Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CYCLIN-DEPENDENT KINASE DEGRADERS AND METHODS OF USE
Document Type and Number:
WIPO Patent Application WO/2019/160890
Kind Code:
A1
Abstract:
The present application provides bifunctional compounds of Formula (Ia): (Ia), or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, which act as protein degradation inducing moieties for one or more of cyclin-dependent kinase 8 (CDK8) and cyclin-dependent kinase 19 (CDK19). The present application also relates to methods for the targeted degradation of CDK8 and/or CDK19 through the use of the bifunctional compounds that link a ubiquitin ligase-binding moiety to a ligand that is capable of binding to CDK8 and/or CDK19 which can be utilized in the treatment of disorders modulated by CDK8 and/or CDK19.

Inventors:
GRAY NATHANAEL (US)
HATCHER JOHN (US)
Application Number:
PCT/US2019/017749
Publication Date:
August 22, 2019
Filing Date:
February 13, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DANA FARBER CANCER INST INC (US)
International Classes:
C07J43/00
Domestic Patent References:
WO2017185034A12017-10-26
WO2016054472A12016-04-07
Foreign References:
US20120190659A12012-07-26
US7875599B22011-01-25
US20130252930A12013-09-26
Other References:
CZAKO, B. ET AL.: "Discovery of Potent and Practical Antiangiogenic Agents Inspired by Cortistatin A", J. AM. CHEM. SOC., vol. 131, 2009, pages 9014 - 9019, XP002693619
HATCHER, J. M. ET AL.: "Development of Highly Potent and Selective Steroidal Inhibitors and Degraders of CDK8", ACS MEDICINAL CHEMISTRY LETTERS, vol. 9, 2018, pages 540 - 545, XP055631975
See also references of EP 3752517A4
Attorney, Agent or Firm:
ERLACHER, Heidi et al. (US)
Download PDF:
Claims:
CLAIMS

1. A bifunctional compound of Formula X:

wherein:

the Targeting Ligand is capable of binding to CDK8 and/or CDK19;

the Linker is a group that covalently binds to the Targeting Ligand and the Degron; and the Degron is capable of binding to a ubiquitin ligase.

2. The bifunctional compound of claim 1 , wherein the Targeting Ligand is of Formula TL-I:

or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, wherein:

X is -0-, NR- . or l ies.: :

Rs is H or Ci-Cr, alkyl;

Het? is a 3- to 7-membered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen;

u, v, and w together form (i), (ii), or (iii):

(i) (ii) (Hi)

y and z together form (iv) or (v):

Heti is heteroaryl comprising one or two 5- or 6-membered rings and 1-4 heteroatoms selected from nitrogen, sulfur, and oxygen, and optionally substituted with Nil?.; wherein the Targeting Ligand is bonded to a Linker via the

3. The bifunctional compound of claim 2, wherein u, v, and w together form 4. The bifunctional compound of claim 2, wherein u, v, and w together form

5. The bifunctional compound of claim 2, wherein u, v, and w together form

6 The bifunctional compound of any one of claims 2-5, wherein y and z together form

7. The bifunctional compound of any one of claims 2-5, wherein y and z together form

8. The bifunctional compound of any one of claims 2-7, wherein X is -0-.

9. The bifunctional compound of any one of claims 2-7, wherein X is -NR?---.

10 The bifunctional compound of claim 9, wherein Rs is methyl.

11 The bifunctional compound of any one of claims 2-7, wherein X is -Met?-.

12. The bifunctional compound of any one of claims 2-7 and 11, wherein Het2 is selected from aziridine, azetidine, pyrrolidine, dihydropyrrole, piperidine, piperazine, dihydropyridine, tetrahydropyndme, azepane, oxazindme, oxazetidme, isoxazoline, isoxazolidme, oxazoline, oxazolidine, thiazoline, thiazolidine, isothiazoline, isothiazolidine, pyrazoline, pyrazolidine, imidazoline, imidazoiidine, morpholine, oxazinane, thiomorphoiine, or thiazinane.

13. The bifunctional compound of any one of claims 2-7, 11, and 12, wherein Het? is triazole.

14. The bifunctional compound of any one of claims 2-7, 11, and 12, wherein Het?. is morpholine.

15. The bifunctional compound of any one of claims 2-7, 11, and 12, wherein Het? is piperazine.

16. The bifunctional compound of any one of claims 2-15, wherein Heti is selected from isoquinolinyl, quinolinyl, indazolyl, cinnolinyl, phthalazinyl, pyridinyl, pyridazinyl, indolyl, acridinyl, pyrazinyl, benzoquino!inyl, pyrazo!yl, pyrroly!, pyrimidinyi, purinyl,

pyrrolopyrimidinyl, quinoxalinyl, and quinazolinyl.

17. The bifunctional compound of any one of claims 2-16, wherein Heti is 6-isoqumolinyl, 7- isoquinolmyl, 6-qumazoiinyl, 6-phthalazmyl, 6-indazolyl, 6-indazolyl-3-atnine, or 5-indazolyl.

18. The bifunctional compound of any one of claims 2-17, wherein Heti is 7-isoquino!inyl.

19. The bifunctional compound of claim 2, wherein the Targeting Ligand is of Formula

(iv(a)’), (', (a (iv(a)”), (v(a)”), (iv(b)’), (v(b)’), (iv(b)”), (v(b)”), (iv(c)’), or (v(c)), (iv(c)”), or (v(c)”):

20. The bifunctional compound of claim 2, wherein the Targeting Ligand is of one of the following formulae:

wherein:

a, b, c, and d are each independently CRe or N; and

each Re is independently H or NH2. 21. The bifunctional compound of claim 2, wherein the Targeting Ligand is of one of the following formulae:

22. The bifunctional compound of claim 2, wherein the targeting Ligand is of one of the following formulae:

wherein:

e and f are each independently CR? or N; and

each R? is independently be H or Nth.

23. The bifunctional compound of claim 2, wherein the Targeting Ligand is selected from Table 1.

24. The bifunctional compound of any one of claims 1-23, wherein the Linker is of Formula 1.0:

or an enantiomer, diastereomer, or stereoisomer thereof, wherein

pi is an integer selected from 0 to 12;

p2 is an integer selected from 0 to 12;

p3 is an integer selected from 0 to 6;

each W is independently absent, CH2, O, S, NH, or NRis;

Z3 is absent, C(O), (CH2)jC(0)NH, CH2, O, NH, or NRux

each Rig is independently C1-C3 alkyl; j is 1, 2, or 3; and

Q is absent, CHz, C(O), or NHC(0)CHz, wherein the Linker is covalently bonded to a Degron via the next to Q, and covalently bonded to a Targeting Ligand via the next to Z3.

25. The bifunctional compound of claim 24, wherein the Linker is of Formula TO:

wherein:

p2 is 0;

p3 is 2;

each W is ();

Zi is C(O); and

Q is absent; wherein the Linker is covalently bonded to the Degron via the next to Q, and covalently bonded to the Targeting Ligand via the next to Z3.

26. The bifunctional compound of claim 24, wherein the Linker is selected from:

27. The bifunctional compound of claim 24, wherein the Linker is

28. The bifunctional compound of any one of claims 1-27, wherein the Degron is of Formula

or an enantiomer, diastereomer, or stereoisomer thereof, wherein:

Y is a bond, (CH2)I-6, (CH2)o-6-0, (CH2)o-6-C(0)NRii, (CH2)o-6-NRiiC(0), (CH2)o-6-NH, or (CH2)O-6-NRI 2;

Z2 is C(O) or C(Ri3)2;

Rn is H or Ci-Ce alkyl;

RI2 is Ci-Ce alkyl or C(0)-Ci-C6 alkyl;

each Rii is independently H or C -C3 alkyl;

each Ri4 is independently C1-C3 alkyl; R15 is H, deuterium, C1-C3 alkyl, F, or Cl;

each Rio is independently halogen, OH, Ci-Ce alkyl, or Ci-Ce alkoxy;

q is 0, 1, or 2; and

s is 0, 1 , 2, or 3, wherein the Degron is covalently bonded to a Linker via

29. The bifunctional compound of claim 28, wherein Zi is C(O).

30. The bifunctional compound of claim 28, wherein Z2 is C(O).

31. The bifunctional compound of claim 28, wherein Zi and Z2 are each is C(Q).

32. The bifunctional compound of claim 28, wherein Zi is C(0) and Z2 is (Ί 1 ··. 33. The bifunctional compound of any one of claims 28-32, wherein Y is a bond.

34. The bifunctional compound of claim 28 or 33, wherein the Degron is of the following formula:

35. The bifunctional compound of any one of claims 28-34, wherein Rn is H.

36. The bifunctional compound of any one of claims 28, 33, 34, and 35, wherein the Degron is of the following formula:

wherein R13 is H.

37 The bifunctional compound of claim 1, having the following structure:

39. The bifunctional compound of claim 1, having the following structure:

40. The bifunctional compound of claim 1, having the following structure:

41. The bifunctional compound of claim 1, having the following structure:

42. A pharmaceutical composition comprising a therapeutically effective amount of the bifunctional compound of any one of claims 1-41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

43. A method of inhibiting or modulating cyc!in-dependent kinase 8 (CDK8) and/or cyclin- dependent kinase-19 (CDK19), comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1-41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 42.

44. A method of treating or preventing a disease in winch one or more of CDK8 and CDK19 plays a role, comprising administering to a subject in need thereof an effective amount of a compound of any one of claims 1 -41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 42.

45. The method of claim 41, wherein the disease is cancer or a proliferation disease.

46. A bifunctional compound of any one of claims 1-41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 42, for use in the manufacture of a medicament for modulating one or more of CDK8 and CDK19 or treating or preventing a disease m which one or more of CDK8 and CDK19 plays a role.

47. A bifunctional compound of any one of claims 1 -41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 42, for use in modulating one or more of CDK8 and CDKI 9 or treating or preventing a disease in which one or more of CDK8 and CDK19 plays a role.

48. Use of a bifunctional compound of any one of claims 1-41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof or the pharmaceutical composition of claim 42, in modulating one or more of CDK8 and CDKi 9 or treating or preventing a disease in which one or more of CDK8 and CDKI 9 plays a role.

49. A kit comprising a compound of any one of claims 1 -41, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 42.

Description:
CYCLIN-DEPENDENT KINASE DEGRADERS AND METHODS OF USE

CROSS-REFERENCE TO RELATED APPLICATION

This application claims priority to, and the benefit of, US. Provisional Application No. 62/629,753, filed on February 13, 2018, the entire contents of which are incorporated herein by reference.

BACKGROUND

Cyclin-dependent kinase is a kinase family integrating multiple signaling pathways to control either cell cycle or gene transcription. CDK! , 2, 4 and 6 are the critical enzymes that drive cell cycle transition. CDK7, 9 and 12 are known enzymes that regulate transcription instead of directly promoting cell cycles. CDK8 forms part of a mediator complex that regulates the transcriptional activity of RNA polymerase II, and thereby regulates cellular proliferation and differentiation. CDK8 has also been shown to modulate the transcriptional output from distinct transcription factors involved m oncogenic control, including the Wnt/p-catenin pathway, Notch, p53, and TGF-b. CDK8 has been found to be amplified and overexpressed in colon and gastric cancer by acting through the Wnt pathway and its key signaling molecule b-eatenin. In addition, CDK8 gene expression correlates with increased mortality in breast and ovarian cancers, and is essential for cell proliferation in melanoma. Similar to CDK8, CDK19 is one of the components of this mediator co-activator complex.

Ubiquitin-Proteasome Pathway (UPP) is a critical pathway that regulates proteins and degrades misfolded or abnormal proteins. UPP is central to multiple cellular processes, and if defective or imbalanced, leads to pathogenesis of a variety of diseases. The covalent attachment of ubiquitin to specific protein substrates is achieved through the action of E3 ubiquitin ligases. These ligases comprise over 500 different proteins and are categorized into multiple classes defined by the structural element of their E3 functional activity. For example, cereblon (CRBN) interacts with damaged DNA binding protein 1 and forms an E3 ubiquitin hgase complex with Cullin 4 in which the proteins recognized by CRBN are ubiquitinated and degraded by proteasomes. Various immunomodulatory drugs (IMiDs) bind to CRBN and modulate CRBN’s role in the uhiquitination and degradation of protein factors involved in maintaining regular cellular function. Bifunctional compounds composed of a target protein-binding moiety and an E3 ubiquitin ligase-binding moiety have been shown to induce proteasome-mediated degradation of selected proteins.

Deregulation of CDKs has been shown to have a significant impact on the cell state and is frequently identified as oncogenic. Numerous selective or pan-CDK small molecule inhibitors have been identified, however, most of the known inhibitors have failed m clinic trials due to the lack of high systemic drug concentration. Alternative strategies to inhibit cyclin-dependent kinases, such as CDKS and/or CDK19, are needed. At present, compounds with alternative mechanisms of action targeting CDKS and/or CDK19 are not available. The present application addresses the need.

SUMMARY

The present application relates to novel bifunctional compounds, which function to recruit CDK8 and/or CDK19 to E3 ubiquitin hgase for degradation, and methods of preparation and uses thereof. The bifunctional compound is of Formula X:

wherein:

the Targeting Ligand is capable of binding to CDK8 and/or CDK19;

the Linker is a group that covalently binds to the Targeting Ligand and the Degron; and the Degron is capable of binding to a ubiquitin tigase, such as an E3 ubiquitin ligase (e.g., cereblon).

The present application also relates to targeted degradation of CDK8 and/or CDK! 9 through the use of bifunctional compounds, including bifunctional compounds that link an E3 ubiquitin ligase-binding moiety to a ligand that binds to CDK8 and/or CDK19.

The present application also relates to a bifunctional compound of Formula la:

Targeting Ligand

or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, wherein:

7 X, u, v, w, y, z, and Heti are each as defined herein;

in Formula (la) and the Degron;

the Degron is capable of binding to a ubiquitin ligase, such as an E3 ubiquitm ligase (e.g., cereblon); and

the Targeting Ligand is capable of binding to CDK8 and/or CDKI9.

The present application also relates to a pharmaceutical composition comprising a therapeutically effective amount of a bifunctional compound of the application, or an

enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

Another aspect of the present application relates to a method of modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cyclin-dependent kinase, such as CDK8 and/or CDK19). The method comprises administering to a subject in need thereof an effective amount of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

Another aspect of the present application relates to a method of treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role). The method comprises administering to a subject in need thereof an effective amount of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application. In one aspect, the disease is a kinase (e.g., CDK8 and/or CDK19) mediated disorder. In one aspect, the disease is a proliferative disease (e.g., a proliferative disease in which CDK8 and/or CDK19 plays a role).

Another aspect of the present application relates to a method of treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or an activated CDK19 or wherein the subject is identified as being in need of CDK8 and/or CDK19 inhibition for the treatment or prevention of cancer. The method comprises administering to the subject an effective amount of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

Another aspect of the present application relates to a kit comprising a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof.

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use m the manufacture of a medicament for modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., CDK8 and/or CDK19).

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in the manufacture of a medicament for treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role). In one aspect, the disease is a kinase (e.g., CDK8 and/or CDK19) mediated disorder. In one aspect, the disease is a proliferative disease (e.g, a proliferative disease in which CDK8 and/or CDK19 plays a role).

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in the manufacture of a medicament for treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or an activated CDK19 or wherein the subject is identified as being in need of CDK8 and/or CDK19 inhibition for the treatment or prevention of cancer.

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g, CDK8 and/or CDK19).

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role). In one aspect, the disease is a kmase (e.g., CDK8 and/or CDK19) mediated disorder. In one aspect, the disease is a proliferative disease (e.g., a proliferative disease in winch CDK8 and/or CDK19 plays a role).

Another aspect of the present application relates to a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or an activated CDK19 or wherein the subject is identified as being in need of CDK8 and/or CDK19 inhibition for the treatment or prevention of cancer.

Another aspect of the present application relates to use of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, in modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., CDK8 and/or CDK19).

Another aspect of the present application relates to use of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, in treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role). In one aspect, the disease is a kmase (e.g., CDK8 and/or CDK19) mediated disorder. In one aspect, the disease is a proliferative disease (e.g., a proliferative disease in which CDK8 and/or CDK19 plays a role).

Another aspect of the present application relates to use of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, in treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or an activated CDK19 or wherein the subject is identified as being in need of CDK8 and/or CDK19 inhibition for the treatment or prevention of cancer.

The present application provides modulators (e.g., degraders or inhibitors) of CDK8 and/or CDK19 that are therapeutic agents in the treatment or prevention of diseases such as cancer and metastasis.

The present application further provides compounds and compositions with an improved efficacy and/or safety profile relative to known CDK8 and/or CDK19 inhibitors. The present application also provides agents with novel mechanisms of action toward CDK8 and/or CDK19 kinases in the treatment of various types of diseases including cancer and metastasis. The compounds and methods of the present application address unmet needs in the treatment of diseases or disorders in which pathogenic or oncogenic endogenous proteins f e.g., CDK8 and/or CDK! 9) play a role, such as cancer.

The details of the disclosure are set forth m the accompanying description below.

Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present application, illustrative methods and materials are now described. In the case of conflict, the present specification, including definitions, wall control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting. Other features, objects, and advantages of the disclosure wall be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwase. Unless defined otherwase, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.

