of the treatment regimen comprising administration of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof for two cycles of continuous daily administration (CS) followed by daily administration for only the first seven days of each subsequent (IS) cycle, the CS and IS cycles are 28-day cycles, wherein the IS cycle is repeated until disease regression is no longer observed. In some or additional embodiments, if disease progression is observed in the subject, the subject resumes the 28-day cycles of continuous daily administration (CS) until disease regression or stabilization are observed

[0244] In certain instances of the treatment regimen comprising administration of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof for two 28-day cycles of continuous daily administration (CS) followed by daily administration for only the first seven days of each subsequent (IS) 28-day cycle; wherein disease regression or stabilization is no longer observed in the subject on the intermittent dosing schedule (IS) cycle, the subject resumes 28-day cycles of continuous daily administration (CS) until disease regression or stabilization are observed.

[0245] In some embodiments, about 60 mg of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject once daily for a period of 7 consecutive days in a 28-day cycle followed by 21 days without therapy, with cycles repeated every 28 days.

[0246] In some embodiments, administration of about 60 mg of the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to a subject in need thereof once daily for a period of 7 consecutive days followed by 21 days without treatment in a 28 -day cycle results in steady-state plasma concentrations sufficient to inhibit PI3K5 in target malignant B-cells. In further or additional embodiments, the subsequent 21 days without treatment is sufficient to repopulate TREG (i.e., 7 days to clear the compound of Formula (I) from the plasma (~7 half-lives) and 14 days for reconstitution of TREG after the compound of Formula (I) is cleared from the plasma.

[0247] In certain instances, the method comprises a continuous daily dosing schedule (CS) for at least two CS 28-day cycles, followed by an intermittant dosing schedule (IS), comprising administering to subject the compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof once daily for 7 consecutive days followed by 21 days without treatment in a 28-day cycle after the at least two CS 28-day cycles. In some embodiments, the dosing schedule avoids or reduces adverse or unwanted side effects associated with the use of the PI3K inhibitor, such as enterocolitis (manifested as diarrhea), cutaneous toxicities, liver toxicity (manifested as elevation of transaminases), pulmonary toxicity (manifested as non-infectious pneumonitis), and infections. In some embodiments, the dosing schedule avoids or reduces enterocolitis, rash, transaminitis, or combinations thereof.

[0248] In some instances, the method for the administration of multiple compounds comprises administering compounds within 48 hours or less of each other. In some embodiments, administration occurs within 24 hours, 12 hours, 6 hours, 3 hours, 1 hour, or 15 minutes. In some instances, the compounds are administered simultaneously. One example of simultaneous administration is the injection of one compound immediately before, after, or during the oral administration of the second compound, immediately referring to a time less than about 5 minutes.

[0249] In some instances, the method for the administration of multiple compounds occurs in a sequential order, wherein the PI3K inhibitor is administered before the T-cell activating bispecific antigen-binding molecule. In another instance, the T-cell activating bispecific antigen-binding molecule is administered before the PI3K inhibitor.

[0250] In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule is cyclically administered to a patient. As discussed above, cycling therapy involves the administration of an active agent or a combination of active agents for a period of time, followed by a rest for a period of time, and repeating this sequential administration. In some embodiments, cycling therapy reduces the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improves the efficacy of the treatment.

[0251] In some embodiments, the compound of Formula (I) is administered daily, every other day, every other day 3 times a week, every 2 weeks, every 3 weeks, every 4 weeks, every 5 weeks, every 3 days, every 4 days, every 5 days, every 6 days, weekly, bi-weekly, 3 times a week, 4 times a week, 5 times a week, 6 times a week, once a month, twice a month, 3 times a month, once every 2 months, once every 3 months, once every 4 months, once every 5 months, or once every 6 months. In some embodiments, the compound of Formula (I) is administered daily. In some embodiments, the compound of Formula (I) is administered daily for a period of up to about 28 days. In some embodiments, the compound of Formula (I) is administered daily for a period of up to about 7 days.

[0252] In some embodiments, the T-cell activating bispecific antigen -binding molecule is administered daily, every other day, every other day 3 times a week, every 3 days, every 4 days, every 5 days, every 6 days, weekly, every 8 days, every 9 days, every 10 days, every 11 days, every 12 days, every 13 days, every 2 weeks, every 15 days, every 3 weeks, every 4 weeks, every 5 weeks, bi-weekly, 3 times a week, 4 times a week, 5 times a week, 6 times a week, once a month, twice a month, 3 times a month, once every 2 months, once every 3 months, once every 4 months, once every 5 months, or once every 6 months.