The contents of all references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties by reference. The references cited herein are not admitted to be prior art to the application.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a western blot showing the levels of CDK8, pS727-STATl , and aetin in Jurkat cells treated with 0.1 mM or 1 mM Compound I- 1 for either 6 h or 24 h.

FIG. 2 is a western blot showing the levels of CDK8, pS727-STATl , and aetin in MC38 colorectal cancer cells treated with 50 nM, 100 nM, 500 nM, or 1 mM Compound 1-1 for either 6 h or 24 h.

FIG. 3 is a western blot showing the levels of CDK8 and aetin in WT Molt4 cells or CZ?Z¾V-/-Molt4 cells treated with 0.1 mM or 1 mM Compound I- 1

FIG. 4 is a western blot illustrating inhibition of phosphorylation of S727-STAT1, a known CDK8 substrate, to form pS727~STATl pS727-STATl and total STAT1 were from HepG cells independently treated for 1 8 h with interferon gamma (IFNy) and 0.5 mM, I mM, or 5 mM Compound 33, or with IFNy and 0.1 mM or 1 mM dehydrocortistatin A (“DCA”). DCA has an CDK8 ICso value of 17 nM. Compound 33 and DCA induced dose-dependent inhibition of S727 phosphorylation in HepG2 cells.

FIG. 5 is a western blot showing the levels of CDK8, pS727-STATl, and actin in Jurkat cells independently treated with 0.1 mM or 1 mM Compound 1-1 or Compound 1-4 for either 6 h or 24 h.

FIG. 6 is a western blot showing the levels of CDK8 and actin in WT Molt4 cells or Ci?SAV-Molt4 cells treated with increasing concentrations of Compound 1-1. Lysates w¾re analyzed after 24 h.

FIG. 7 is a western blot showing the levels of CDK8 and actin in Jurkat cells treated with increasing concentrations of Compound 1-2. Lysates were analyzed after 24 h.

FIG. 8 is a western blot showing the levels of CDK8 and actin in Jurkat cells treated with increasing concentrations of Compound 1-5. Lysates were analyzed after 24 h.

DETAILED DESCRIPTION

Compounds of the Application

The present application relates to bifunctional compounds having utility as modulators of ubiquitination and proteosomal degradation of targeted proteins, especially compounds comprising a moiety capable of binding to a polypeptide or a protein that is degraded and/or otherwise inhibited by the bifunctional compounds of the present application. In particular, the present application is directed to compounds which contain a moiety, e.g., a small molecule moiety (i.e., having a molecular weight of below 2,000, 1 ,000, 500, or 200 Daltons), such as a thalidomide-like moiety, which is capable of binding to an E3 ubiquitm ligase, such as cereblon, and a ligand that is capable of binding to a target protein, in such a way that the target protein is placed in proximity to the ubiquitm ligase to effect degradation (and/or inhibition) of that protein.

In one embodiment, the present application provides a bifunctional compound of Formula X:

wherein:

the Targeting Ligand is capable of binding to CDK8 and/or CDK19; the Linker is a group that covalently binds to the Targeting Ligand and the Degron; and the Degron is capable of binding to a ubiquitin ligase, such as an E3 ubiquitin ligase (e.g., cereblon).

In one embodiment, the present application provides a compound of Formula la:

Targeting Ligand

or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, wherein:

X, u, v, w, y, z, and Heti are each as defined herein;

the Degron; the Degron is capable of binding to a ubiquitin ligase, such as an E3 ubiquitin ligase (e.g., cereblon); and

the Targeting Ligand is capable of binding to CDK8 and/or CDK19

Targeting Ligand (TL) (or target protein moiety or target protein ligand or ligand) is a small molecule which is capable of binding to a target protein of interest, such CDK8 and/or

CDK19.

In one embodiment, a Targeting Ligand is a compound of Formula TL-I:

or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, wherein:

X is— O— , -MRs-, or -Het2-; R5 is H or Ci-Ce alkyl;

Het?. is a 3- to 7-membered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen;

u, v, and w together form (1), (ii), or (iii):

y and z together form (iv) or (v):

Heti is heteroaryl comprising one or two 5- or 6-membered rings and 1-4 heteroatoms selected from nitrogen, sulfur, and oxygen, and optionally substituted with NH?; wherein the Targeting Ligand is bonded to a Linker via the ¾ .

For a compound of Formula TL-I, in various embodiments where applicable, X, u, v, w, y, z, Rs, Heti, and Het? are each as described below.

In one embodiment, X is -0--. In one embodiment, X is (R) 0-. In one embodiment, X is (S) 0 .

In one embodiment X is -NRs- In one embodiment, X is (Z?)-NR5- In one embodiment, X is (5)-NR5-.

In one embodiment X is -0-, (5)-0- (R} 0-, -NRs-, (5)-NR5-, or (i?)-NRs-.

R or S configuration may be determined by methods readily known in art, such as by assigning priority groups about a stereogenic carbon.

In one embodiment, Ri is H.

In one embodiment, Rs is Ci-Ce straight-chain or C3-C6 branched alkyl (e.g., methyl, ethyl, «-propyl, /-propyl, «-butyl, /-butyl, x-butyl, /-butyl, pentyl, or hexyl).

In one embodiment, Rs is C1-C4 straight-chain or C3-C4 branched alkyl (e.g., methyl, ethyl, «-propyl, /-propyl, «-butyl, /-butyl, x-butyl, or /-butyl).

In one embodiment, R5 is Ci-CX alkyl optionally substituted with OH, 0-(Ci-C6 alkyl), NFL·, NH(Ci-Ce alkyl), or N(Ci-Ce alkyl) 2 . In one embodiment R? is methyl.

In one embodiment, X is -Het2-. In one embodiment, X is (i¾)-Iiei2-. In one embodiment, X is {X)---Het2---.

In one embodiment, Beta is a 3- to 7-membered heterocyclic ring. In one embodiment, the heterocyclic ring is selected from aziridine, azetidine, pyrrolidine, dihydropyrrole, piperidine, piperazine, dihydropyridine, tetrahydropyridine, azepane, oxaziridine, oxazetidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, thiazoline, thiazolidine, isothiazoline, isothiazolidine, pyrazoline, pyrazolidine, imidazoline, imidazolidine, morpholine, oxazinane, thiomorpholme, thiazinane, or the like. In one embodiment, the heterocyclic ring is morpholine. In one embodiment, Hetz is a 5-, 8-, or 9-membered heteroaryl ring (e.g., pyrrole, pyrazole, imidazole, triazole, tetrazole, indole, benzimidazole, indazole, azaindoleand the like). In one embodiment, the heteroaryl ring is selected from pyrrole, imidazole, 1 ,2,3-triazole, 1,2,4-triazole, and tetrazole. In one embodiment, the heteroaryl ring is triazole. In one embodiment, the heteroaryl ring 1 ,2,3-triazole;

In one embodiment, u, v, and w together form

In one embodiment, u, v, and w together form

In one embodiment, u, v, and w together form

In one embodiment, v and z together form

In one embodiment, y and z together form M

In one embodiment Heti is heteroaryl selected from isoquinolinyl, quinolinyl, indazoiyl, cinnolinyl, phthalazinyl, pyridinyl, pyridazinyl, indolyl, acridinyl, pyrazinyl, benzoquinolinyl, pyrazolyi, pyrrolyl, pyrimidinyl, purinyl, pyrrolopyrimidinyl, quinoxalinyl, and quinazolinyl. In one embodiment, Heti is selected from 6-isoquinolinyl, 7-isoquinolinyl, 6-quinazolinyl, 6- phthalazinyl, 6-indazolyl, 6-indazolyl-3-amine, and 5-indazo!yl. In one embodiment, Heti is 7- isoquinolmyl.

In one embodiment, Heti is in the R configuration. In one embodiment, Heti is in the S configuration.

In one embodiment, a Targeting Ligand is not a compound of the following formula

Any of the groups described herein for any of X, u, v, w, y, z, Rs, Heti , and Het? can be combined with any of the groups described herein for one or more of the remainder of X, u, v, w, y, z, Rs, Heti, and Het?..

(1) In one embodiment, u, v, and w together form , and y and z together

form

(2) In one embodiment, u, v, and w together form , and y and z together

form

(3) In one embodiment, u, v, and w together form , and y and z together

form

(4) In one embodiment, u, v, and w together form (Ϊ) and y and z together

form

(5) In one embodiment, u, v, and w together form , and y and z together

form ' V i

(6) In one embodiment, u, v, and w together form , and y and z together

(7) in one embodiment, u, v, w, y, and z are each as defined and combined in any one of (l)-(6), and X is -Q-. In a further embodiment, X is (-S )- 0-. In a further embodiment, X is (ii)- (>-.

(8) In one embodiment, u, v, w, y, and z are each as defined and combined m any one of

(l)-(6), and X is -NRs- In a further embodiment, X is (i?)-NR5- In a further embodiment, X is (5)-NR5-

(9) In one embodiment, u, v, w, y, and z are each as defined and combined in any one of (l)-(6), and X is -Het2- In a further embodiment, X is (i?)-Het2- In a further embodiment, X is (5}-Het2-

(10) In one embodiment, u, v, w, y, z, and X are each as defined and combined, where applicable, m any one of (l)-(6) and (8), and Rs is Ci-Ce alkyl as described herein. In a further embodiment, R is methyl (1 1) In one embodiment, u, v, w, y, z, and X are each as defined and combined, where applicable, in any one of (l )-(6) and (8), and Rs is H.

(12) In one embodiment, u, v, w, y, z, X, and Rs are each as defined and combined, where applicable, m any one of (1 )-(l 1), and Heti, where applicable, is in the (S) configuration.

(13) In one embodiment, u, v, w, y, z, X, and Rs are each as defined and combined, where applicable, in any one of (l)-(l 1), and Heti is heteroaryl comprising two 5- or 6-membered rings and 1-4 heteroatoms selected from nitrogen, sulfur, and oxygen. In a further embodiment, Heti is heteroaryl comprising two 5- or 6-membered rings and 1-4 nitrogen atoms. In a further embodiment, Heti, where applicable, is in the (5) configuration. In one embodiment, Heti is selected from the heteroaryl groups described herein.

(14) In one embodiment, u, v, w, y, z, X, and Heti are each as defined and combined, where applicable, in any one of (I)-(6), (9), (12), and (13), Het2 is a 3- to 7-membered heterocyclic or 5-, 8-, or 9-membered heteroaryl ring. In one embodiment, I le· ·. is a 3- to 7- membered heterocyclic ring. In one embodiment, Heti is a 5-, 8-, or 9-membered heteroaryl ring. In one embodiment, Het2 is selected from the heterocyclic groups described herein. In one embodiment, Het 2 is selected from the heteroaryl groups described herein.

In one embodiment a compound of Formula TL~X is of Formula (F), (ii’), (in’), (iv(a)), (iv(b)), (iv(c)), (v(a)), (v(b)), or (v(c)):

For a compound of Formula (F), (iF), (iii’), (iv(a)), (iv(b)), (iv(c)), (v(a)), (v(b)), or (v(c)), where applicable:

In one embodiment, X is -NR5-. For example, X is -N(CH3)- or --NH-.

In one embodiment X is -0-.

In one embodiment, X is -Het2- and Het; is a morpholmyl ring.

In one embodiment, X is -Het2- and Het? is piperazinyl ring.

In one embodiment, X is -Het?- and Het; is triazolyl ring.

In one embodiment, the carbon to which X is attached is designated with R stereochemistry (/.<?., X is (ii)-NR5-, (/?)-()-, or (ii)-Het 2- ).

In one embodiment, the carbon to which X is attached is designated with S stereochemistry

In one embodiment, y and z together

in one embodiment, y and z together form ‘ v

In one embodiment, a compound of Formula TL-I is of Formula (iv(a)’), (v(a)’), (iv(a)”), (v(a)”), (iv(b)’), (v(b)’), (iv(b)”), (v(b)”), (iv(c)’), (v(c)’), ( i\ (c) ' ). or (v(c)”):

wherein:

X is (), -NR5-, or I lei :

Rs is H or Ci-Ce alkyl; and

Het2 is a 3- to 7-membered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen.

In one embodiment,

In one embodiment, X is -NR5-.

In one embodiment, X is -NRs- and Rs is methyl.

In one embodiment, X is -NRs- and Rs is H.

In one embodiment, X is -Het2- In one embodiment, X is -Het2- and Het; is a morpholinyl ring

in one embodiment, X is -Het2- and Het? is piperazinyl ring.

In one embodiment, X is -Het ?- and Het; is triazolyl ring.

in one embodiment, a compound of Formula TL-I is of Formula (iv(a)’), (v(a)’), (iv(a)”), (v(a)”), (iv(b)’), (v(b)’), (iv(b)”), (v(b)”), iv(c)”), or (v(c)”), as described above.

In one embodiment, a compound of Formula TL-I is of Formula (iv(c)’) or (v(c)’), as described above.

In one embodiment, a compound of Formula TL-I is of one of the following formulae:

wherein:

Rs is H or C1-C6 alkyl;

Het2 is a 3- to 7-membered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen;

a, b, c, and d are each independently CRe or N; and

each Re is independently H or NIL.

In one embodiment, X is -()--.

In one embodiment, X is -NRs- In one embodiment, X is -NRs- and Rs is methyl.

In one embodiment, X is -NRs- and Rs is H.

In one embodiment, b and d are each independently CRe; a and c are each N; and one Re is H, and one Re is NHa.

In one embodiment, a, b, and d are each independently CRe; c is N; and each Re is

H.

In one embodiment, X is -Het2-

In one embodiment, X is -Heta- and Het2 is a morpholinyl ring.

In one embodiment, X is -Heta- and Heta is piperazmyl ring.

In one embodiment, X is -Heta- and Heta is triazolyl ring.

In one embodiment, a compound of Formula TL-I is of one of the following formulae:

wherein:

Rs is H or Ci-Ce alkyl; and

Heta is a 3- to 7-mernbered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen.

In one embodiment, X is -()-. In one embodiment, X is -NRs-.

In one embodiment, X is -NR5- and Ra is methyl.

In one embodiment, X is -NRs- and Rs is H.

in one embodiment, X is -Het?.-.

In one embodiment, X is -Beta- and Beta is a morpholinyl ring.

In one embodiment, X is -Beta- and Beta is piperazinyi ring.

In one embodiment, X is -Beta- and Beta is triazoiyl ring.

In one embodiment, a compound of Formula TL-X is of one of the following formulae:

w'herein:

X is— O— , -NR5-, or -Beta-;

Rs is B or Ci-Ce alkyl;

Hefo is a 3- to 7-membered heterocyclic or a 5-, 8-, or 9-membered heteroaryl ring optionally comprising one or more additional heteroatoms selected from nitrogen, sulfur, and oxygen;

R? is H or Ci-Ce alkyl;

e and f are each independently CRs or N; and

each Rs is independently be H or NHa.

In one embodiment X is -O- In one embodiment, X is -NRs-.

In one embodiment, X is -NR5- and R-: is methyl.

In one embodiment, X is -NRs- and Rs is H.

in one embodiment, e is N; f is CRs; and Rs is H.

In one embodiment, e is N; f is CRs: and Rs is NH2.

In one embodiment, e is CRs; f is N; Rs is NHa; and R? may be H.

In one embodiment, e is CRs; f is N; Rs is H; and R? may be H.

In one embodiment, X is -Beta--.

In one embodiment, X is -Beta- and Beta is a morpholinyl ring.

In one embodiment, X is -Beta-- and Beta is piperazinyi ring.

In one embodiment, X is -Bet2- and Beta is triazoiyl ring.

In several embodiments, a compound of Formula TL-I may be represented by one of compounds in the right column in Table 1, where the left column shows the structure of the compounds from which a compound of Formula TL-I may be prepared.

21

22







F Target-Link er-Degrons 1-1, 1-2, and 1-3 prepared from compound 46.

* Target-Linker-Degrons may be prepared from mono-methylated derivative.

p Target-Linker-Degron may be prepared by, for example, via General Scheme 2. in several embodiments, a compound of Formula TL-I may be represented by one of:

In several embodiments, a compound of Formula TL-I may be represented by one of:

In several embodiments, a compound of Formula TL-I may be represented by one of:

In several embodiments, a compound of Formula TL-I may be represented by one of:

In several embodiments, a compound of Formula TL-I may be represented by one of:

in several embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

In some embodiments, a compound of Formula TL-I may be represented by one of:

Degron

The Degron serves to link a targeted protein, through a Linker and a Targeting Ligand, to a ubiquitin !igase for proteosomal degradation. In one embodiment, the Degron is capable of binding to a ubiquitin ligase, such as an E3 ubiquitin ligase. In one embodiment, the Degron is capable of binding to cereblon.

In one embodiment, the Degron is of Formula Dl :

or an enantiomer, diastereomer, or stereoisomer thereof, wherein: Y is a bond, (CH 2 )I-6, (CH 2 )o-6-0, (CH2)o-6-C(0)NRn, (CH 2 )o-6-NRiiC(0), (CH 2 )o-6-NH, or (CH 2 )O-6-NRI 2 ;

ZJ is C(O) or C(Ri3) 2 ;

Z 2 is C(O) or C(Ri3) 2 ;

Rn is H or Ci-Cr, alkyl;

Rn IS C -Cr, alkyl or C(0)-Ci-C6 alkyl;

each Ri3 IS independently H or C1-C3 alkyl;

each R14 is independently C1-C3 alkyl;

Ris is H, deuterium, C1-C3 alkyl, F, or Cl;

each Ris is independently halogen, OH, Ci-Ce alkyl, or C1-C6 alkoxy;

q is 0, 1, or 2; and

s is 0, 1, 2, or 3, wherein the Degron is covalently bonded to a Linker via .

In one embodiment, Zi is C(O).

in one embodiment, Zi is C(Ri F- and each R13 is H. In one embodiment, Zi is C(Ri3) 2 ; and one of Rn is H, and the other is C1-C3 alkyl selected from methyl, ethyl, and propyl. In one embodiment, Zi is C(R ; and each Rn is independently selected from methyl, ethyl, and propyl.

In one embodiment, Z 2 is C(O).

In one embodiment, Z 2 is C(Rn) 2 ; and each Rn is H. In one embodiment, Z 2 i s C(Rn) 2 ; and one of Rn is H, and the other is C1-C3 alkyl selected from methyl, ethyl, and propyl. In one embodiment, Z 2 is C(Rn) 2 ; and each Rn is independently selected from methyl, ethyl, and propyl.

In one embodiment, Zi and Z 2 are each C(O).

In one embodiment, Zi is C(O); and Z 2 is C(RD)2 and each Rn is H. In one embodiment, Z 2 is C(Rn) 2 ; and one of Rn is H, and the other is C1-C3 alkyl selected from methyl, ethyl, and propyl. In one embodiment, Z 2 is C(Rn) 2 ; and each Rn is independently selected from methyl, ethyl, and propyl .

In one embodiment, Y is a bond.

In one embodiment, Y is a bond, O, or NH. In one embodiment, one embodiment, Y is (Cl Hi. (CH?.)?., or (CH?)?. In one embodiment, Y is (CH?)i or (CI-I?)?.

In one embodiment, Y is O,€Ή?-0, (CH?)?-©, (CH?)?-©, (CI-I?)4-0, (P ίΊ'-O. or (CH?)6- O. In one embodiment, Y is O, CH?-0, (CH?)?-0, or (CH?)?-©. In one embodiment, Y is O or CH?-0. In one embodiment, Y is O.

In one embodiment, Y is C(0)NRn, CH?-C(0)NRn, (CH?)?-C(0)NRn, (CH?)?~

C(0)NRn, (CH?) 4 -C(0)NRII, (CH 2 )S-C(0)NRJ I, or (CH 2 )6-C(0)NRii. In one embodiment, Y is C(0)NRn, CH2-C(0)NRI I , (CH?)?-C(0)NRn, or (CH?)3-C(0)NRn. In one embodiment, Y is C(0)NRn or CH?-C(0)NRn. In one embodiment, Y is C(0)NRii.

In one embodiment, Y is NRnC(O), CH?-NRiiC(0), (C l !?}?- NR i iC (O). (CH?)?- NRiiC(O), (CH?)4-NRIIC(0), (CH?)5-NRI IC(0), or (CH?)6-NRI IC(0). In one embodiment, Y is NRiiC(O), CH?~NRiiC(0), (CH?) 2 -NRnC(0), or (CH?)?-NRIIC(0). In one embodiment, Y is NRiiC(O) or CH?-NRiiC(0). In one embodiment, Y is NRnC(O).

In one embodiment, Rn is H. In one embodiment, Rn is selected from methyl, ethyl, propyl, butyl, i-butyl, t-butyl, pentyl, i-pentyl, and hexyl. In one embodiment, Rn is Ci-C? alkyl selected from methyl, ethyl, and propyl.

In one embodiment, Y is NH, (Ή.ΆI I. (CH?)?-NH, (CH?)?-NH, (CH?)4-NH, (CH?)s-NH, or (CH?)6-NH. In one embodiment, Y is NH, CH?-NH, (CH?)?-NH, or (CH?)?-NH. In one embodiment, Y is NH or CH?-NH. In one embodiment, Y is NH.

In one embodiment, Y is NRi?, CH?-NRi?, (CH?)?-NRi?, (CH?)?-NRi?, (CH?) 4 -NRi2, (CH?)5-NRI?, or (CH?)6-NRI?. In one embodiment, Y is NRi?, CH?~NRi?, (CH?)?-NRi?, or (CH?)?~NRi?. In one embodiment, Y is NRi? or CH?-NRi?. In one embodiment, Y is NRi?

In one embodiment, Ri? is selected from methyl, ethyl, propyl, butyl, i-butyl, t-butyl, pentyl, i-pentyl, and hexyl. In one embodiment, Rr? is Ci-C? alkyl selected from methyl, ethyl, and propyl.

In one embodiment, Ri? is selected from C(0)-methyl, C(0)-ethyl, C(0)-propyJ, C(O)- butyl, C(0)-i-butyl, C(0)-t-butyl, C(0)-pentyl, C(0)-i-pentyl, and C(0)-hexyl. In one embodiment, Ri? is C(0)-Ci-C? alkyl selected from C(0)-methyl, C(0)-ethyl, and C(0)-propyl.

In one embodiment, Ri? is H.

In one embodiment, Ri? is Ci-C? alkyl selected from methyl, ethyl, and propyl. In one embodiment, Rn is methyl. In one embodiment, q is 0.

in one embodiment, q is 1.

In one embodiment, q is 2.

in one embodiment, each RM is independently C1-C3 alkyl selected from methyl, ethyl, and propyl.

In one embodiment, s is 0.

In one embodiment, s is 1.

In one embodiment, s is 2.

In one embodiment, s is 3.

In one embodiment, each Rie is independently selected from halogen (e.g., F, Cl, Br, and I), OH, Ci-Ce alkyl (e.g., methyl, ethyl, propyl, butyl, i-butyl, t- butyl, pentyl, i-pentyl, and hexyl), and Ci-Ce alkoxy (e.g., methoxy, ethoxy, propoxy, butoxy, i-hutoxy, t-butoxy, and pentoxyj. In a further embodiment each Rie is independently selected from F, Cl, OH, methyl, ethyl, propyl, butyl, i-butyl, t-butyl, methoxy, and ethoxy.

In one embodiment, Ris is H, deuterium, or C1-C3 alkyl. In another embodiment, R15 is H or C1-C3 alkyl. In a further embodiment, R15 is in the (S) or (R) configuration. In a further embodiment, R l5 is in the (S) configuration. In one embodiment, the compound comprises a racemic mixture of (<S)-Ris and (Afi-Ris.

In one embodiment, Ris is H.

In one embodiment, Ris is deuterium.

In one embodiment, Ris is C1-C3 alkyl selected from methyl, ethyl, and propyl. In one embodiment, Ris is methyl.

In one embodiment, Ris is F or Cl. In a further embodiment, Ris is in the (S) or (R) configuration. In a further embodiment, Ris is in the f R) configuration. In one embodiment, the compound comprises a racemic mixture of (.S>-Ri· ; and (i?)-Ris. In one embodiment, Ris is F.

Any of the groups described herein for any of Y, Zi, Z2, R11, R12, R13, R14, Ris, Ri6, q and s can be combined with any of the groups described herein for one or more of the remainder of Y, Zi, Zz, R11, R12, R13, RM, RIS, R16, q and s, and may further be combined with any of the groups described herein for the Linker.

For a Degron of Formula Dl :

(1) In one embodiment, Zi is C(O) and Y is a bond. (2) In one embodiment, Zi is C(O) and Y is NH.

(3) in one embodiment, Zi is C(O) and Y is (CH 2 )o-6-0. In a further embodiment, Y is O.

(4) In one embodiment, Zi is C(O); Y r is a bond; and q and s are each 0.

(5) In one embodiment, Zi is C(O); Y is NH; and q and s are each 0.

(6) In one embodiment, Zi is C(Q); ' is (CH 2 )o-6-Q; and q and s are each 0. In a further embodiment, Y is O.

(7) In one embodiment, Zi is C(Q); Y' is a bond; and Ris is H.

(8) In one embodiment, Zi is C(O); Y is a bond; and Ris is H.

(9) In one embodiment, Zi is C(Q); Y' is NH; and Rn is H.

(10) In one embodiment, Zi is C(O); Y is NH; and Ris is H.

(11) In one embodiment, Zi is C(Q); Y is a bond; Ri3 is H; and Ris is H.

(12) In one embodiment, Zi is C(O); Y is NH; Ris is H; and Ris is H.

(13) In one embodiment, Zi is C(O); is (CHilo-e-O; and Ris is H. In a further embodiment, Y is O.

(14) In one embodiment, Zi is C(O); is (CH2I0-6-O; and R15 is H. In a further

embodiment, Y is O.

(15) In one embodiment, Zi is C(O); Y is (CH2I0-6-O; R13 is H; and Ris is H. In a

further embodiment, Y is O.

(16) In one embodiment, q and s are each 0; and Y " , Zi, R13, Ris, and Rie are each as defined in any of (1 ) --- (3) and (7) --- (15).

(17) In one embodiment, Zi is C(O) and Z 2 i s C(O).

(18) In one embodiment, Zi is C(O); Z2 is C(O); and Ris is H.

(19) In one embodiment, Zi is C(O) and Z2 is C(RI 3 ) 2,

(20) In one embodiment, Zi is C(O); Z 2 is C(Ri3) 2 ; and Ris is H.

(21) In one embodiment, Zi is C(O); Z 2 is C(Ri3) 2 ; R13 is H; and Ris is H.

In one embodiment, the Degron is of one of the following formulae:

or an enantiomer, diastereomer, or stereoisomer thereof, wherein Y, R l3 , RM, Ri6, q, and s are each as defined above m Formula Dl, and can be selected from any moieties or combinations thereof described above.

In one embodiment, the Degron is one of the preceding formulae Dla-DIT and R l3 is H.

In one embodiment the Degron is one of the preceding formulae Dla-Dll’ and R is Ci~ ( ' · alkyl. In one embodiment, R l3 is (Ή ·.

In one embodiment, Y is a bond, O, or NH. In one embodiment, Y is a bond. In one embodiment, Y is O. In one embodiment, Y is NH.

Linker

The Linker is a bond or a carbon chain that serves to link a Targeting Ligand with a Degron In one embodiment, the carbon chain optionally comprises one, two, three, or more heteroatoms selected from N, O, and S. In one embodiment, the carbon chain comprises only saturated chain carbon atoms. In one embodiment, the carbon chain optionally comprises two or more unsaturated chain carbon atoms (e.g., c=c or c~ºc). In one embodiment, one or more chain carbon atoms in the carbon chain are optionally substituted with one or more substituents (e.g., oxo, Ci-Co alkyl, C2-C6 alkenyl, Ci-Ce alkynyl, Ci-C 3 alkoxy, OH, halogen, NH2, NH(CI-C 3 alkyl), N(€I~C 3 alkyl)2, CN, C 3 -Cg cycloalkyl, heterocyclyl, phenyl, and heteroaryl).

In one embodiment, the Linker comprises at least 5 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises less than 25 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises less than 20 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 5, 7, 9, 11, 13, 15, 17, or 19 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 5, 7, 9, or 11 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 11, 13, 15, 17, or 19 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 11, 13, 15, 17, 19, 21, or 23 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 6, 8, 10, 12, 14, 16, 18, 20, 22, or 24 chain atoms f e.g., C, O, N, and S). In one embodiment, the Linker comprises 6, 8, 10, 12, 14, 16, 18, or 20 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 6, 8, 10, or 12 chain atoms (e.g., C, O, N, and S). In one embodiment, the Linker comprises 12, 14, 16, 18, or 20 chain atoms (e.g., C, O, N, and S).

In one embodiment, the Linker comprises from 11 to 19 chain atoms (e.g., C, Q, N, and S).

In one embodiment, the Linker is a carbon chain optionally substituted with non-bulky substituents (e.g., oxo, Ci-Ce alkyl, Cz-Ce alkenyl, Ci-Ce alkynyl, C1-C3 alkoxy, OH, halogen, Nth, NH(CI-C3 alkyl), MC i-C · alkyl)2, and CN). In one embodiment, the non-bulky substitution is located on the chain carbon atom proximal to the Degron (i.e., the carbon atom is separated from the carbon atom to which the Degron is bonded by at least 3, 4, or 5 chain atoms in the Linker). In one embodiment, the non-bulky substitution is located on the chain carbon atom proximal to the Targeting Ligand (i.e., the carbon atom is separated from the carbon atom to which the Degron is bonded by at least 3, 4, or 5 chain atoms in the Linker).

In one embodiment the Linker is of Formula L0:

or an enantiomer, diastereomer, or stereoisomer thereof, wherein

pi is an integer selected from 0 to 12;

p2 is an integer selected from 0 to 12;

p3 is an integer selected from 0 to 6;

each W is independently absent, CH2, O, S, NTT, or NR19;

Z-. is absent, ( ' (()). (CH 2 ) .j C(0)NH, Cl h. O, NTT, or NR19;

each R19 IS independently C1-C3 alkyl;

j is 1, 2, or 3; and

Q is absent, CTI2, C(O), or NHC(0)CH 2 ,

wherein the Linker is covalently bonded to a Degron via the ¾ next to Q, and covalently bonded to a Targeting Ligand via the next to Z 3 .

In one embodiment, the total number of chain atoms in the Linker is less than 30. In a further embodiment, the total number of chain atoms in the Linker is less than 20.

For a Linker of Formula L0:

In one embodiment, pi is an integer selected from 0 to 10.

In one embodiment, pi is an integer selected from 1 to 10.

In one embodiment, pi is selected from 1, 2, 3, 4, 5, and 6.

In one embodiment, pi is 0, 1, 3, or 5.

In one embodiment pi is 0, 1 , 2, or 3.

In one embodiment, pi is 0.

In one embodiment pi is 1.

In one embodiment, pi is 3.

In one embodiment. pi is 5.

In one embodiment, p2 is an integer selected from 0 to 10.

In one embodiment. p2 is selected from 0, 1, 2, 3, 4, 5, and 6.

In one embodiment, p2 is 0, 1, 2, or 3.

In one embodiment. p2 is 0.

In one embodiment, p2 is 1.

In one embodiment. p3 is an integer selected from 1 to 5.

In one embodiment, p3 is 2, 3, 4, or 5.

In one embodiment, p3 is 0, 1 , 2, or 3.

In one embodiment, p3 is 0.

In one embodiment, p3 is 1.

In one embodiment, p3 is 2.

In one embodiment, p3 is 3.

In one embodiment, p3 is 6.

In one embodiment, at least one W is CH 2 .

In one embodiment, at least one W is O.

In one embodiment, at least one W is S. In one embodiment, at least one W is NH.

in one embodiment, at least one W is NR19; and each R19 is independently C1-C3 alkyl selected from methyl, ethyl, and propyl

in one embodiment, each W is O.

In one embodiment, j is 1, 2, or 3.

In one embodiment, j is 1.

In one embodiment, j is 2.

In one embodiment, j is 3.

In one embodiment, j is 2 or 3.

In one embodiment, j is 1 or 2.

In one embodiment, Q is absent.

In one embodiment, Q is NHC(0)CH2.

In one embodiment Q is C(O).

In one embodiment, Q is CH2.

In one embodiment, Z3 is absent.

In one embodiment, Z3 is CH2

In one embodiment, Z3 is O.

In one embodiment, Z 3 is C(O).

In one embodiment, Z3 is (CH 2 )jC(0)NH

In one embodiment, Z 3 is NR19; and R19 is C1-C3 alkyl selected from methyl, ethyl, and propyl.

In one embodiment, pi is 1, 2, 3, or 4. In one embodiment, pi is 1. In one embodiment, pi is 2. In one embodiment, pi is 3 In one embodiment, pi is 4

In one embodiment, pi is 1 and Z3 is absent.

In one embodiment, pi is 1, Z3 is absent, and W is CH 2 .

In one embodiment, pi is 1, Z3 is absent, and p3 is 1.

In one embodiment, pi is 1, Z3 is absent, and p3 is 2.

In one embodiment, pi is 1, Z3 is absent, and p2 is 0.

In one embodiment, pi is 1, Z3 is absent, p3 is 2, and p2 is 0.

In one embodiment, pi is 1, Z3 is absent, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pi is 1, Z3 is absent, p3 is 2, p2 is 0, each W is O, and Q is absent In one embodiment, pl is 3 and Z3 is absent.

In one embodiment, pl is 3, Z3 is absent, and p3 is 2.

In one embodiment, pl is 3, Z3 is absent, and p2 is 0.

In one embodiment, pl is 3, Z is absent, p3 is 2, and p2 is 0.

In one embodiment, pl is 3, Z3 is absent, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pl is 3, Z 3 is absent, p3 is 2, p2 is 0, each W is O, and Q is absent In one embodiment, pl is 5 and Z3 is absent.

In one embodiment, pl is 5, Z 3 is absent, and p3 is 2.

In one embodiment, pl is 5, Z3 is absent, and p2 is 0.

In one embodiment, pl is 5, Z 3 is absent, p3 is 2, and p2 is 0.

In one embodiment, pl is 5, Z 3 is absent, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pi is 5, Z 3 is absent, p3 is 2, p2 is 0, each W is O, and Q is absent In one embodiment pl is 1 and Z3 is C(O).

In one embodiment, pi is 1, Z 3 is C(O), and p3 is 2.