[0253] In some embodiments, the T-cell activating bispecific antigen -binding molecule is administered in cycles. In some embodiments, the T-cell activating bispecific antigen-binding molecule is administered in 28-day cycles. In some embodiments, loading doses of the T-cell activating bispecific antigen-binding molecule are administered for one cycle (e.g., on Day 1, Day 8, and Day 15 or on Day 1, Day 2, Day 8, and Day 15) followed by administration of the T-cell activating bispecific antigen-binding molecule once monthly for 2-6 or 2-8 cycles (e.g., on Day 1). In some embodiments, treatment with the T-cell activating bispecific antigen-binding molecule is continued after 2-6 or 2-8 cycles every two months for up to two years. In some embodiments, the loading dose of the T-cell activating bispecific antigen-binding molecule is 1,000 mg. In other embodiments, the loading dose of the T-cell activating bispecific antigen-binding molecule is 900 mg. In other embodiments, the loading dose of the T-cell activating bispecific antigen binding molecule is 100 mg. In some embodiments, the loading dose of the T-cell activating bispecific antigen-binding molecule is 900 mg on Day 1, 100 mg on Day 2, and 1,000 mg on Day 15. In some embodiments, the loading dose of the T-cell activating bispecific antigen-binding molecule is 900 mg on Day 1, 100 mg on Day 2, and 1,000 mg on Day 15 of Cycle 1 followed by administration of 1,000 mg on Day 1 for Cycles 2-6. In some embodiments, a compound of Formula (I), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, is administered to the subject orally on Day 1 and Day 15 of Cycles 1-6 in combination therapy with the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab).

[0254] In some instances, the compound of Formula (I) or the T-cell activating bispecific antigen -binding molecule is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug holiday”). In some embodiments, the length of the drug holiday varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 12 days, 14 days, 15 days, 20 days, 21 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes from 10%-100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.

[0255] In certain embodiments, the methods provided herein comprise administering a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab), to a patient simultaneously or sequentially by the same or different routes of administration. In certain embodiments, the methods provided herein comprise administering a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule) (e.g., obinutuzumab) to a patient simultaneously or sequentially by the same or different routes of administration. In some embodiments, the compound of Formula (I) is Compound A35 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A36 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A68 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A70 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A37 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A38 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A41or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A42 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A43 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A44 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A62 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A63 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A64 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A65 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A66 or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the compound of Formula (I) is Compound A67or an isotopic variant, pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof.

[0256] The suitability of a particular route of administration employed for a particular active agent will depend on the active agent itself (e.g., whether it can be administered orally without decomposing prior to entering the blood stream) and the disease being treated. Recommended routes of administration for the second active agents are known to those of ordinary skill in the art. See, e.g., Physicians’ Desk Reference, 1755-1760 (56th ed., 2002).

[0257] In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) are administered simultaneously, at essentially the same time, or sequentially. In some embodiments, administration takes place sequentially and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered before or after administration of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered before administration of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered simultaneously with administration of a compound of Formula (I), an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered after the administration of a compound of Formula (I), an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. In some embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) need not be administered by means of the same vehicle. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) and a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof are administered in different vehicles. The T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) may be administered one or more times, and the number of administrations of each component of the combination may be the same or different. In addition, a compound of Formula (I), or an isotopic variant thereof, or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) need not be administered at the same site.

[0258] In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) are cyclically administered to a patient. Cycling therapy involves the administration of an active agent or a combination of active agents for a period of time, followed by a rest for a period of time, and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improves the efficacy of the treatment. In some embodiments, the cycles are 28 days.

[0259] In certain embodiments, in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof generally is ranging from about 1 to 1,000 mg, from about 1 to about 500 mg, from about 5 to about 500 mg, from about 5 to about 200 mg, from about 5 to about 250 mg, or from about 10 to about 150 mg, which can be administered in single or multiple doses. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50,

55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 450, 500, or 1,000 mg. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 60 mg, about 120 mg, about 150 mg, or about 180 mg. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 60 mg.

[0260] In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120,

125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325,

350, 375, 400, 450, 500, or 1,000 mg/day.

[0261] In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 45 mg/day. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 60 mg/day. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 90 mg/day. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 120 mg/day. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 150 mg/day. In certain embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered in an amount of about 180 mg/day.

[0262] For oral administration, the pharmaceutical compositions provided herein can be formulated in the form of tablets or capsules containing from about 1.0 to about 1,000 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, in one embodiment, about 1, about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 225, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 450, about 500, about 550, about 600, about 650, about 700, about 750, about 800, about 850, about 900, about 950, or about 1,000 mg of the a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof for the symptomatic adjustment of the dosage to the patient to be treated. The pharmaceutical compositions can be administered on a regimen of one (1) to four (4) times per day, including once, twice, three times, and four times per day. In some embodiments, the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered once per day. In some embodiments, about 30 mg, about 45 mg, or about 60 mg of the compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered once per day.

[0263] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 45 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 45 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 45 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 45 mg daily for 56 days.

[0264] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 60 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 60 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 60 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 60 mg daily for 56 days.