In one embodiment pi is 1, Z3 is C(O), and p2 is 0.

In one embodiment, pl is 1, Z 3 is C(O), p3 is 2, and p2 is 0.

In one embodiment. pi is i, Z3 is C(O), p3 is 2, p2 is 0, and each W is O.

In one embodiment, pl is 1, Z3 is C(O), p3 is 2, p2 is 0, each W is O, and Q is absent. In one embodiment. pi is 3 and Z3 is C(O).

In one embodiment, pl is 3, Z3 is C(O), and p3 is 2.

In one embodiment. pi is 3, Z 3 i s C(O), and p2 is 0.

In one embodiment, pl is 3, Z3 is C(O), p3 is 2, and p2 is 0

In one embodiment. pi is 3, Z 3 i s C(O), p3 is 2, p2 is 0, and each W is O.

In one embodiment, pl is 3, Z3 is C(O), p3 is 2, p2 is 0, each W is O, and Q is absent. In one embodiment, pl is 5 and Z3 is CfO).

In one embodiment, pl is 5, Z3 is C(O), and p3 is 2.

In one embodiment, pl is 5, Z3 is C(O), and p2 is 0.

In one embodiment, pl is 5, Z3 is C(O), p3 is 2, and p2 is 0.

In one embodiment, pl is 5, Z3 is C(O), r3 is 2, p2 is 0, and each W is O.

In one embodiment, pl is 5, Z3 is C(O), p3 is 2, p2 is 0, each W is O, and Q is absent. In one embodiment, p2 is 0 and Q is absent. In one embodiment, p2 is 0; Q is absent; and each W is O.

in one embodiment, p2 is 0; Q is absent; and pi is 2-4.

In one embodiment, p2 is 0; Q is absent; and pi is 2.

In one embodiment, p2 is 0; Q is absent; and pi is 4.

In one embodiment, p2 is 0; Q is absent; and p3 is 2.

In one embodiment, p2 is 0; Q is absent; and Z 3 is C(O).

In one embodiment, p2 is 0; Q is absent; each W is O; and pi is 2-4.

In one embodiment, p2 is 0; Q is absent; each W is O; and pi is 2.

In one embodiment, p2 is 0; Q is absent; each W is O; and pi is 4.

In one embodiment, p2 is 0; Q is absent; each W is O; and p3 is 2.

In one embodiment, p2 is 0; Q is absent; each W is O; and Z¾ is C(O).

In one embodiment, p2 is 0; Q is absent; each W is O; p3 is 2; and Z3 is C(O) In one embodiment p3 is 3 and Z3 is absent

In one embodiment, p3 is 3, Z3 is absent, and pi is 0.

In one embodiment p3 is 3, Z 3 i s absent, pi is 0, and Q is absent

In one embodiment, p3 is 4 and Z 3 is absent

In one embodiment p3 is 4, Z 3 i s absent, and p i is 0.

In one embodiment, p3 is 4, Z3 is absent, pi is 0, and Q is absent

In one embodiment, p3 is 2, and Z3 is absent

In one embodiment, pi is 3 and Z3 is (CH 2 )jC(0)NH.

In one embodiment, pi is 3 and Z3 is (CH2)C(0)NH.

In one embodiment, pi is 3 and Z3 is (CH2) 2 C(0)NH.

In one embodiment, pi is 3 and Z3 is (CH2)3C(0)NH.

In one embodiment, pi is 3, Z3 is (CH2)jC(0)NH, and p3 is 2.

In one embodiment, pi is 3, Z3 is (CH2)C(0)NH, and p3 is 2.

In one embodiment, pi is 3, Z3 is (CH2)2C(0)NH, and p3 is 2.

In one embodiment, pi is 3, Z3 is (CH2)3C(0)NH, and p3 is 2.

In one embodiment, pi is 3, Z3 is (CH2)jC(0)NH, p3 is 2, and p2 is 0.

In one embodiment, pi is 3, Z3 is (CH2)C(0)NH, p3 is 2, and p2 is 0.

In one embodiment, pi is 3, Z3 is (CH2)2C(0)NH, p3 is 2, and p2 is 0.

In one embodiment, pi is 3, Z3 is (CH2.)3C(Q)NH, p3 is 2, and p2 is 0. In one embodiment, pi is 3, Zs is (CH2)jC(0)NH, p3 is 2, p2 is 0, and each W is O. in one embodiment, pi is 3, Z 3 i s (CH2)C(0)NH, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pi is 3, Zs is (CH2)2C(0)NH, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pi is 3, Z3 is (CH2)3C(0)NH, p3 is 2, p2 is 0, and each W is O.

In one embodiment, pi is 3, Z3 is (CH2.)jC(Q)NH, p3 is 2, p2 is 0, each W is O, and Q is absent.

In one embodiment, pi is 3, Z3 is (CH2.)C(0)NH, p3 is 2, p2 is 0, each W is O, and Q is absent.

In one embodiment, pi is 3, Z3 is (CH2.)2C(Q)NH, p3 is 2, p2 is 0, each W is O, and Q is absent.

In one embodiment, pi is 3, Z 3 is (CH2) 3 C(0)NH, p3 is 2, p2 is 0, each W is O, and Q is absent.

In one embodiment pi is 3 and Z3 is CH 2 C(0)NH.

In one embodiment, pi is 3, Z 3 is CH2C(0)NH, and Q is absent.

In one embodiment pi is 4 and Z3 is absent.

In one embodiment, pi is 4, Z 3 is absent, and p2 is 1.

In one embodiment, pi is 4, Z 3 is absent, p2 is 1, and Q is absent.

In one embodiment, pi is 4, Z 3 is absent, p2 is 1 , and p3 is 3.

In one embodiment, pi is 4, Z 3 i s absent, p2 is 1, p3 is 3, and Q is absent.

In one embodiment, pi is 3 and Z 3 is absent.

In one embodiment, pi is 3, Z 3 i s absent, and p3 is 3.

In one embodiment, pi is 3, Q is absent, and p3 is 3.

In one embodiment, pi is 4, Z 3 i s absent, and p3 is 3.

In one embodiment, pi is 4, Z3 is absent, p3 is 3, and Q is absent.

In one embodiment, pi is 4, Z3 is absent, p3 is 3, Q is absent, and p2 is 0.

In one embodiment, pi is 4, Z3 is absent, and Q is absent.

In one embodiment, pi is 3, Z3 is CH2.C(0)NH, and Q is absent.

In one embodiment, pi is 3, Z3 is CH2.C(0)N1-I, Q is absent, and p3 is 2.

In one embodiment, pi is 4, Q is absent, and p3 is 1.

In one embodiment, pi is 4, Q is absent, p3 is 1, and p2 is 0.

In one embodiment, pi is 4, Q is absent, and p3 is 3. In one embodiment, pi is 4, Q is absent, p3 is 3, and p2 is 0.

in one embodiment, pi is 2, Q is absent, p2 is 0, ZB is absent, and p3 is 6.

In one embodiment, the Linker-Targeting Ligand (TL) has the structure selected from Table L:

Table L

wherein Q, TL, pi, p3, and ] are each as described above.

Any one of the Degrons described herein can be covalently bound to any one of the Linkers described herein. Any one of the Targeting Ligands described herein can be covalently bound to any one of the Linkers described herein.

In one embodiment the present application relates to the Degron-Linker (DL), wherein the Degron is of Formula Dl, and the Linker is selected from Li - L7 In one embodiment, the Degron is of any one of Formulae D!a-Dlf , and the Linker is selected from LI --- L7. In one embodiment, the Degron is of any one of Formulae Dig-Dll’, and the Linker is selected from LI - L7. In one embodiment, the Degron is of any one of Formulae Dla-Dlf, and the Linker is LI, L2, or L3. In one embodiment, the Degron is of any one of Formulae Dig-Dll’, and the Linker is LI, L2, or L3. In one embodiment, the Degron is of any one of Formulae Dla-Dlf, and the Linker is L4, L5, L6, or L7. In one embodiment, the Degron is of any one of Formulae Dig- Dll’, and the Linker is L4, L5, L6, or L7. In one embodiment, the Degron is of Formula Dla or Dla’, and the Linker is LI, L2, or L3. In one embodiment, the Degron is of Formula Dig or Dig’, and the Linker is LI, L2, or L3. In one embodiment, the Degron is of Formula Dla or Dla’, and the Linker is L4, L5, L6, or L7. In one embodiment, the Degron is of Formula Dig or Dig’, and the Linker is L4, L5, L6, or L7. In one embodiment, the Degron is of Formula Dla or Dla’, and the Linker is 1,2. In one embodiment, the Degron is of Formula Dig or Dig’, and the Linker is 1,2.

In one embodiment, the Linker is designed and optimized based on SAR (structure- activity' relationship) and X-ray crystallography of the Targeting Ligand with regard to the location of atachment for the Linker.

In one embodiment the optimal Linker length and composition vary by the Targeting Ligand and can be estimated based upon X-ray structure of the Targeting Ligand bound to its target. Linker length and composition can be also modified to modulate metabolic stability and pharmacokinetic (PK) and pharmacodynamics (PD) parameters.

Some embodiments of present application relate to the bifunctional compounds having the following structures in Table A:

Some of the foregoing compounds can comprise one or more asymmetric centers, and thus can exist in various isomeric forms, e.g., stereoisomers and/or diastereomers. Accordingly, compounds of the application may be in the form of an individual enantiomer, diastereomer or geometric isomer, or may be in the form of a mixture of stereoisomers. In one embodiment, the compounds of the application are enantiopure compounds. In another embodiment, mixtures of stereoisomers or diastereomers are provided.

Furthermore, certain compounds, as described herein, may have one or more double bonds that can exist as either the Z or E isomer, unless otherwise indicated. The application additionally encompasses the compounds as individual Z/E isomers substantially free of other E/Z isomers and alternatively, as mixtures of various isomers.

In one embodiment, the present application relates to compounds that target proteins, such as CDK8 and/or CDK19 for degradation, which have numerous advantages over inhibitors of protein function (e.g., kinase activity) and can a) overcome resistance in certain cases; b) prolong the kinetics of drug effect by destroying the protein, thus requiring resynthesis of the protein even after the compound has been metabolized; e) target all functions of a protein at once rather than a specific catalytic activity or binding event; d) expand the number of drug targets by including all proteins that a ligand can be developed for, rather than proteins whose activity (e.g., kinase activity) can be affected by a small molecule inhibitor, antagonist or agonist; and e) have increased potency compared to inhibitors due to the possibility of the small molecule acting catalytically.

Some embodiments of the present application relate to degradation or loss of 30% to 100% of the target protein. Some embodiments relate to the loss of 50-100% of the target protein. Other embodiments relate to the loss of 75-95% of the targeted protein.

A bi functional compound of the present application (e.g., a bifunctional compound of any of the formulae described herein, or selected from any bifunctional compounds described herein) is capable of modulating (e.g., decreasing) the amount of a targeted protein (e.g, CDK8 and/or CDK19). A bifunctional compound of the present application (e.g, a bifunctional compound of any of the formulae described herein, or selected from any bifunctional compounds described herein) is also capable of degrading a targeted protein (e.g., CDK8 and/or CDK19) through the UPP pathway. Accordingly, a bifunctional compound of the present application (e.g., a bifunctional compound of any of the formulae described herein, or selected from any

bifunctional compounds described herein) is capable of treating or preventing a disease or disorder in which CDK8 and/or CDK19 plays a role. A bifunctional compound of the present application (e.g., a bifunctional compound of any of the formulae described herein, or selected from any bifunctional compounds described herein) is also capable of treating or preventing a disease or disorder in which CDK8 and/or CDK19 plays a role or in which CDK8 and/or CDK19 is deregulated f e.g., overexpressed).

Modulation of CDK8 and/or CDK19 through UPP-mediated degradation by a bifunctional compound of the application, such as those described herein, provides a novel approach to the treatment, prevention, or amelioration of diseases or disorders in which CDK8 and/or CDK19 plays a role including, but not limited to, cancer and metastasis, inflammation, arthritis, systemic lupus erthematosus, skin-related disorders, pulmonary disorders,

cardiovascular disease, ischemia, neurodegenerative disorders, liver disease, gastrointestinal disorders, viral and bacterial infections, central nervous system disorders, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, spinal cord injury', and peripheral neuropathy. Further, modulation of CDK8 and/or CDK19 through UPP-mediated degradation by a bifunctional compound of the application, such as those described herein, also provides a new paradigm for treating, preventing, or ameliorating diseases or disorders in which CDK8 and/or CDK! 9 is deregulated.

In one embodiment, a bifunctional compound of the present application (e.g., a bifunctional compound of any of the formulae described herein, or selected from any

bifunctional compounds described herein) is more efficacious in treating a disease or condition (e.g., cancer) than, or is capable of treating a disease or condition resistant to, the Targeting Ligand, when the Targeting Ligand is administered alone (i.e., not bonded to a Linker and a Degron). In one embodiment, a bifunctional compound of the present application (e.g., a bifunctional compound of any of the formulae described herein, or selected from any

bifunctional compounds described herein) is capable of modulating (e.g., decreasing) the amount of CDK8 and/or CDK19, and thus is useful in treating a disease or condition (e.g., cancer) in winch CDK8 and/or CDK19 plays a role.

In one embodiment, the bifunctional compound of the present application that is more efficacious in treating a disease or condition than, or is capable of treating a disease or condition resistant to, the Targeting Ligand, when the Targeting Ligand is administered alone (i.e., not bonded to a Linker and a Degron), is more potent in inhibiting the growth of cells (e.g., cancer cells) or decreasing the viability' of cells (e.g., cancer cells), than the Targeting Ligand, when the Targeting Ligand is administered alone (i.e., not bonded to a Linker and a Degron). In one embodiment, the bifunctional compound inhibits the growth of cells (e.g., cancer cells) or decreases the viability of cells (e.g., cancer cells) at an ICso that is lo wer than the ICso of the Targeting Ligand (when the Targeting Ligand is administered alone (i.e., not bonded to a Linker and a Degron)) for inhibiting the growth or decreasing the viability of the cells in one embodiment, the ICso of the bifunctional compound is at most 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 8%, 5%, 4%, 3%, 2%, 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 50%, 40%, 30%, 20%, 10%, 8%, 5%, 4%, 3%, 2%, 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 30%, 20%, 10%, 8%, 5%, 4%, 3%, 2%, 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 10%, 8%, 5%, 4%, 3%, 2%, 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 5%, 4%, 3%, 2%, 1%, 0.8%, 0 5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 2%, 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the ICso of the bifunctional compound is at most 1%, 0.8%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1% of the ICso of the Targeting Ligand. In one embodiment, the bifunctional compound inhibits the growth of cells (e.g., cancer cells) or decreases the viability of cells (e.g., cancer cells) at an Emax that is lower than the Emax of the Targeting Ligand (when the Targeting Ligand is administered alone (i.e., not bonded to a Linker and a Degron)) for inhibiting the growth or decreasing the viability of the cells. In one embodiment, the Emax of the bifunctional compound is at most 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 8%, 5%, 4%, 3%, 2%, or 1% of the Emax of the Targeting Ligand. In one embodiment, the Emax of the bifunctional compound is at most 50%, 40%, 30%, 20%, 10%, 8%, 5%, 4%, 3%, 2%, or 1% of the Emax of the Targeting Ligand. In one embodiment, the Emax of the bifunctional compound is at most 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, or 10% of the Emax of the Targeting Ligand.

In some embodiments, the inhibition or degradation of CDK8 and/or CDK19 activity is measured by ICso.

In some embodiments, the inhibition or degradation of CDK8 and/or CDK19 activity is measured by ECso. Potency of the inhibitor or degrader can he determined by ECso value. A compound with a lower ECso value, as determined under substantially similar conditions, is a more potent inhibitor or degrader relative to a compound with a higher ECso value in some embodiments, the substantially similar conditions comprise determining a CDK8- and/or CDK19-dependent phosphorylation level, in vitro or in vivo (e.g., m cells expressing a wild-type CDK8 and/or CDK19, a mutant CDK8 and/or CDK19, or a fragment of any thereof).

Potency of the inhibitor or degrader can also be determined by ICso value. A compound with a lower ICso value, as determined under substantially similar conditions, is a more potent inhibitor or degrader relative to a compound with a higher ICso value. In some embodiments, the substantially similar conditions comprise determining a CDK8- and/or CDK19-dependent phosphorylation level, in vitro or in vivo (e.g., m cells expressing a wild-type CDK8 and/or CDK19, a mutant CDK8 and/or CDK19, or a fragment of any thereof).

In one embodiment the bifunctional compounds of the present application are useful as anticancer agents, and thus may be useful in the treatment of cancer, by effecting tumor cell death or inhibiting the growth of tumor cells. In certain exemplary embodiments, the disclosed anticancer agents are useful in the treatment of cancers and other proliferative disorders, including, but not limited to breast cancer, cervical cancer, colon and rectal cancer, leukemia, lung cancer (e.g., non-small cell lung cancer), melanoma, multiple myeloma, non-Hodgkin’s lymphoma, ovarian cancer, pancreatic cancer, prostate cancer, gastric cancer, leukemias (e.g., myeloid, lymphocytic, myelocytic and lymphoblastic leukemias), malignant melanomas, and T- cell lymphoma

A“selective CDK 8 inhibitor or degrader” or“selective CDK19 inhibitor or degrader,” can be identified, for example, by comparing the ability of a compound to inhibit the kinase activity of or degrade CDK8 or CDK19 to its ability to inhibit or degrade the other members of the CDK kinase family or other kinases. For example, a substance may be assayed for its ability to inhibit CDK 8 kinase activity or degrade CDK8, as well as CDK1 CDK2, CDK4, CDK6, CDK?, CDK9, CDK1 1 , CDK12, CDK13 CDK14 and other kinases. In some embodiments, the selectivity can be identified by measuring the ECso or ICso of the compounds.