[0265] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 90 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 90 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 90 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 90 mg daily for 56 days.

[0266] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 120 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 120 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 120 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 120 mg daily for 56 days.

[0267] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 150 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 150 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 150 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 150 mg daily for 56 days. [0268] In some embodiments, the pharmaceutical compositions provided herein can be formulated in the form of tablets containing about 180 mg of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof. The pharmaceutical compositions can be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day. In certain embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 180 mg daily for 28 days or 56 days. In certain specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 180 mg daily for 28 days. In other specific embodiments, a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof is administered to a patient in need thereof in an amount of about 180 mg daily for 56 days.

[0269] In the methods of treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level of a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) generally is ranging from about 0.1 to 2,000 milligrams per day, per week, per month, or every two months. For example, 100, 900, or 1,000 milligrams once or multiple times per month may be effective to obtain the desired results.

[0270] In certain embodiments, the T-cell activating bispecific antigen -binding molecule is

obinutuzumab and the amount of obinutuzumab that is administered is from about 10 mg/day up to, and including, 2,000 mg/day. In certain embodiments, the amount of obinutuzumab that is administered is from about 10 mg/day to 1,000 mg/day. In certain embodiments, the amount of obinutuzumab that is administered is from about 100 mg/day to 1,000 mg/day. In certain embodiments, in the treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level of a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) generally is ranging from about 100 to 1,000 mg, from about 250 to about 1,000 mg, from about 500 to about 1,000 mg, from about 750 to about 1,000 mg, from about 900 to about 1,000 mg, or from about 10 to about 150 mg, which can be administered in single or multiple doses. In certain embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered in an amount of about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90,

95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 450, 500, 1,000, or 2,000 mg. In certain embodiments, the T- cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered in an amount of about 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120,

125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325,

350, 375, 400, 450, 500, 1,000, or 2,000 mg/day.

[0271] In the methods of treatment, prevention, or amelioration of one or more symptoms of the disorders, diseases, or conditions described herein, an appropriate dosage level of a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) generally is ranging from about 1 to 2,000 milligrams per day, per week, per month, or every two months. For example, 100, 900, or 1,000 milligrams once or multiple times per month may be effective to obtain the desired results.

[0272] For oral administration, the pharmaceutical compositions provided herein can be formulated in the form of tablets or capsules containing from about 1 to about 2,000 mg. For administrion by infusion, the pharmaceutical compositions provided herein can be formulated as a solution of 1,000 mg/40mL (25 mg/mL) of a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab). The

pharmaceutical compositions can be administered on a regimen of one (1) to four (4) times per month, including once, twice, three times, and four times per month. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered once per month. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered twice per month. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered three times per month. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered four times per month. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered once every other month. In some embodiments, the T-cell activating bispecific antigen binding molecule (e.g., obinutuzumab) is administered twice per day. In some embodiments, about 100 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion. In some embodiments, about 900 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion. In some embodiments, about 1,000 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion for up to six 28-day cycles. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion for at least six 28- day cycles. In some embodiments, about 100 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion on day one of the 28-day cycle. In some embodiments, about 900 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion on day two of the 28-day cycle. In some embodiments, about 1,000 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is

administered by infusion on days 8 and 15 of the 28-day cycle. In some embodiments, about 100 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion on day one Cycle 1, 900 mg on day two Cycle 1, 1,000 mg on day 8 and 15 of Cycle 1 and 1,000 mg on day one of Cycles 2-6 of the 28-day cycle. In some embodiments, about 100 mg of the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion on day 1, 8, and 15 of Cycle 1, and 1,000 mg on day one of Cycles 2-6 of the 28-day cycle. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 25 mg/hr over 4 hours. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 50 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 100 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 150 mg/hr. In some embodiments, the T-cell activating bispecific antigen binding molecule (e.g., obinutuzumab) is administered by infusion at 200 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 250 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 300 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 350 mg/hr. In some embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is administered by infusion at 400 mg/hr.

[0273] In certain embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is co-administered (e.g., in a single dosage form) with a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, once per day. In certain embodiments, the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) is co-administered (e.g., in a single dosage form) with a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof, twice per day.

[0274] It will be understood, however, that the specific dose level and frequency of dosage for any particular patient can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.

ADDITIONAL COMBINATION THERAPY

[0275] In certain embodiments, the methods of combination therapy comprising a compound of Formula (I) an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) can also be combined or used in combination with a third agent or therapies useful in the treatment, prevention, or amelioration of one or more symptoms of a proliferative disorders, diseases, or conditions.