In some embodiments, the bifunctional compounds of the present application containing a Target Ligand inhibit or degrade CDK8 and/or CDK! 9 more selectively over other cyclin- dependent kinases and/or other kinases than the Target Ligand alone (i.e., a Target Ligand itself compared to the Target Ligand covalently bound to a Linker and a Degron). in certain embodiments, the bifunctional compounds of the application are about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90% or about 99% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 10%, about 20%, about 30%, about 40%, or about 50% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 20%, about 30%, about 40%, about 50% or about 60% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 30%, about 40%, about 50%, about 60% or about 70% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 40%, about 50%, about 60%, about 70%, or about 80% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 50%, about 60%, about 70%, about 80%, or about 90% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In certain embodiments, the bifunctional compounds of the application are about 60%, about 70%, about 80%, about 90%, or about 99% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone.

In other embodiments, the bifunctional compounds of the application are between about 10% and about 99% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 10% and about 30% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 20% and about 40% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 30% and about 50% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone in other embodiments, the bifunctional compounds of the application are between about 40% and about 60% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 50% and about 70% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 60% and about 80% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 70% and about 90% more selective at inhibiting or degrading CDK8 and/or CDK19 than the Target Ligand alone. In other embodiments, the bifunctional compounds of the application are between about 80% and about 99% more selective at inhibiting or degrading CDK8 and/or CDK! 9 than the Target Ligand alone.

In some embodiments, the compounds of the present application are selective over other kinases. As used herein,“selective”,“selective CDK8 inhibitor,”“selective CDK19 inhibitor,” “selective CDK8 degrader,”“selective CDK19 degrader,”“selective CDK8 compound,” or “selective CDK19 compound” refers to a compound, for example a bifunctional compound of the application, that effectively inhibits or degrades CDK8 and/or CDK! 9 kinase to a greater extent than any other kinase enzyme, particularly any enzyme from the Cyclic-dependent kinase family (e.g., CDK1, CDK2, CDK4, CDK6, CDK7, CDK9, CDK1 1 , CDK12 CDK13. CDK14. r/c. i

In certain embodiments, the compounds of the application are CDK8 and/or CDK19 inhibitors or degraders that exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100- fold selectivity over other kinases (e.g., CDK1, CDK2, CDK4, CDK6, CDK7, CDK9, CDK1 1 , CDK12, CDK13, CDK14, etc.). In various embodiments, the compounds of the application exhibit 1000-fold selectivity over other kinases.

In certain embodiments, the compounds of the application are CDK8 and/or CDK19 inhibitors or degraders that exhibit at least 2-fold, 3-fold, 5-fold, 10-fold, 25-fold, 50-fold or 100- fold selectivity over other cyc!in-dependent kinases f e.g., CDK1, CDK2, CDK4, CDK6, CDK7, CDK9, CDK1 1 , CDK12, CDK13, CDK14, etc. ). In various embodiments, the compounds of the application exhibit 1000-fold selectivity over other cyclin-dependent kinases. Definitions

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

The term "alkyl," as used herein, refers to saturated, straight or branched-chain hydrocarbon radicals containing, in certain embodiments, between one and six carbon atoms. Examples of C -Cs alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, neopentyl, and n-hexyl radicals.

The term "alkenyl," as used herein, denotes a monovalent group derived from a hydrocarbon moiety containing, in certain embodiments, from two to six carbon atoms having at least one carbon-carbon double bond. The double bond may or may not be the point of attachment to another group. Alkenyl groups include, but are not limited to, for example, etheny!, propenyl, butenyl, l-methyl-2-buten-l-yl and the like.

The term "alkoxy" refers to an -O-aikyl radical.

The terms "hal," "halo," and "halogen," as used herein, refer to an atom selected from fluorine, chlorine, bromine and iodine.

The term "aryl," as used herein, refers to a mono- or poly-cyclic carhoeychc ring system having one or more aromatic rings, fused or non-fused, including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, indenyl and the like.

The term "aralkyl," as used herein, refers to an alkyl residue attached to an aryl ring. Examples include, but are not limited to, benzyl, phenethyl and the like.

The term "cycloalkyl," as used herein, denotes a monovalent group derived from a monocyclic or polycyclic saturated or partially unsaturated carbocyclic ring compound.

Examples of Ci-Cg cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopentyl and cyclooctyl; and examples of C3-Ci2-cycloalkyl include, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1 ] heptyl, and bicyclo [2.2.2] octyl. Also contemplated is a monovalent group derived from a monocyclic or polycyclic carbocyclic ring compound having at least one carbon-carbon double bond by the removal of a single hydrogen atom. Examples of such groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cye!ooctenyl, and the like. The term "heteroaryl," as used herein, refers to a mono- or poly-cyclic (e.g., bi-, or tri cyclic or more) fused or non-fused, radical or ring system having at least one aromatic ring, having from five to ten ring atoms of which one ring atoms is selected from S, O, and N; zero, one, or two ring atoms are additional heteroatoms independently selected from S, O, and N; and the remaining ring atoms are carbon. Heteroaryl includes, but is not limited to, pyridinyl, pyrazinyi, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, tiuazolyi, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolmyl, benzimidazolyl, benzooxazolyl, quinoxalinyl, and the like.

The term "heteroaralkyl," as used herein, refers to an alkyl residue attached to a heteroaryl ring. Examples include, but are not limited to, pyridinyimethyl, pyrimidinyiethyl and the like.

The term“heterocyclyl,” or“heterocycloalkyl,” as used herein, refers to a non-aromatic 3-, 4-, 5-, 6- or 7-membered ring or a bi- or tri-cyclic group fused of non-fused system, where (i) each ring contains between one and three heteroatoms independently selected from oxygen, sulfur and nitrogen, (ii) each 5-membered ring has 0 to 1 double bonds and each 6-membered ring has 0 to 2 double bonds, (hi) the nitrogen and sulfur heteroatoms may optionally be oxidized, and (iv) the nitrogen heteroatom may optionally be quatemized. Representative heterocycloalkyl groups include, but are not limited to, [l,3]dioxo!ane, pyrrolidinyl, pyrazoimyl, pyrazolidinyi, imidazo!inyl, imidazolidinyl, piperidinyl, piperaziny!, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl.

The term "alkylamino" refers to a group having the structure -NH(Ci-Ci2 alkyl), e.g., - NH(CI-C6 alkyl), where C1-C12 alkyl is as previously defined.

The term " dialky latnino" refers to a group having the structure -N(Ci-Ci2 alky 1)2, e.g., - NH(CI-C6 alkyl), where C1-C12 alkyl is as previously defined.

The term "acyl" includes residues derived from acids, including but not limited to carboxylic acids, carbamic acids, carbonic acids, sulfonic acids, and phosphorous acids.

Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyis, aliphatic sulfinyis, aromatic phosphates and aliphatic phosphates. Examples of aliphatic carbonyls include, but are not limited to, acetyl, propionyl, 2-fluoroacetyl, butyryl, 2-hydroxy acety l, and the like. In accordance with the application, any of the aryls, substituted aryls, heteroaryls and substituted heteroaryls described herein, can be any aromatic group. Aromatic groups can be substituted or unsubstituted.

As described herein, compounds of the application and moieties present in the compounds may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the application. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted." In general, the term "substituted”, whether preceded by the term "optionally" or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. The terms "optionally substituted", "optionally substituted alkyl," "optionally substituted "optionally substituted alkenyl," "optionally substituted alkynyl", "optionally substituted cycloalkyl," "optionally substituted cycloalkenyl," "optionally substituted aryl", "optionally substituted heteroaryl," "optionally substituted aralkyl", "optionally substituted heteroaralkyl," "optionally substituted heterocycloalkyl," and any other optionally substituted group as used herein, refer to groups that are substituted or unsubstituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to:

-F, -Cl, -Br, -I, -OH, protected hydroxy, -NO2, -CN, -NH2, protected amino, -NH-C1-C12- alkyi, -NH-C2-Ci2-alkenyi, -NH-C2-Ci2-alkenyi, -NH -C3-Ci2-eycloalkyi,

-NH-aryl, -NH -heteroaryl, -NH -heterocycloalkyl, -dialkylamino, -diarylammo,

-diheteroarylamino, -O-Ci-Cn-alkyl, -Q-Cz-Ciz-alkenyl, -O-C2-C 12-alkenyl,

-0-C3-Ci2-cycloalkyl, -O-aryl, -O-heteroaryl, -O-heterocycloalkyl, -C(0)-Ci-Ci2-alkyl, -C(O)- C2.-Ci2-alkenyl, -C(0)-C 2 -Ci2-alkenyl, -C(0)-C3-Ci2-cycloalkyl, -C(0)-aryl, -C(0)-heteroaryl, -C(0)-heterocycloalkyl, -CONH2, -CONH-Ci-Cn-alkyl, -CONH-Cz-Ciz-alkenyl,

-CONH-C 2 -Ci2-alkenyl, -CONH-Cs-Cn-cycloalkyl, -CONH-aryl, -CONH-heteroaryl,

-CONH-heterocycloalkyl,-OC02-Ci-Ci2-alkyl, -OCOz-Cz-Ciz-alkenyl, -OCOz-Cz-Ciz-alkenyl, -OC02-C3-Ci2-cycloalkyl, -OC0 2 -aryl, -OCOz-heteroaryl, -OC0 2 -heterocycloalkyl, -OCONH2, -OCONH-Ci-Ci2-alkyl, -OCONH- C2-C12-alkenyl, -OCONH- C2-C12-alkenyl, -OCONH-C3-Ci2-cy oaIkyl, -OCONH-aryl, -OCONH-heteroaryl, -OCONH-heterocycloalkyl, -NHC(0)-Ci-Ci 2-alkyl, -NHC(0)-C2-Ci2-alkenyl, -NHC(0)-C2-Ci2-alkenyl,

-NHC(0)-C3-Ci2-cycloalkyl, -NHC(0)-aryl, -NHC(0)-heteroaryl, -NHC(0)-heterocvcloalkyl, -NHCCh-Ci -Ci 2-alkyl, -NHC02-C2-Ci2-alkeny 1, -NHC02-C2-Ci2-alkenyl,

-NHC02-C3-Ci2-cycloalkyl, -NHCOa-aryl, -NHC02-heteroaryl, -NHCO2- heterocycloalkyl, NHC(0)NH2, -NHC(0)NH-C 1 -Ci 2-alkyl, -NHC(0)NH-C2-Ci2-alkenyl,

-NHC(0)NH-C2-Ci2-alkenyl, -NHC(0)NH-C3-Ci2-cycloalkyl, -NHC(0)NH-aryl,

-NHC(0)NH-heteroaryl, NHC(0)NH-heterocycloalkyl, -NHC(S)NH2,

-NHC( S)NH-Ci-Ci2-alkyl, -NHC(S)NH-C2-Ci2-alkenyl,

-NHC(S)NH-C2-Ci2-alkenyl, -NHC(S)NH-C 3 -Ci 2-cycloalkyl, -NHC(S)NH-aryl,

-NHC( S)NH-heteroaryl, -NHC(S)NH-heterocycloalkyl, -NHC(NH)NH 2 ,

-NHC(NH)NH- Ci-C 12-alkyl, -NHC(NH)NH-C2-Ci2-alkenyl, -NHC(NH)NH-C2-Ci2-alkenyl, -NHC(NH)NH-C3-C 12-cycloalkyl, -NHC(NH)NH-aryl, -NHC(NH)NH-heteroaryl,

-NHC(NH)NHheterocycloalkyl, -NHC(NH)-Ci-Ci2-alkyl, -NHC(NH)-C2-Ci2-alkenyl,

-NHC(NH)-C 2 -Ci2-alkenyl, -NHC(NH)-C3-Ci 2 -cycloalkyl, -NHC(NH)-aryl,

-NHC(NH)-heteroaryl, -NHC(NH)-heterocycloalkyl, -C(NH)NH-Ci-Ci2-alkyl,

-C(NH)NH-C 2 -Ci2-alkenyl, -C(NH)NH-C 2 -Ci2-alkenyl, C(NH)NH-C3-Ci 2 -cycloalkyl,

-C(NH)NH-aryl, -C(NH)NH-heteroaiyl, -C(NH)NHheterocycloalkyl,

-S(0)-Ci-Ci 2 -alkyl,- S(0)-C 2 -Ci2-alkenyl,- S(0)-C 2 -Ci2-alkenyl,

-S(0)-C3-C l2 -cycloalkyl,- S(0)-aryl, -S(0)-heteroaryl, -S(0)-heterocycloalkyl -SO2NH2, -SC) 2 NH-Ci-Ci 2 -alkyl, -S0 2 NH-C 2 -Ci 2 -alkenyl, -S0 2 NH-C 2 -Ci2-alkenyl,

-S0 2 NH-C 3 -Ci 2 -cycloalky3, -S0 2 NH-aryl, -S0 2 NH-heteroaryl, -S() 2 M-I-heterocycloalkyl, -NHSO2-C i-C i 2 -alkyl, -NHS0 2 -C 2 -Ci 2 -alkenyl,- NHS0 2 -C 2 -Ci2-alkenyl,

-NHS0 2 -C3-Ci 2 -cycloalkyl, -NHS0 2 -aryl, -NHS0 2 -heteroaryl, -NHS0 2 -heterocycloalkyl, -CH2NH2, -CH2.SO2.CH3, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, -heterocycloalkyl, -C3-Ci2-cycloalkyl, polyalkoxyalkyl, polyalkoxy, -methoxymethoxy, -methoxyethoxy, -SH, -S-Ci-Ci2-alkyl, -S-C 2 -Ci 2 -alkenyl, -S-C 2 -Ci 2 -alkenyl, -S-C 3 -Ci2-cycloalkyl, -S-aryl,

-S-heteroaryl, -S-heteroeycloalkyl, or methylthiomethyl.

it is understood that the aryls, heteroaryls, alkyls, and the like can be substituted.

The term "cancer" includes, but is not limited to, the following cancers: epidermoid oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma, and teratoma; Lung: bronchogemc carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal, rectum; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), eholangioearcmoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma,

osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma,

meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma (serous cystadenocarcmoma, mucinous

cystadenocarcmoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgermmoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous ceil carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia (acute and chrome), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma) hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; medullary thyroid carcinoma, undifferentiated thyroid cancer, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary 7 thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma. Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above- identified conditions.

The term“CDK8" herein refers to cyclin-dependent kinase 8.

The term“CDK19” herein refers to cyclin-dependent kinase 19.

The term "subject" as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be referred to herein as a patient.

"Treat", "treating" and "treatment" refer to a method of alleviating or abating a disease and/or its attendant symptoms.

As used herein,“preventing” or“prevent” describes reducing or eliminating the onset of the symptoms or complications of the disease, condition or disorder.

The term“targeted protem(s)” is used interchangeably with“target protem(s)”, unless the context clearly dictates otherwise. In one embodiment, a“targeted protein” is CDK.

The term "subject" as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be referred to herein as a patient.

The terms“disease(s)”,“disorder(s)”, and“condition(s)” are used interchangeably , unless the context clearly dictates otherwise.

The term "therapeutically effective amount" of a bifunctional compound or

pharmaceutical composition of the application, as used herein, means a sufficient amount of the bifunctional compound or pharmaceutical composition so as to decrease the symptoms of a disorder in a subject. As is well understood in the medical arts a therapeutically effective amount of a bifunctional compound or pharmaceutical composition of this application will be at a reasonable benefit/nsk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present application will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory ' dose for any particular patient wall depend upon a variety of factors including the disorder being treated and the severity- of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.

As used herein, the term "pharmaceutically acceptable salt" refers to those salts of the compounds formed by the process of the present application which are, within the scope of sound medical judgment, suitable for use m contact with the tissues of humans and low'er animals without undue toxicity', irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, etal. describes pharmaceutically acceptable salts in detail in J.

Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the application, or separately by reacting the free base or acid function with a suitable acid or base.

Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts: salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate,

benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodeey!sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate,

palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, /7-toluenesulfonate, undecanoate, valerate salts, and the like. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternar ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl having from 1 to 6 carbon atoms, sulfonate and aiyl sulfonate.

As used herein, the term "pharmaceutically acceptable ester" refers to esters of the bifunctional compounds formed by the process of the present application which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.

The term "pharmaceutically acceptable prodrugs" as used herein, refers to those prodrugs of the bifunctional compounds formed by the process of the present application which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity', irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitteriomc forms, where possible, of the compounds of the present application. "Prodrug", as used herein, means a compound which is convertible in vivo by metabolic means (e.g., by hydrolysis) to afford any compound delineated by the formulae of the instant application.

Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzymology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). "Design and Application of Prodrugs, Textbook of Drug Design and Development, Chapter 5, 113-191 (1991 ); Bundgaard, et al, Journal of Drug Deliver Reviews, 8: 1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Deliver} Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer, "Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzymology," John Wiley and Sons, Ltd. (2002). This application also encompasses pharmaceutical compositions containing, and methods of treating disorders through administering, pharmaceutically acceptable prodrugs of bifunctional compounds of the application. For example, compounds of the application having free amino, amido, hydroxy or carboxylic groups can be converted into prodrugs. Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues is covalently joined through an amide or ester bond to a free ammo, hydroxy or carboxylic acid group of compounds of the application. The amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes 4-hydroxyproline, hydroxylysine, demosine, isodemosme, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methionine sulfone. Additional types of prodrugs are also encompassed. For instance, free carboxyl groups can be derivatized as amides or alkyl esters. Free hydroxy groups may be derivatized using groups including but not limited to hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxy carbonyls, as outlined in Advanced Drug Delivery Reviews, 1996, 19, 1 15. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers wherein the acyl group may be an alkyl ester, optionally substituted with groups including but not limited to ether, amine and carboxylic acid functionalities, or where the acyl group is an ammo acid ester as described above, are also encompassed. Prodrugs of this type are described in J Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including but not limited to ether, amine and carboxylic acid functionalities.

The application also provides for a pharmaceutical composition comprising a

therapeutically effective amount of a bifunctional compound of the application, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

In another aspect, the application provides a kit comprising a bifunctional compound capable of inhibiting the activity of or degrading CDK8 and/or CDK19 selected from one or more compounds disclosed herein, or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, optionally in combination with a second agent and instructions for use m treating cancer.

In another aspect, the application provides a method of synthesizing a bifunctional compound disclosed herein.

The synthesis of the bifunctional compounds of the application can be found herein and in the Examples below.

Other embodiments are a method of making a bifunctional compound of any of the formulae herein using any one, or combination of, reactions delineated herein. The method can include the use of one or more intermediates or chemical reagents delineated herein.

Another aspect is an isotopically labeled bifunctional compound of any of the formulae delineated herein. Such compounds have one or more isotope atoms which may or may not be radioactive (e.g., 3 H, 2 H, l4 C, 13 C, 1S F, 5 S, 32 P, 1/5 I, and m I) introduced into the bifunctional compound. Such compounds are useful for drug metabolism studies and diagnostics, as well as therapeutic applications.

A bifunctional compound of the application can be prepared as a pharmaceutically acceptable acid addition salt by reacting the free base form of the compound with a

pharmaceutically acceptable inorganic or organic acid. Alternatively, a pharmaceutically acceptable base addition salt of a bifunctional compound of the application can be prepared by reacting the free acid form of the bifunctional compound with a pharmaceutically acceptable inorganic or organic base.

Alternatively, the salt forms of the bifunctional compounds of the application can be prepared using salts of the starting materials or intermediates.

The free acid or free base forms of the bifunctional compounds of the application can be prepared from the corresponding base addition salt or acid addition salt from, respectively. For example, a bifunctional compound of the application in an acid addition salt form can be converted to the corresponding free base by treating with a suitable base (e.g., ammonium hydroxide solution, sodium hydroxide, and the like). A bifunctional compound of the application in a base addition salt form can be converted to the corresponding free acid by treating with a suitable acid (e.g., hydrochloric acid, etc.).

Prodrugs of the bifunctional compounds of the application can be prepared by methods known to those of ordinary skill in the art (e.g., for further details see Saulnier et ah, (1994), Bioorganic and Medicinal Chemistry Letters, Vol. 4, p. 1985). For example, appropriate prodrugs can be prepared by reacting a non-denvatized bifunctional compound of the application with a suitable carbamy fating agent f e.g., 1,1 -acyloxyalkylearbanochloridate, para-nitrophenyl carbonate, or the like).

Protected derivatives of the bifunctional compounds of the application can be made by means known to those of ordinary skill in the art. A detailed description of techniques applicable to the creation of protecting groups and their removal can be found in T. W. Greene, "Protecting Groups in Organic Chemistry", 3rd edition, John Wiley and Sons, Inc., 1999.

Compounds of the present application can be conveniently prepared, or formed during the process of the application, as solvates (e.g., hydrates). Hydrates of bifunctional compounds of the present application can be conveniently prepared by recrystallization from an

aqueous/organic solvent mixture, using organic solvents such as dioxin, tetrahydrofuran or methanol.

Acids and bases useful in the methods herein are known in the art. Acid catalysts are any acidic chemical, which can be inorganic (e.g., hydrochloric, sulfuric, nitric acids, aluminum trichloride) or organic (e.g., camphorsu!fomc acid, p-toluenesulfonic acid, acetic acid, ytterbium inflate) in nature. Acids are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions. Bases are any basic chemical, which can be inorganic (e.g., sodium bicarbonate, potassium hydroxide) or organic (e.g., triethylamine, pyridine) m nature. Bases are useful m either catalytic or stoichiometric amounts to facilitate chemical reactions.

Combinations of substituents and variables envisioned by this application are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds winch possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).

When any variable (e.g., Rw) occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with one or more Ri4 moieties, then R l4 at each occurrence is selected independently from the definition of RM. Also, combinations of substituents and/or variables are permissible, but only if such combinations result in stable compounds within a designated atom’s normal valency. In addition, some of the compounds of this application have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomerie mixtures, and cis- or trans- or E- or Z- double isomeric forms, and other stereoisomeric forms that may be defined, m terms of absolute stereochemistry, as (R)- or (S)-, or as (D)~ or (L)- for ammo acids. When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration unless the text so states; thus a carbon-carbon double bond depicted arbitrarily herein as trans may be cis, trans, or a mixture of the two in any proportion. All such isomeric forms of such compounds are expressly included in the present application.

Optical isomers may be prepared from their respective optically active precursors by the procedures described herein, or by resolving the racemic mixtures. The resolution can be carried out in the presence of a resolving agent, by chromatography or by repeated crystallization or by some combination of these techniques which are known to those skilled in the art. Further details regarding resolutions can be found in Jacques, et ah, Enantiomers, Racemates, and Resolutions (John Wiley & Sons, 1981).

“Isomerism” means compounds that have identical molecular formulae but differ in the sequence of bonding of their atoms or m the arrangement of their atoms in space. Isomers that differ in the arrangement of their atoms in space are termed“stereoisomers”. Stereoisomers that are not mirror images of one another are termed“diastereoisomers”, and stereoisomers that are non-superimposabie mirror images of each other are termed“enantiomers” or sometimes optical isomers. A mixture containing equal amounts of individual enantiomeric forms of opposite chirality is termed a“racemic mixture”.

A carbon atom bonded to four non-identical substituents is termed a“chiral center”.

“Chiral isomer” means a compound with at least one chiral center. Compounds with more than one chiral center may exist either as an individual diastereomer or as a mixture of diastereomers, termed“diastereomerie mixture”. When one chiral center is present, a stereoisomer may be characterized by the absolute configuration (R or S) of that chiral center. Absolute configuration refers to the arrangement in space of the substituents attached to the chiral center. The substituents attached to the chiral center under consideration are ranked in accordance with the Sequence Ride of Cahn, Ingold and Prelog. (Cahn et al., Angew. Chern.

Inter. Edit. 1966, 5, 385; errata 511; Cahn et al. , Angew. i ' hem. 1966, 78, 413; Cahn and Ingold, J. Chew. Soc. 1951 (London), 612; Cahn et al., Experientia 1956, 12, 81; Cahn, J. Chem. Educ. 1964, 41, 116).

“Geometric isomer” means the diastereomers that owe their existence to hindered rotation about double bonds. These configurations are differentiated in their names by the prefixes cis and trails, or Z and E, which indicate that the groups are on the same or opposite side of the double bond in the molecule according to the Cahn-Ingoid-Prelog rules.

Furthermore, the structures and other compounds discussed in this application include all atropic isomers thereof. “Atropic isomers” are a type of stereoisomer in which the atoms of two isomers are arranged differently in space. Atropic isomers owe their existence to a restricted rotation caused by hindrance of rotation of large groups about a central bond. Such atropic isomers typically exist as a mixture, however as a result of recent advances in chromatography techniques; it has been possible to separate mixtures of two atropic isomers in select cases.

“Tautomer” is one of two or more structural isomers that exist in equilibrium and is readily converted from one isomeric form to another. This conversion results in the formal migration of a hydrogen atom accompanied by a switch of adjacent conjugated double bonds. Tautomers exist as a mixture of a tautomeric set in solution. In solid form, usually one tautomer predominates. In solutions where tautomenzation is possible, a chemical equilibrium of the tautomers will be reached. The exact ratio of the tautomers depends on several factors, including temperature, solvent and pH. The concept of tautomers that are interconvertable by

tautomers zati on s is called tautomerism.

Of the various types of tautomerism that are possible, two are commonly observed. In keto-enol tautomerism a simultaneous shift of electrons and a hydrogen atom occurs. Ring-chain tautomerism arises as a result of the aldehyde group (-CHO) in a sugar chain molecule reacting with one of the hydroxy groups (-OH) in the same molecule to give it a cyclic (ring-shaped) form as exhibited by glucose. Common tautomeric pairs are: ketone-enol, amide-nitrile, lactam- lactim, amide-imidic acid tautomerism in heterocyclic rings (e.g, in nucleobases such as guanine, thymine and cytosine), amine-enannne and enamine-enannne. The compounds of this application may also be represented in multiple tautomeric forms, in such instances, the application expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the application expressly includes all such reaction products).

in the present application, the structural formula of the bifunctional compound represents a certain isomer for convenience in some cases, but the present application includes all isomers, such as geometrical isomers, optical isomers based on an asymmetrical carbon, stereoisomers, tautomers, and the like. In the present specification, the structural formula of the compound represents a certain isomer for convenience m some cases, but the present application includes all isomers, such as geometrical isomers, optical isomers based on an asymmetrical carbon, stereoisomers, tautomers, and the like.

Additionally, the compounds of the present application, for example, the salts of the bifunctional compounds, can exist in either hydrated or unhydrated (the anhydrous) form or as solvates with other solvent molecules. Non-limiting examples of hydrates include

monohydrates, dihydrates, etc. Non-limiting examples of solvates include ethanol solvates, acetone solvates, etc.

“Solvate” means solvent addition forms that contain either stoichiometric or non- stoichiometric amounts of solvent. Some compounds have a tendency to trap a fixed molar ratio of solvent molecules in the crystalline solid state, thus forming a solvate. If the solvent is water the solvate formed is a hydrate; and if the solvent is alcohol, the solvate formed is an alcoholate. Hydrates are formed by the combination of one or more molecules of water with one molecule of the substance in which the water retains its molecular state as H2O.

The synthesized bifunctional compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid

chromatography, or recrystallization. As can be appreciated by the skilled artisan, further methods of synthesizing the bifunctional compounds of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired bridged macrocyclic products of the present application. Sy nthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wilts, Protective Groups m Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.

The compounds of this application may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

The compounds of the application are defined herein by their chemical structures and/or chemical names. Where a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and chemical name conflict, the chemical structure is determinative of the compound's identity.

The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

Method of Synthesizing the Compounds

Compounds of the present application can be prepared m a variety of ways using commercially available starting materials, compounds known in the literature, or from readily prepared intermediates, by employing standard synthetic methods and procedures either known to those skilled in the art, or which will be apparent to the skilled artisan in light of the teachings herein. Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be obtained from the relevant scientific literature or from standard textbooks m the field. Although not limited to any one or several sources, classic texts such as Smith, M. B., March, J., March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure , 5 th edition, John Wiley & Sons: Ne ' York, 2001; and Greene, T.W., Wuts, P.G. M., Protective Groups in Organic Synthesis, 3 rd edition, John Wiley & Sons: New York, 1999, incorporated by reference herein, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of compounds of the present application. The processes generally provide the desired final compound at or near the end of the ov erall process, although it may be desirable m certain instances to further convert the compound to a pharmaceutically acceptable salt, ester or prodrug thereof. Suitable synthetic routes are depicted in the schemes below.

Those skilled in the art will recognize if a stereocenter exists in the compounds disclosed herein. Accordingly, the present application includes both possible stereoisomers (unless specified in the synthesis) and includes not only racemic compounds but the individual enantiomers and/or diastereomers as well. When a compound is desired as a single enantiomer or diastereomer, it may be obtained by stereospecific synthesis or by resolution of the final product or any convenient intermediate. Resolution of the final product, an intermediate, or a starting material may be affected by any suitable method known in the art. See, for example, "Stereochemistry' of Organic Compounds" by E. L. Eliel, S. H. Widen, and L. N. Mander (Wiley- lnterscience, 1994).

The compounds of the present application can be prepared in a number of ways well known to those skilled in the art of organic synthesis. By way of example, compounds of the present application can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or variations thereon as appreciated by those skilled in the art. Preferred methods include but are not limited to those methods described below.

Compounds of the present application can be synthesized by following the steps outlined in General Schemes 1-4 which comprise different sequences of assembling intermediates.

Starting materials are either commercially available or made by known procedures in the reported literature or as illustrated.

Targeting Ligands of the present application can be synthesized by following the steps outlined in General Schemes 5 and 6 which comprise different sequences of assembling intermediates. Starting materials are either commercially available or made by known procedures in the reported literature or as illustrated. General Scheme 1: Synthesis of Thalidomide- based Degraders - Amides and Esters

wherein R13, RM, R15, Ri6, Z2, W, pi, q, and s are as defined herein above.

The general way of preparing representative compounds of the present application {i.e., Compound of Formula (I) shown above) using intermediates la, lb, lc, Id, le, If, and Ig is outlined in General Scheme 1 Reaction of glutarinnde (or d-valerolactam) derivatives la with phthalic anhydride derivatives lb in the presence of a base, i.e., diisopropylethylamine (DIPEA), and in a solvent, i.e., dimethy!formamide (DMF), provided intermediates lc. Reaction of esters Id with lc provided intermediates le. Deprotection of le in the presence of TFA in a solvent, i.e., dichloromethane (DCM) or methanol (MeOFI), provided If. Coupling of If and Target Ligands Ig, wherein“X” is \1 i(C! 1 H. under standard peptide coupling conditions using a coupling reagent, i.e., 1 -ethyl-3 -(3 -dimethylaminopropyl) carbodiimide (EDC) and

hydroxybenzotriazole, in a solvent, i.e., DCM or DMF, provided bifunctional compound of Formula (I).

Alternatively, glutarinnde derivative la may be replaced with the corresponding d-!actam derivative, e.g., d-valerolactam, to provide bifunctional compounds including degrons of Formulae Dig and Dig’.

Alternatively, targeting ligands Ig, wherein“X” is OH, may be coupled to intermediates If via known esterification conditions, e.g., Fischer esterification conditions by treatment with acid, Yamaguchi esterification conditions via conversion of the acid to an appropriate anhydride, or Steglich esterification conditions by treatment with a coupling agent such as

dicyclohexyl carbodiimide. General Scheme 2: Synthesis of Thalidomide- based Degraders - Triazoles

wherein R13, R14, Ris, Ri6, Z2, W, pi , q, and s are as defined herein above.

A general way of preparing representative compounds of the present application (i.e.,

Compound of Formula (II) shown above) using intermediates la, lb, lc, lh, li, lj, and Ik is outlined in General Scheme 2. Intermediates lc may be prepared according to General Scheme 1, and subsequently coupled to amines lh under the conditions provided in General Scheme 1 to provide intermediates li. The aldehyde of intermediate li may be converted to the terminal alkyne of intermediate lj under typical Corey-Fuchs homologation conditions, i.e., treatment with carbon tetrahromide in the presence of triphenylphosphirie, followed by treatment of the resulting 1,1 -dibromoolefin with «-butyllithium. Alternatively, amines lh may be protected with suitable protecting groups, subjected to Corey-Fuchs conditions, and further deprotected prior to coupling with lc. Alkyne lj may be coupled to Targeting Ligands Ik, wherein X is Ns, under standard Click reaction conditions, i.e., by treatment with a copper catalyst.

General

A general way of preparing representative compounds of the present application (i.e.. Compound of Formula (III) shown above) usmg intermediates la, lb, le, lh, li, 11, and lm is outlined in General Scheme 3. Intermediates lc may be prepared according to General Scheme 1, and subsequently coupled to amines lb under the conditions provided in General Scheme 1 to provide intermediates li. Olefination under standard Wittig conditions may afford the corresponding olefin (not shown) and subsequent treatment with bromine may provide dibromide 11. Targeting ligand lm may be prepared in a similar manner, i.e., under Mitsunobu conditions with ethanoiamine, ethyienediamine, or derivatives thereof, as shown in the examples below. Alternatively, ethanoiamine or ethyienediamine moieties may be installed via reductive animation between Targeting Ligand-NEb and O-protected 2-hydroxyacetaldehyde or TV- protected ammoaeetaldehyde, respectively. Under conditions effective to encourage substitution, e.g., in the presence of a base catalyst, reaction between Target Ligand lm and dibromide 11 may afford compounds of Formula (III).

It should be understood that the steps in any of the schemes disclosed herein may be performed in a different order. For example, the Wittig olefination of li may instead be performed on aldehyde lb.

General Scheme 4: Synthesis of Thalidomide-based Degraders - Heterocycles (b)

wherein R13, Ru, Ris, R16, Z2, W, pi , q, and s are as defined herein above.