[0276] Suitable third agent of therapies include, but are not limited to, (1) alpha-adrenergic agents; (2) antiarrhythmic agents; (3) anti-atherosclerotic agents, such as ACAT inhibitors; (4) antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; (5) anticancer agents and cytotoxic agents, e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes; (6) anticoagulants, such as acenocoumarol, argatroban, bivalirudin, lepirudin, fondaparinux, heparin, phenindione, warfarin, and ximelagatran, (7) anti -diabetic agents, such as biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins, meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, and glipizide), thiozolidinediones (e.g., troglitazone, rosiglitazone, and pioglitazone), and PPAR-gamma monists; (8) antifungal agents, such as amorolfme, amphotericin B, anidulafungin, bifonazole, butenafme, butoconazole, caspofungin, ciclopirox, clotrimazole, econazole, fenticonazole, fdipin, fluconazole, isoconazole, itraconazole, ketoconazole, micafungin, miconazole, naftifme, natamycin, nystatin, oxyconazole, ravuconazole, posaconazole, rimocidin, sertaconazole, sulconazole, terbinafme, terconazole, tioconazole, and voriconazole; (9) antiinflammatories, e.g., non-steroidal anti-inflammatory agents, such as aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoricoxib, faislamine, fenbufen, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lomoxicam, loxoprofen, lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide, oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinpyrazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin; (10) antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; (11) anti-platelet agents, such as GPIIb/IlIa blockers (e.g., abciximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), cilostazol, dipyridamole, and aspirin; (12) antiproliferatives, such as methotrexate, FK506 (tacrolimus), and mycophenolate mofetil; (13) anti-TNF antibodies or soluble TNF receptor, such as etanercept, rapamycin, and leflunimide; (14) aP2 inhibitors; (15) beta-adrenergic agents, such as carvedilol and metoprolol; (16) bile acid secjuestrants, such as questran; (17) calcium channel blockers, such as amlodipine besylate; (18) chemotherapeutic agents; (19) cyclooxygenase -2 (COX-2) inhibitors, such as celecoxib and rofecoxib; (20) cyclosporins; (21) cytotoxic drugs, such as azathioprine and cyclophosphamide; (22) diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzothiazide, ethacrynic acid, ticrynafen, chlorthalidone, furosenide, muzolimine, bumetanide, triamterene, amiloride, and spironolactone; (23) endothelin converting enzyme (ECE) inhibitors, such as phosphoramidon; (24) enzymes, such as L-asparaginase; (25) Factor Vila Inhibitors and Factor Xa Inhibitors; (26) fame syl -protein transferase inhibitors; (27) fibrates; (28) growth factor inhibitors, such as modulators of PDGF activity; (29) growth hormone secretagogues; (30) HMG CoA reductase inhibitors, such as pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, nisvastatin, or nisbastatin), and ZD-4522 (also known as rosuvastatin, atavastatin, or visastatin); neutral endopeptidase (NEP) inhibitors; (31) hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, and octreotide acetate; (32) immunosuppressants; (33) mineralcorticoidreceptor antagonists, such as spironolactone and eplerenone; (34) microtubule-disruptor agents, such as ecteinascidins; (35) microtubule -stabilizing agents, such as pacitaxel, docetaxel, and epothilones A-F; (36) MTP Inhibitors; (37) niacin; (38) phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, and vardenafil); (39) plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and taxanes; (40) platelet activating factor (PAF) antagonists; (41) platinum coordination complexes, such as cisplatin, satraplatin, and carboplatin; (42) potassium channel openers; (43) prenyl -protein transferase inhibitors; (44) protein tyrosine kinase inhibitors; (45) renin inhibitors; (46) squalene synthetase inhibitors; (47) steroids, such as aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone, hydrocortisone (cortisol), prednisolone, prednisone, methylprednisolone, dexamethasone, and triamcinolone; (48) TNF -alpha inhibitors, such as tenidap; (49) thrombin inhibitors, such as hirudin; (50) thrombolytic agents, such as anistreplase, reteplase, tenecteplase, tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase,

prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC); (51) thromboxane receptor antagonists, such as ifetroban; (52) topoisomerase inhibitors; (53) vasopeptidase inhibitors (dual NEP-ACE inhibitors), such as omapatrilat and gemopatrilat, and (54) other miscellaneous agents, such as, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, and gold compounds.

[0277] In certain embodiments, the third therapies that may be used in combination with the methods provided herein include, but are not limited to, surgery, endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, and agents to attenuate any adverse effects (e.g., antiemetics).

[0278] In certain embodiments, the third therapeutic agents that may be used in combination with the compounds provided herein include, but are not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, melphalan, and ifosfamide), antimetabolites (cytarabine (also known as cytosine arabinoside or Ara-C), and methotrexate), purine antagonists and pyrimidine antagonists (6- mercaptopurine, 5-fluorouracil, cytarbine, and gemcitabine), spindle poisons (vinblastine, vincristine, and vinorelbine), podophyllotoxins (etoposide, irinotecan, and topotecan), antibiotics (daunorubicin, doxorubicin, bleomycin, and mitomycin), nitrosoureas (carmustine and lomustine), enzymes

(asparaginase), and hormones (tamoxifen, leuprolide, flutamide, and megestrol), imatinib, adriamycin, dexamethasone, and cyclophosphamide. For a more comprehensive discussion of updated cancer therapies; See, the world wide web at nci.nih.gov/, a list of the FDA approved oncology drugs at the world wide web fda.gov/cder/cancer/dniglistframe.htm, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference for such disclosure.

[0279] In another embodiment, the methods provided herein comprise administration of a compound of Formula (I), or an isotopic variant thereof, or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab), together with administration of one or more chemotherapeutic agents and/or therapies selected from: alkylation agents (e.g., cisplatin, carboplatin); antimetabolites (e.g., methotrexate and 5-FU); antitumor antibiotics (e.g., adriamymycin and bleomycin); antitumor vegetable alkaloids (e.g., taxol and etoposide); antitumor hormones (e.g., dexamethasone and tamoxifen); antitumor immunological agents (e.g., interferon a, b, and g); radiation therapy; and surgery. In certain embodiments, the one or more chemotherapeutic agents and/or therapies are administered to the subject before, during, or after the administration of a compound of Formula (I), or an isotopic variant thereof, or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab).

[0280] Such other agents, or drugs, can be administered, by a route and in an amount commonly used therefor, simultaneously or sequentially with a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab). When a compound of Formula (I) and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) are used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) can be utilized, but is not required. Accordingly, the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to a compound of Formula (I).

PHARMACEUTICAL COMPOSITIONS AND ROUTES OF ADMINISTRATION

[0281] Provided herein is a pharmaceutical composition comprising a compound provided herein (a compound of Formula (I) and/or a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) and a pharmaceutically acceptable excipient, adjuvant, carrier, buffer, or stabilizer. In some embodiments, the compound of Formula (I) and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) are present in the same pharmaceutical composition. In some

embodiments, the compound of Formula (I) and the T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab) are in different pharmaceutical compositions.

[0282] In one embodiment, the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers. The pharmaceutical compositions provided herein that are formulated for oral administration may be in tablet, capsule, powder, or liquid form. In some embodiments, a tablet comprises a solid carrier or an adjuvant. Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil, or synthetic oil. Physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol may be included. In some embodiments, a capsule comprises a solid carrier such as gelatin.

[0283] In another embodiment, the pharmaceutical compositions are provided in a dosage form for parenteral administration, which comprise a compound provided herein, and one or more

pharmaceutically acceptable excipients or carriers. Where pharmaceutical compositions may be formulated for intravenous, cutaneous or subcutaneous injection, the active ingredient will be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has a suitable pH, isotonicity, and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles, such as Sodium Chloride injection, Ringer’s injection, or Lactated Ringer’s injection. In some embodiments, preservatives, stabilisers, buffers, antioxidants, and/or other additives are included as required.

[0284] In yet another embodiment, the pharmaceutical compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, and one or more pharmaceutically acceptable excipients or carriers.

[0285] The pharmaceutical compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, and programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd Edition, Rathbone et al, Eds., Marcel Dekker, Inc.: New York, NY, 2008).

[0286] The pharmaceutical compositions provided herein can be provided in a unit-dosage form or multiple-dosage form. A unit-dosage form, as used herein, refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit-dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit- dosage form may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons.

[0287] The pharmaceutical compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the

administration of the formulations.

[0288] In certain embodiments, the pharmaceutical compositions provided herein further comprise one or more chemotherapeutic agents as defined herein.

A. Oral Administration

[0289] The pharmaceutical compositions provided herein for oral administration can be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration also includes buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, fastmelts, chewable tablets, capsules, pills, strips, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, bulk powders, effervescent or non-effervescent powders or granules, oral mists, solutions, emulsions, suspensions, wafers, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions can contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye -migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide.

[0290] Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as com starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose,

methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL -PH-101, AVICEL-PH- 103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, PA); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre -gelatinized starch, and mixtures thereof. The amount of a binder or filler in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.

[0291] Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets. The amount of a diluent in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.

[0292] Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation -exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as com starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.

[0293] Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® 200 (W.R. Grace Co., Baltimore, MD) and CAB-O-SIL® (Cabot Co. of Boston, MA); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant. [0294] Suitable glidants include, but are not limited to, colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, MA), and asbestos-free talc. Suitable coloring agents include, but are not limited to, any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water- soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Suitable flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Suitable sweetening agents include, but are not limited to, sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include, but are not limited to, gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suitable suspending and dispersing agents include, but are not limited to, sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose,

hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Suitable wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Suitable solvents include, but are not limited to, glycerin, sorbitol, ethyl alcohol, and syrup. Suitable non-aqueous liquids utilized in emulsions include, but are not limited to, mineral oil and cottonseed oil. Suitable organic acids include, but are not limited to, citric and tartaric acid. Suitable sources of carbon dioxide include, but are not limited to, sodium bicarbonate and sodium carbonate.