A general way of preparing representative compounds of the present application (i.e., Compound of Formula (IV) shown above) using intermediates la, lb, lc, In, lo, Ip, and Iq is outlined in General Scheme 4. Intermediates lc may be prepared according to General Scheme 1, and subsequently coupled to amines In under the conditions provided in General Scheme 1 to provide intermediates lo. An example amine In may be prepared by polymerizing ethylene oxide with L-protected ethanoiamine, wherein W is O, followed by /V-deprotection. The alcohol of intermediates lo may be converted to a suitable leaving group, such as a tosylate (not shown). The resulting tosylate may substituted with azide and the azide compound (not shown) may further be reduced under standard Staudmger conditions, e.g., treatment with phosphine, to afford amine intermediates Ip. Targeting Ligand Iq may be prepared, for example, via two sequential reductive animation events between Targeting Ligand-Nth and an (^-protected 2- hydroxyacetaldehyde, i.e., two separate reductive animation reactions, each with different O- protecting groups that are cleavable under different conditions, e.g., -OBn and -OtBu. O- deprotection, followed by conversion of the resulting aicohol(s) to a suitable leaving group, such as tosylate. The resulting tosylate may undergo substitution with amine Ip, in the presence of an acid or base catalyst, to provide compounds of Formula (IV).

reductive amination alkylation, or on

ketone vinyi trif!ate

f (or d)

deprotection formation

In General Scheme 5, compound (a) may be obtained commercially as a single stereoisomer, and as either saturated or unsaturated as indicated.

Step i) Compound (a) may be protected as ketal (b), for example, with a suitable diol or thiol in the presence of an acid, e.g., ethylene glycol and tosylic acid. Ketal (b) may be subjected to Mitsunobu conditions (e.g. , a phosphine such as tnphenyiphosphine; an azodicarboxylate such as diethylazodicarboxylate; and a nucleophile such as an azide, e.g., diphenyl phosphoryl azide) to form azide (e). Other nucleophiles may be readily envisioned.

Step ii) Azide (c) may undergo a Click reaction in the presence of a suitable alkyne, such as TMS-acetylene, and a copper catalyst. Azide (e) may also be used to prepare various amino- substituted intermediates. For example, saturated azide (c) may be hydrogenated under standard hydrogenation conditions, e.g., H 2 and Pd/C. Unsaturated azide (c) may similarly be reduced under Staudinger conditions to provide amine (e), i.e., by treatment with a phosphine such as trimethylphosphine. Amine (e) may undergo reductive amination and/or alkylation. For example, reductive amination conditions may include inline formation with a suitable aldehyde, e.g., formaldehyde, or ketone, followed by treatment with a reducing agent such as sodium borohydride. For example, alkylation conditions may include treating amine (e), in the presence of a base, with a suitable alkylating agent such as an alkyl halide, e.g., ethyl iodide, 1,4- dibromoethane, or 2-bromoethyi ether. Alternatively, amine (e) may undergo mono-protection prior to alkylation, for example, as the boc amine, by treatment with boc anhydride.

Deprotection of a boc amine may be afforded by treatment with an acid, such as trifluoroacetic acid. For example, deprotection may be carried out on compound (i) or (j).

Step hi) The ketones of amine (!) or triazole (d) may be deprotected by treatment with a suitable acid such as tosylic acid and subsequently converted to the corresponding vinyl inflate (h). For example, inflate (h) may be provided by treatment of ketone (g) with a non- nucleophilic base, such as KHMDS, and suitable electrophilic triflate source, such as iV-pheny bis(trifluoromethanesulfonimide) or triflic anhydride.

Step iv) Triflate (h) may be coupled to a variety of heteraryl groups (“HET”). For example, triflate (h) may be contacted with a HET-boronic acid or HET-boronate ester in the presence of a suitable palladium catalyst, i.e., Suzuki cross-coupling. Other cross-coupling reactions are readily envisioned, for example contacting triflate (h) with a HET-Snfalkylfl reagent in the presence of a palladium catalyst, i.e . Stifle cross-coupling. Compound (i) may further be reduced to afford compound (j), for example, by treatment with a dhmide reducing agent, such as potassium azodicarboxylate.

In General Scheme 6, compound (m) may be obtained commercially as a single stereoisomer, and as either saturated or unsaturated as indicated by the figure.

Compound (m) may be subjected to cross-coupling conditions m order to install a suitable heteroaryl group (“HET”), and the resulting compound (n) may further be reduced. Each step may be carried out as described above in General Scheme 5.

Biological Assays

The biological activities of the compounds of the present application can be assessed through a variety of methods known in the art. For example, a kinase competitive binding assay may be used to determine the activity of the compounds of the present application to bind to CDS and/or CD 19. In the competitive binding assay, a reporting ligand capable of binding to CDS and/or CD 19, such as a fluoro- or radio-labeled compound, is incubated with CDS and/or CD 19 in the absence of a compound of the present application or with increasing concentrations of the compound. The changes in the signal produced by the reporting ligand when the compound is present relative when the compound is absent are recorded, which are calculated to provide the ICso of the compound. Methods of the i-

ln one aspect, the present application provides a method of modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cyclin-dependent kinase, such as CDK8 and/or CDK! 9). The method comprises administering to a subject in need thereof an effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

In one aspect the present application provides a method of modulating (e.g., inhibiting or decreasing the amount of) CDK8 and/or CDK19. The method comprises administering to a subject in need thereof an effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

In some embodiments, the inhibition or degradation of CDK8 and/or CDK19 activity is measured by ICso. In some embodiments, the inhibition or degradation of CDK8 and/or CDK19 activity is measured by ECso.

A bifunctional compound of the present application (e.g., a compound of any of the formulae described herein, or selected from any compounds described herein) is capable of treating or preventing a disease or disorder in which CDK8 and/or CDK 19 plays a role or in which CDK8 and/or CDK 19 is deregulated (e.g., overexpressed).

In one aspect, the present application provides a method of treating or preventing a disease responsive to modulation of CDK 8 and/or CDK! 9. The method comprises administering to a subject in need thereof an effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

In one aspect, the present application provides a method of treating or preventing a disease. The method comprises administering to a subject m need thereof an effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application. In one aspect, the disease is a kinase (e.g., CDK8 and/or CDK 19) mediated disorder. In one aspect, the disease is a proliferative disease (e.g., a proliferative disease m which CDK8 and/or CDK 19 plays a role). In one aspect, the present application provides a method of treating or preventing cancer m a subject, wherein the cell of the cancer comprises an activated CDK8 and/or activated CDK19 or wherein the subject is identified as being in need of inhibition of CDK8 and/or CDK! 9 for the treatment or prevention of cancer. The method comprises administering to the subject an effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

In one embodiment, the disease ( e.g ., cancer) is mediated by CDK8 and/or CDK19 ( e.g ., CDK8 and/or CDK19 plays a role m the initiation or development of the disease).

In one embodiment, the CDK8 activation is selected from mutation of CDK8, amplification of CDK8, overexpression of CDK8, and ligand mediated activation of CDK8.

In one embodiment, the CDK19 activation is selected from mutation of CDK19, amplification of CDK19, overexpression of CDK19, and ligand mediated activation of CDK19.

In one embodiment, the present application provides a method of treating or preventing any of the diseases, disorders, and conditions described herein, wherein the subject is a human.

In one embodiment, the application provides a method of treating. In one embodiment, the application provides a method of preventing.

As inhibitors or degraders of CDK8 and/or CDK19, the compounds and compositions of this application are particularly useful for treating or lessening the severity of a disease, condition, or disorder where a protein kinase is implicated in the disease, condition, or disorder. In one embodiment, the present application provides a method for treating or lessening the severity of a disease, condition, or disorder where a protein kinase is implicated m the disease state. In one embodiment, the present application provides a method for treating or lessening the severity of a kinase disease, condition, or disorder where inhibition of enzymatic activity is implicated in the treatment of the disease. In one embodiment, the present application provides a method for treating or lessening the severity of a disease, condition, or disorder with compounds that inhibit enzymatic activity or degrade the enzymes by binding to the protein kmase. In one embodiment, the present application provides a method for treating or lessening the severity of a kinase disease, condition, or disorder by inhibiting enzymatic activity of the kmase or degrading the kinase with a protein kmase inhibitor or degrader.

In one embodiment, the disease or disorder is cancer or a proliferation disease. In one embodiment, the disease or disorder is lung cancer, colon cancer, breast cancer, prostate cancer, liver cancer, pancreas cancer, brain cancer, kidney cancer, ovarian cancer, stomach cancer, skin cancer, bone cancer, gastric cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, hepatocellular carcinoma, papillary renal carcinoma, head and neck squamous cell carcinoma, leukemias, lymphomas, myelomas, or solid tumors.

In one embodiment, the disease or disorder is inflammation, arthritis, rheumatoid arthritis, spondyiarthropathies, gouty arthritis, osteoarthritis, juvenile arthritis, and other arthritic conditions, systemic lupus erthematosus (SLE), skin-related conditions, psoriasis, eczema, bums, dermatitis, neuroinflammation, allergy, pain, neuropathic pain, fever, pulmonary disorders, lung inflammation, adult respiratory distress syndrome, pulmonary sarcoisosis, asthma, silicosis, chronic pulmonary inflammatory disease, and chronic obstructive pulmonary disease (COPD), cardiovascular disease, arteriosclerosis, myocardial infarction (including post-myocardial infarction indications), thrombosis, congestive heart failure, cardiac reperfusion injury, as well as complications associated with hypertension and/or heart failure such as vascular organ damage, restenosis, cardiomyopathy, stroke including ischemic and hemorrhagic stroke, reperfusion injury, renal reperfusion injury, ischemia including stroke and brain ischemia, and ischemia resulting from cardiac/coronary' bypass, neurodegenerative disorders, liver disease and nephritis, gastrointestinal conditions, inflammatory' bowel disease, Crohn's disease, gastritis, irritable bowel syndrome, ulcerative colitis, ulcerative diseases, gastric ulcers, viral and bacterial infections, sepsis, septic shock, gram negative sepsis, malaria, meningitis, HIV infection, opportunistic infections, cachexia secondary to infection or malignancy, cachexia secondary to acquired immune deficiency syndrome (AIDS), AIDS, ARC (AIDS related complex), pneumonia, herpes virus, myalgias due to infection, influenza, autoimmune disease, graft vs. host reaction and allograft rejections, treatment of bone resorption diseases, osteoporosis, multiple sclerosis, cancer, leukemia, lymphoma, colorectal cancer, brain cancer, bone cancer, epithelial call-derived neoplasia (epithelial carcinoma), basal cell carcinoma, adenocarcinoma,

gastrointestinal cancer, lip cancer, mouth cancer, esophageal cancer, small bowel cancer, stomach cancer, colon cancer, liver cancer, bladder cancer, pancreas cancer, ovarian cancer, cervical cancer, lung cancer, breast cancer, skin cancer, squamous cell and/or basal cell cancers, prostate cancer, renal cell carcinoma, and other known cancers that affect epithelial cells throughout the body, chronic myelogenous leukemia (CIVIL), acute myeloid leukemia (AML) and acute promyelocytic leukemia (APL), angiogenesis including neoplasia, metastasis, central nervous system disorders, central nervous system disorders having an inflammatory or apoptotic component, Alzheimer's disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, spinal cord injury, peripheral neuropathy, or B-Cell Lymphoma.

In one embodiment, the disease or disorder is inflammation, arthritis, rheumatoid arthritis, spondylarthropathies, gouty arthritis, osteoarthritis, juvenile arthritis, and other arthritic conditions, systemic lupus erthematosus (SLE), skin-related conditions, psoriasis, eczema, dermatitis, pain, pulmonary disorders, lung inflammation, adult respiratory distress syndrome, pulmonary sarcoisosis, asthma, chronic pulmonary inflammatory disease, and chronic obstructive pulmonary disease (COPD), cardiovascular disease, arteriosclerosis, myocardial infarction (including post-myocardial infarction indications), congestive heart failure, cardiac reperfusion injury', inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome, leukemia, or lymphoma.

In one embodiment, the disease or disorder is selected from autoimmune diseases, inflammatory' diseases, proliferative and hyperproliferative diseases, immunologically-mediated diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cardiovascular diseases, hormone related diseases, allergies, asthma, and Alzheimer's disease. In one embodiment, the disease or disorder is selected from a proliferative disorder and a neurodegenerative disorder.

In one embodiment, the disease or disorder is characterized by excessive or abnormal cell proliferation. Such diseases include, but are not limited to, a proliferative or hyperproliferative disease, and a neurodegenerative disease. Examples of proliferative and hyperproliferative diseases include, without limitation, cancer.

The term’’cancer" includes, but is not limited to, the following cancers: breast; ovary; cervix; prostate; testis, genitourinary tract; esophagus; larynx, glioblastoma; neuroblastoma; stomach; skin, keratoacanthoma; lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung adenocarcinoma; bone; colon; colorectal; adenoma; pancreas, adenocarcinoma; thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma; seminoma;

melanoma; sarcoma; bladder carcinoma; liver carcinoma and biliar passages; kidney carcinoma; myeloid disorders; lymphoid disorders, Hodgkin's, hairy cells; buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx; small intestine; coionrectum, large intestine, rectum, brain and central nervous system; chronic myeloid leukemia (CML), and leukemia. The term’’cancer" includes, but is not limited to, the following cancers: myeloma, lymphoma, or a cancer selected from gastric, renal, or and the following cancers: head and neck, oropharangeal, non-small cell lung cancer (NSCLC), endometrial, hepatocarcinoma, Non-Hodgkins lymphoma, and pulmonary.

The term "cancer" also refers to any cancer caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, lymphomas and the like. For example, cancers include, but are not limited to, mesothelioma, leukemias and lymphomas such as cutaneous T-cell lymphomas (CTCL), noncutaneous peripheral T-cell lymphomas, lymphomas associated with human T-cell lymphotrophic virus (HTLV) such as adult T-cell leukemia/lymphoma (ATLL), B-cell lymphoma, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, acute myelogenous leukemia, lymphomas, and multiple myeloma, non-Hodgkin lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), Hodgkin’s lymphoma, Burkitt lymphoma, adult T-cell leukemia lymphoma, acute-myeloid leukemia (AML), chronic myeloid leukemia (CML), or hepatocellular carcinoma. Further examples include myelodisplastic syndrome, childhood solid tumors such as brain tumors, neuroblastoma, retinoblastoma, Wilms' tumor, bone tumors, and soft-tissue sarcomas, common solid tumors of adults such as head and neck cancers (e.g., oral, laryngeal, nasopharyngeal and esophageal), genitourinary cancers (e.g., prostate, bladder, renal, uterme, ovarian, testicular), lung cancer (e.g., small-cell and non-small cell), breast cancer, pancreatic cancer, melanoma and other skin cancers, stomach cancer, brain tumors, tumors related to Gorlin's syndrome (e.g , medulloblastoma, meningioma, etc.), and liver cancer.

Additional exemplary forms of cancer winch may be treated by the subject compounds include, but are not limited to, cancer of skeletal or smooth muscle, stomach cancer, cancer of the small intestine, rectum carcinoma, cancer of the salivary gland, endometrial cancer, adrenal cancer, anal cancer, rectal cancer, parathyroid cancer, and pituitary cancer.

Cancer may also include colon carcinoma, famitiary adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, or melanoma. Further, cancers include, but are not limited to, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, thyroid cancer (medullary and papillary thyroid carcinoma), renal carcinoma, kidney parenchyma carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, testis carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, gall bladder carcinoma, bronchial carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroidea melanoma, seminoma, rhabdomyosarcoma, cramopharyngeoma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma, and piasmocytoma.

Cancer may also include colorectal, thyroid, breast, and lung cancer; and

myeloproliferative disorders, such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, chronic myelogenous leukemia, chronic myelomonocytic leukemia, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, and systemic mast cell disease. In one embodiment, the compounds of this application are useful for treating hematopoietic disorders, in particular, acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia, and acute lymphocytic leukemia (ALL).

Examples of neurodegenerative diseases include, without limitation,

Adrenoleukodystrophy (AID), Alexander's disease, Alper’s disease, Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's Disease), Ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjogren-Batten disease). Bovine spongiform encephalopathy (BSE), Canavan disease, Cockayne syndrome, Corticobasal degeneration, Creutzfeldt- Jakob disease, Familial fatal insomnia. Frontotemporal lobar degeneration, Huntington's disease, HIV- associated dementia, Kennedy's disease, Krabbe's disease, Lewy body dementia,

Neuroborreliosis, Machado-Joseph disease (Spinocerebellar ataxia type 3), Multiple System Atrophy, Multiple sclerosis, Narcolepsy, Niemann Pick disease, Parkinson's disease, Pelizaeus- Merzbacher Disease, Pick's disease, Primary lateral sclerosis, Prion diseases, Progressive Supranuclear Palsy, Refsum's disease, Sandhoff disease, Schilders disease, Subacute combined degeneration of spinal cord secondary to Pernicious Anaemia, Spielmeyer-Vogt-Sjogren-Batten disease (also known as Batten disease), Spinocerebellar ataxia (multiple types with varying characteristics), Spinal muscular atrophy, Steele-Richardson-Olszewski disease, Tabes dorsalis, and Toxic encephalopathy.