[0295] It should be understood that many carriers and excipients may serve several functions, even within the same formulation.

[0296] The pharmaceutical compositions provided herein for oral administration can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or fdm -coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film -coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to,

hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets. [0297] The tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants.

Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

[0298] The pharmaceutical compositions provided herein for oral administration can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.

[0299] Coloring and flavoring agents can be used in all of the above dosage forms.

[0300] The pharmaceutical compositions provided herein for oral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

B. Parenteral Administration

[0301] The pharmaceutical compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrastemal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.

[0302] The pharmaceutical compositions provided herein for parenteral administration can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see. Remington: The Science and Practice of Pharmacy, supra).

[0303] The pharmaceutical compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water- miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases. [0304] Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Suitable non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, com oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Suitable water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, L'-mcthy 1 -2-pyrrol idone. N,N- dimethylacetamide, and dimethyl sulfoxide.

[0305] Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl-and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and

polyvinylpyrrolidone. Suitable emulsifying agents are those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include but are not limited to EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including a-cyclodextrin, b-cyclodextrin, hydroxypropyl -b-cyclodextrin, sulfobutylether^-cyclodextrin, and sulfobutylether 7^-cyclodextrin (CAPTISOL®, CyDex, Lenexa, KS).

[0306] When the pharmaceutical compositions provided herein are formulated for multiple dosage administration, the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungi static concentrations. All parenteral formulations must be sterile, as known and practiced in the art.

[0307] In one embodiment, the pharmaceutical compositions for parenteral administration are provided as ready-to-use sterile solutions. In another embodiment, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In yet another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In yet another embodiment, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In still another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.

[0308] The pharmaceutical compositions provided herein for parenteral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms. [0309] The pharmaceutical compositions provided herein for parenteral administration can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In one embodiment, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.

[0310] Suitable inner matrixes include, but are not limited to, polymethylmethacrylate, polybutyl- methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene -vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and metliacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.

[0311] Suitable outer polymeric membranes include but are not limited to, polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.

C. Modified Release

[0312] The pharmaceutical compositions provided herein can be formulated as a modified release dosage form. As used herein, the term“modified release” refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Modified release dosage forms include, but are not limited to, delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix-controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphism of the active ingredient(s).

[0313] Examples of modified release include, but are not limited to, those described in U.S. Pat. Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356; 5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961; 6,589,548; 6,613,358; and 6,699,500.

[0314] Provided herein also are kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a subject. In certain embodiments, the kit provided herein includes one or more containers and a dosage form of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab).

[0315] In certain embodiments, the kit provided herein includes one or more containers and a dosage form of a compound of Formula (I), or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof and a T-cell activating bispecific antigen-binding molecule (e.g., obinutuzumab). Kits provided herein can further include devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes and needle-less injectors drip bags.

[0316] Kits provided herein can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate -free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: aqueous vehicles, including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer’s Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer’s Injection; water-miscible vehicles, including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles, including, but not limited to, com oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.

[0317] The disclosure will be further understood by the following non-limiting examples.

EXAMPLES

[0318] As used herein the symbols and conventions used in these processes, schemes and examples, regardless of whether a particular abbreviation is specifically defined, are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Specifically, but without limitation, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams); mL (milliliters); pL, (microliters); M (molar); mM (millimolar), mM (micro molar); eq. (equivalent); mmol (millimoles), Hz (Hertz), MHz (megahertz); hr or hrs (hour or hours); min (minutes); and MS (mass spectrometry).

[0319] For all of the following examples, standard work-up and purification methods known to those skilled in the art can be utilized. Unless otherwise indicated, all temperatures are expressed in °C (degrees Centigrade). All reactions conducted at room temperature unless otherwise noted. Synthetic

methodologies illustrated herein are intended to exemplify the applicable chemistry through the use of specific examples and are not indicative of the scope of the disclosure.

[0320] Synthesis of Compound A35 is described in US Patent No. 9,056,852 B2, which is incorporated by reference for such disclosure. Example 1: Synthesis of 4-(2-(difluoromcthyl)-l//-bcnzo|£/|imidazol- 1 -yl)-N-(2-mcthyl-l -(2-( 1 - methylpiperidin-4-yl)phenyl)propan-2-yl)-6-morpholino-l,3,5- triazin-2-amine, Compound A35

[0321] A mixture of 4-(2-(difluoromethyl)-lH-benzo[d]imidazol-l-yl)-N-(2-methyl- l-(2-(piperidin-4- yl)phenyl)propan-2-yl)-6-morpholino-l,3,5-triazin-2-amine (80 mg, 0.14 mmol), aq. formaldehyde (37%, 23 mg), and sodium cyanoborohydride (11 mg, 0.17 mmol) in methanol (2 mL) was stirred at room temperature for 1 hr. The crude product was purified by prep-HPLC to give compound A35 (11 mg, 13% yield) as a white solid: 99% purity (HPLC); MS m/z 577.3 (M+l); ¾ NMR (CDCE, 500 MHz) d 8.37 (d, 1H), 7.90 (d, 1H), 7.64 (t, 1H), 7.42 (m, 2H), 7.32 (d, 1H), 7.24 (1, 1H), 7.13 (t, 1H), 7.07 (d, 1H), 5.15 (s, 1H), 4.00-3.70 (m, 8H), 3.28 (s, 2H), 2.94 (m, 2H), 2.78 (m, 2H), 2.28 (s, 3H), 1.891.60 (m, 6H), 1.53 (s, 6H) ppm.

Example 2: Study of a Combination of a PI3K Inhibitor and obinutuzumab in Patients with Chronic Lymphocyctic Leukemia (CLL)

[0322] The purpose of this study is to evaluate the safety and effectiveness of Compound A35, A36,

A68, or A70 and obinutuzumab, in patients with CLL.

[0323] Primary Outcome Measures: Determine Acceptable Adverse Events That Are Related to

Treatment [Time Frame: 6 months of therapy]. To determine the incidence of adverse events, any potential abnormal laboratory results and any dose-limiting toxicities.

[0324] Secondary Outcome Measures: Overall Response Rate [Time Frame: up to one year]. The overall response rate (ORR) in patients with CLL treated with a combination of Compound A35, A36, A68, or A70 and obinutuzumab.

Arms Assigned Interventions

Experimental: obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 30 infusion

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily (oral agent)

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 45 infusion

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 Obinutuzumab oral daily dose - 10-500 mg or A70 a once daily oral agent

Compound A35, A36, A68, or A70 oral daily dose - 60 Obinutuzumab injection, for intravenous mg infusion

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 120 infusion Arms Assigned Interventions

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 150 infusion

mg

[0325] Patients should not have had exposure to the compounds prior to the study entry. Patients must not have received treatment for their cancer within 2 weeks of beginning the trial. Treatments include the use of chemotherapy, hematopoietic growth factors, and biologic therapy such as monoclonal antibodies. Patients must have recovered from all toxicities (to grade 0 or 1) associated with previous treatment. All subjects are evaluated for safety and all blood collections for pharmacokinetic analysis are collected as scheduled. All studies are performed with institutional ethics committee approval and patient consent.

[0326] Doses of the compounds may be held or modified for toxicity based on assessments as outlined below. Treatment repeats every 28 days in the absence of unacceptable toxicity. Dose limiting toxicities are determined according to the definitions and standards set by the National Cancer Institute (NCI) Common Terminology for Adverse Events (CTCAE) Version 3.0 (August 9, 2006).

[0327] Blood Sampling Serial blood is drawn by direct vein puncture before and after administration of the compound. Venous blood samples (5 mL) for determination of serum concentrations are obtained at about 10 minutes prior to dosing and at approximately the following times after dosing: days 1, 8, and 15. Each serum sample is divided into two aliquots. All serum samples are stored at -20 °C. Serum samples are shipped on dry ice.

[0328] Pharmacokinetics: Patients undergo plasma/serum sample collection for pharmacokinetic evaluation before beginning treatment and at days 1, 8, and 15. Pharmacokinetic parameters are calculated by model independent methods on a Digital Equipment Corporation VAX 8600 computer system using the latest version of the BIOAVL software. The following pharmacokinetics parameters are determined: peak serum concentration (C maX ); time to peak serum concentration (t maX ); area under the concentration time curve (AUC) from time zero to the last blood sampling time (AUCo- 72 ) calculated with the use of the linear trapezoidal rule; and terminal elimination half-life (ti / 2), computed from the elimination rate constant. The elimination rate constant is estimated by linear regression of consecutive data points in the terminal linear region of the log-linear concentration-time plot. The mean, standard deviation (SD), and coefficient of variation (CV) of the pharmacokinetic parameters are calculated for each treatment. The ratio of the parameter means (preserved formulation/non-preserved formulation) is calculated.

[0329] Patient Response to combination therapy: Patient response is assessed via imaging with X-ray, CT scans, and MRI, and imaging is performed prior to beginning the study and at the end of the first cycle, with additional imaging performed every four weeks or at the end of subsequent cycles . Imaging modalities are chosen based upon the cancer type and feasibility/availability, and the same imaging modality is utilized for similar cancer types as well as throughout each patient’s study course. Patient response is also assessed via complete blood cell count and/or marrow biopsy. Response rates are determined using the RECIST criteria. (Therasse et al, J. Natl. Cancer Inst. 2000 Feb 2; 92(3):205-16; http://ctep.cancer.gov/forms/TherasseRECISTJNCI.pdf). After completion of study treatment, patients are followed periodically for 4 weeks.

Example 3: Study of a Combination of a PI3K Inhibitor and obinutuzumab in Patients with Follicular Lymphoma (FL)

[0330] The purpose of this study is to evaluate the safety and effectiveness of Compound A35, A36,

A68, or A70 and obinutuzumab, in patients with FL.

[0331] Primary Outcome Measures: Determine Acceptable Adverse Events That Are Related to Treatment [Time Frame: 6 months of therapy]. To determine the incidence of adverse events, any potential abnormal laboratory results and any dose-limiting toxicities.

[0332] Secondary Outcome Measures: Overall Response Rate [Time Frame: up to one year]. The overall response rate (ORR) in patients with FL treated with a combination of Compound A35, A36, A68, or A70 and obinutuzumab.

Arms Assigned Interventions

Experimental: obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 30 infusion

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 45 infusion

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 Obinutuzumab oral daily dose - 10-500 mg or A70 a once daily oral agent

Compound A35, A36, A68, or A70 oral daily dose - 60 Obinutuzumab injection, for intravenous mg infusion

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral agent

Obinutuzumab dose - 10-1000 mg Obinutuzumab injection, for intravenous

Compound A35, A36, A68, or A70 oral daily dose - 120 infusion

mg

Experimental: Obinutuzumab + Compound A35, A36, PI3K Inhibitor: Compound A35, A36, A68, A68, or A70 or A70 a once daily oral

Obinutuzumab dose - 10-1000 mg agentObinutuzumab injection, for

Compound A35, A36, A68, or A70 oral daily dose - 150 intravenous infusion

mg [0333] Patients should not have had exposure to the compounds prior to the study entry. Patients must not have received treatment for their cancer within 2 weeks of beginning the trial. Treatments include the use of chemotherapy, hematopoietic growth factors, and biologic therapy such as monoclonal antibodies. Patients must have recovered from all toxicities (to grade 0 or 1) associated with previous treatment. All subjects are evaluated for safety and all blood collections for pharmacokinetic analysis are collected as scheduled. All studies are performed with institutional ethics committee approval and patient consent.

[0334] Doses of the compounds may be held or modified for toxicity based on assessments as outlined below. Treatment repeats every 28 days in the absence of unacceptable toxicity. Dose limiting toxicities are determined according to the definitions and standards set by the National Cancer Institute (NCI) Common Terminology for Adverse Events (CTCAE) Version 3.0 (August 9, 2006).

[0335] Blood Sampling Serial blood is drawn by direct vein puncture before and after administration of the compound. Venous blood samples (5 mL) for determination of serum concentrations are obtained at about 10 minutes prior to dosing and at approximately the following times after dosing: days 1, 8, and 15. Each serum sample is divided into two aliquots. All serum samples are stored at -20 °C. Serum samples are shipped on dry ice.

[0336] Pharmacokinetics: Patients undergo plasma/serum sample collection for pharmacokinetic evaluation before beginning treatment and at days 1, 8, and 15. Pharmacokinetic parameters are calculated by model independent methods on a Digital Equipment Corporation VAX 8600 computer system using the latest version of the BIOAVL software. The following pharmacokinetics parameters are determined: peak serum concentration (C maX ); time to peak serum concentration (t maX ); area under the concentration time curve (AUC) from time zero to the last blood sampling time (AUCo- 72 ) calculated with the use of the linear trapezoidal rule; and terminal elimination half-life (ti / 2), computed from the elimination rate constant. The elimination rate constant is estimated by linear regression of consecutive data points in the terminal linear region of the log-linear concentration-time plot. The mean, standard deviation (SD), and coefficient of variation (CV) of the pharmacokinetic parameters are calculated for each treatment. The ratio of the parameter means (preserved formulation/non-preserved formulation) is calculated.

[0337] Patient Response to combination therapy: Patient response is assessed via imaging with X-ray, CT scans, and MRI, and imaging is performed prior to beginning the study and at the end of the first cycle, with additional imaging performed every four weeks or at the end of subsequent cycles. Imaging modalities are chosen based upon the cancer type and feasibility/availability, and the same imaging modality is utilized for similar cancer types as well as throughout each patient’s study course. Patient response is also assessed via complete blood cell count and/or marrow biopsy. Response rates are determined using the RECIST criteria. (Therasse et al, J. Natl. Cancer Inst. 2000 Feb 2; 92(3):205-16; http://ctep.cancer.gov/forms/TherasseRECISTJNCI.pdf). After completion of study treatment, patients are followed periodically for 4 weeks

[0338] The examples set forth above are provided to give those of ordinary skill in the art with a complete disclosure and description of how to make and use the claimed embodiments and are not intended to limit the scope of what is disclosed herein. Modifications that are obvious to persons of skill in the art are intended to be within the scope of the following claims.