In one aspect, the present application also provides a method of treating or preventing cell proliferative disorders such as hyperplasias, dy splasias, or pre-cancerous lesions. Dysplasia is the earliest form of pre-cancerous lesion recognizable in a biopsy by a pathologist. The compounds of the present application may he administered for the purpose of preventing hyperplasias, dysplasias, or pre-cancerous lesions from continuing to expand or from becoming cancerous. Examples of pre-cancerous lesions may occur in skin, esophageal tissue, breast, and cervical mtra-epithelial tissue.

As inhibitors or degraders of CDK8 and/or CDK19, the compounds and compositions of this application are also useful in assessing, studying, or testing biological samples. One aspect of the application relates to inhibiting protein kinase activity or degrading protein kinase m a biological sample, comprising contacting the biological sample with a compound or a composition of the application.

The term "biological sample", as used herein, means an in vitro or an ex vivo sample, including, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Inhibition of protein kinase activity or degradation of protein kinase in a biological sample is useful for a variety of purposes that are known to one of skill in the art.. Examples of such purposes include, but are not limited to, blood transfusion, organ

transplantation, and biological specimen storage.

Another aspect of this application relates to the study of CDK8 and/or CDK! 9 in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such protein kinases; and the comparative evaluation of new protein kinase inhibitors. Examples of such uses include, but are not limited to, biological assays such as enzyme assays and cell-based assays.

The activity of the compounds and compositions of the present application as CDK8 and/or CDK19 inhibitors or degraders may be assayed in vitro , in vivo, or in a cell line. In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity or degradation of the activated kinase. Alternate in vitro assay s quantitate the ability of the inhibitor or degrader to bind to the protein kinase and may be measured either by radio labelling the inhibitor or degrader prior to binding, isolating the inhibitor/kinase or degrader/kinase complex and determining the amount of radio label bound, or by running a competition experiment where new inhibitors or degraders are incubated with the kinase bound to known radioligands. Detailed conditions for assaying a compound utilized in this application as an inhibitor or degrader of various kinases are set forth in the Examples below. In accordance with the foregoing, the present application provides a method for preventing or treating any of the diseases or disorders described above in a subject m need of such treatment, comprising administering to the subject a therapeutically effective amount of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

For any of the above uses, the required dosage will vary depending on the mode of

administration, the particular condition to be treated and the effect desired.

Compounds and compositions of the application can be administered in therapeutically effective amounts in a combinational therapy with one or more therapeutic agents

(pharmaceutical combinations) or modalities, e.g., an anti-proliferative, anti-cancer,

immunomodulatory or anti-inflammatory agent. Where the compounds of the application are administered in conjunction with other therapies, dosages of the co-administered compounds will of course vary depending on the type of co-drug employed, on the specific drug employed, on the condition being treated and so forth. Compounds and compositions of the application can be administered in therapeutically effective amounts in a combinational therapy with one or more therapeutic agents (pharmaceutical combinations) or modalities, e.g., anti-proliferative, anti- cancer, immunomodulatory or anti-inflammatory agent, and/or non-drug therapies, etc. For example, synergistic effects can occur with anti-proliferative, anti -cancer, immunomodulatory or anti-inflammatory substances. Where the compounds of the application are administered in conjunction with other therapies, dosages of the co-administered compounds will of course vary depending on the type of co-drug employed, on the specific drug employed, on the condition being treated and so forth.

Combination therapy includes the administration of the subject compounds in further combination with one or more other biologically active ingredients (such as, but not limited to, a second CDK8 inhibitor, a second CDK19 inhibitor, a second and different anti neoplastic agent, a second cy elm-dependent kinase inhibitor (i.e., CDK1, CDK2, CDK4, CDK6, CDK7, CDK9 CDK1 1 , CDK12, CDK! 3, CDK14, etc.) and non-drug therapies (such as, but not limited to, surgery or radiation treatment). For instance, the compounds of the application can be used in combination with other pharmaceutically active compounds, preferably compounds that are able to enhance the effect of the compounds of the application. The compounds of the application can be administered simultaneously (as a single preparation or separate preparation) or sequentially to the other drug therapy or treatment modality. In general, a combination therapy envisions administration of two or more drugs during a single cycle or course of therapy.

In another aspect of the application, the compounds may be administered in combination with one or more separate pharmaceutical agents, e.g., a chemotherapeutic agent, an

immunotherapeutic agent, or an adjunctive therapeutic agent.

Another aspect of the present application relates to a kit comprising a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application.

Another aspect of the present application relates to a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in the manufacture of a medicament for modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cyclin- dependent kinase, such as CDK8 and/or CDK19), for treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role), or for treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or activated CDK19 or wherein the subject is identified as being in need of inhibition of CDK8 and/or CDK19 for the treatment or prevention of cancer.

Another aspect of the present application relates to use of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, in the manufacture of a medicament for modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cyclin-dependent kinase, such as CDK8 and/or CDK19), for treating or preventing a disease (e.g, a disease in which CDK8 and/or CDK19 plays a role), or for treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or activated CDK19 or wherein the subject is identified as being in need of inhibition of CDK8 and/or CDK19 for the treatment or prevention of cancer.

Another aspect of the present application relates to a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, for use in modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cvclin-dependent kinase, such as CDK8 and/or CDK19), m treating or preventing a disease (e.g., a disease m which CDK8 and/or CDK19 plays a role), or in treating or preventing cancer in a subject, wherein the cell of the cancer composes an activated CDK8 and/or activated CDK19 or wherein the subject is identified as being in need of inhibition of CDK8 and/or CDK19 for the treatment or prevention of cancer.

Another aspect of the present application relates to use of a bifunctional compound of the application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the application, in modulating (e.g., inhibiting or decreasing the amount of) a kinase (e.g., a cyclin-dependent kinase, such as CDK8 and/or CDK19), m treating or preventing a disease (e.g., a disease in which CDK8 and/or CDK19 plays a role), or in treating or preventing cancer in a subject, wherein the cell of the cancer comprises an activated CDK8 and/or activated CDK19 or wherein the subject is identified as being in need of inhibition of CDK8 and/or CDK19 for the treatment or prevention of cancer.

Pharmaceutical Compositions

In another aspect, the application provides a pharmaceutical composition comprising a therapeutically effective amount of a bifunctional compound of the present application or an enantiomer, diastereomer, stereoisomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

Compounds of the application can be administered as pharmaceutical compositions by any conventional route, in particular entera!ly, e.g., orally, e.g., in the form of tablets or capsules, or parenteraliy, e.g., in the form of injectable solutions or suspensions, or topically, e.g. , in the form of lotions, gels, ointments or creams, or in a nasal or suppository form.

Pharmaceutical compositions comprising a bifunctional compound of the present application in free form or in a pharmaceutically acceptable salt form in association with at least one pharmaceutically acceptable carrier or diluent can be manufactured in a conventional manner by mixing, granulating or coating methods. For example, oral compositions can be tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g, magnesium aluminum silicate, starch paste, gelatin, tragacanth, methy!cellulose, sodium carboxymethylcellulose and or polyvinylpyrrolidone; if desired d) disintegrants, e.g, starches, agar, algimc acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners. Injectable compositions can be aqueous isotonic solutions or suspensions, and suppositories can be prepared from fatty emulsions or suspensions. The compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances. Suitable formulations for transdermal applications include an effective amount of a bifunctional compound of the present application with a carrier. A carrier can include absorbable

pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin. Matrix transdermal formulations may also be used. Suitable formulations for topical application, e.g., to the skin and eyes, are preferably aqueous solutions, ointments, creams or gels well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.

The pharmaceutical compositions of the present application comprise a therapeutically effective amount of a bifunctional compound of the present application formulated together with one or more pharmaceutically acceptable carriers. As used herein, the term "pharmaceutically acceptable carrier" means a non-toxic, inert solid, semi-solid or liquid filler, diluent

encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes,

polyethylenepo!yoxy propylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes, oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate, agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; algimc acid; pyrogen- free water, isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium laury! sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.

The pharmaceutical compositions of this application can be administered to humans and other animals orally, reetaily, parenterally, mtracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buecaily, or as an oral or nasal spray.

Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylfermamide, oils (m particular, cottonseed, groundnut, com, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Injectable preparations, for example, sterile injectable aqueous, or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

In order to prolong the effect of a drug, it is often desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally

administered drug form is accomplished by dissolving or suspending the drug m an oil vehicle.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this application with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppositoiy wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

Solid compositions of a similar type may also be employed as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.

Dosage forms for topical or transderrnal admini tration of a compound of this application include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable earner and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this application.

The ointments, pastes, creams and gels may contain, in addition to an active compound of this application, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Powders and sprays can contain, in addition to the compounds of this application, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.

Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

EXAMPLES

Analytical Methods, Materials, and Instrumentatio

All reactions were monitored by thin layer chromatography (TLC) with 0.25 mm E. Merck pre-coated silica gel plates (60 F254), Waters LCMS system (Waters 2489 UV/Vxsible Detector, Waters 3100 Mass, Waters 515 HPLC pump, Waters 2545 Binary' Gradient Module, Waters Reagent Manager, Waters 2767 Sample Manager) using SunFire™ CI S column (4.6 x 50 mm, 5 pm particle size): solvent gradient = 100% A at 0 min, 1% A at 5 min; solvent A :::

0.035% TEA in Water; solvent B = 0.035% TEA in MeOH; flow' rate : 2.5 mL/min, and/or Waters Acquity UPLC/MS system (W 7 aters PDA eX Detector, QDa Detector, Sample manager - FL, Binary Solvent Manager) using Acquity UPLC® BEH Cl 8 column (2.1 x 50 mm, 1.7 mhi particle size): solvent gradient = 90% A at 0 min, 1% A at 1.8 min; solvent A = 0.1% formic acid in Water; solvent B = 0.1% formic acid in Acetonitrile; flow' rate : 0.6 mL/min. Reaction products were purified by flash column chromatography using CombiFlash ® Rf with Teledyne Isco Redi&p^f High Performance Gold or Silicycle Silia&p™ High Performance columns (4 g, 12 g, 24 g, 40 g, or 80 g). Waters HPLC system using SunFire™ Prep Cl 8 column (19 x 100 mm, 5 mhi particle size): solvent gradient = 80% A at 0 min, 5% A at 25 min; solvent A =

0.035% TEA in Water; solvent B = 0.035% TEA in MeOH; flow' rate : 25 mL/min (Method A), and Waters Acquity UPLC/MS system (Waters PDA el Detector, QDa Detector, Sample manager --- FL, Binary Solvent Manager) using Acquity UPLC® BEH Cl 8 column (2.1 x 50 mm, 1.7 pm particle size): solvent gradient = 80% A at 0 min, 5% A at 2 min; solvent A ::: 0.1% formic acid in Water; solvent B = 0.1% formic acid in Acetonitrile; flow' rate : 0.6 mL/min (method B). The purity of all compounds was over 95% and was analyzed with Waters LCMS system. ¾ NMR w¾s obtained using a 500 MHz Bruker Avance III. Chemical shifts are reported relative to dimethyl sulfoxide (S : = 2.50) or chloroform (0 :::: 7.26) for l H NMR. Data are reported as (hr = broad, s = singlet, d = doublet, t = triplet, q— quartet, m = multiplet).

Abbreviations used in the following examples and elsewhere herein are:

AcOH acetic acid

atm atmosphere

B0C2O di-tert-butyl dicarbonate

BuLi «-butyllithium

br broad

CBi-4 carbon tetrabromide

CDCh deuterated chloroform

DCM dichloromethane

DIEA N,N-diisopropyletIiylamine

DMAP 4-dimethylaminopyridine

DMA N,N-dimethyiaeetamide

DMF N,N-dimethylformamide

DMSO dimethyl sulfoxide

DMSO-de deuterated dimethyl sulfoxide

EDCI 1 -ethyl-3-(3-dimethy!aminopropy!) earbodhmide

ESI electrospray ionization

EtOAc ethyl acetate

HC1 hydrochloric acid

h hour(s)

HAITI bis(dimethylamino)methylene] - 1H- 1 ,2,3 -triazolof 4,5-bjpyridimum 3 - oxide hexafluoro-phosphate

HPLC high-performance liquid chromatography

Hz hertz

KHMDS potassium hexamethylsilazide

LCMS liquid chromatography-mass spectrometry

m multiplet

mL milliliter

MeCN acetonitrile

MeOH methanol

g milligram

mmol millimole

MgS0 4 magnesium sulfate MHz megahertz

min minutes

MS mass spectrometry

NaaCCb sodium carbonate

NaHCCb sodium bicarbonate

NMR nuclear magnetic resonance

Tf inflate

Pd?.(dba)3 tris(dibenzylideneacetone)dipalladium(0)

Pd(PPh 3 )2Cl2 bisitriphenylphosplune)palladium(ll) dichloride

PiiNiSChCTYb N-phenyl-bis(trifluoromethanesulfonamide

PMe 3 trim ethy Iphosphine

PPh 3 triphenyiphosphine

ppm parts per million

PTSA para-toluene sulfonic acid

TBAF tetra-n-butylammonium fluoride

t-BuOH tert-butanol

TFA trifluoroacetic acid

TMS trimethylsilane

THF tetrahydrofuran

TLC thin layer chromatography

pL microiiter

Xphos 2-dicyclohexyiphosphino-2',4',6'-triisopropylbiphenyl

To a solution of (3A,8A,9S, 10/\y 3S,I46)-i 7-iodo-iO,13~dimethyi- 2,3,4,7,8,9,10,1 1 ,12, 13,14, 15-dodecahydro-l H-cyclopenta[a]phenanthren-3-ol (50 mg, 0.13 mmol) in 1 ,4-dioxane (5 ml.) and Na 2 C03 2M aq. (0.32 rnL, 0.63 mmol) was added isoquinoiin- 7-ylboromc acid (25 mg, 0.14 mmol). The solution was thoroughly degassed and then

Pd(PPh3)2.Cb. (5 mg, 0.006 mmol) was added and the mixture was heated to 90 °C for 1 hour. The reaction was quenched with H2O and extracted with EtOAc (3 x 50 mL), dried over MgSQ 4 , and condensed to give a brown oil that was purified by reverse phase chromatography using a gradient of 1-90% MeCN in H2O to give the title compound (1) as a beige solid (40 mg, 80% yield). 41 NMR (500 MHz, DMSO-de): d 9.66 (s, 111).8.55 (d, J = 5Hz, ill).8.35 (s, ill).8.20 (d, ./ 61 lz.1H), 8.13 (s.2H), 6.39 (s, 1H), 5.33 (s, 1H), 3.28 (m, 1H), 3.17 (s, 1H), 2.38-1.99 (m, 6H), 1.81-1.52 (m, 8H), 1.45-1.33 (m, 3H), 1.15 (s.3H), 1.03 (s, 3H). MS m/z 400.39

Compounds 2-7 in Table 1 above were prepared by this procedure from appropriate starting materials and appropriate boronic ester/acid.

Compound 2:

76% yield 'll NMR (500 MHz, DMSO-de): d 9.65 (s, 1H), 8.56 (d, ./ 5Hz, 111).8.32 (s, 1H), 8.17 (d, ./= 6Hz, 1H), 8.12 (s, 2H), 6.37 (s, 1H), 3.38 (m, 1H), 3.17 (s, 1H), 2.41-2.22 (m, 3H), 1.75-1.14 (m, 17H), 1.11 (s, 3H), 084 (s, 3H) LCMS m/z 401.74 |M !lj .

Compound 3:

68% yield 'll N R (500 MHz, DMSO-de): d 9.86 (s, ill).9.80 (s, ill).8.28 (s, ill). 8.24 (d, J= 6Hz, 2H), 6.51 (s, 1H), 5.34 (s, 1H), 3.26 (m, 1H), 3.17 (s, 1H), 241-1.87 (m, 6H), 1.83-1.54 (m, 8H), 1.41-1.30 (m, 3H), 111 (s, 3H), 1.01 (s, 3H) LCMS m/z 400.86 jM 11]

Compound 4:

60% yield. NMR (500 MHz, DMSO-de): d 9.38 (s, 1H), 8.06 (br, 2H), 7.89 (s, 1H), 7.80 (d, J = 6Hz, 1H), 7.54 (d, J = 6Hz, 1H) 6.34 (s, IB), 5.32 (s, IB), 329 (m, 1H), 3.17 (s,

III).1.83-1.21 (m, 1711).0.93 (s, 341).0.44 (s, 311). LCMS m/z 416.37 IM 11 ] .

Compound 5:

54% yield. 1 B NMR (500 MHz, DMSO-de): d 12.96 (s, 1H), 794 (s, 1H), 7.48 (s, 1H), 7.40 (d, J = 6Hz, 1H), 7.19 (d, J = 6Hz, 1H), 6.39 (s, 1H), 5.30 (s, 1H), 5.12 (s, 1H), 4.63 (s, 1H), 3.27 (m, 1H), 2.22-1.20 (m, 1811).0.93 (s, 3H), 0.43 (s, 3H). LCMS m/z 403.61 iM-!!j .

Compound 6:

58% yield. 1 H NMR (500 MHz, DMSO-de): d 12.91 (s, 1H), 7.98 (s, 2H), 7.58 (s, 1H), 7.43 (d, J = 6Hz, 1H), 7.21 (d, J = 6Hz, 1H), 6.38 (s, 1H), 5.30 (s, 1H), 4.63 (s, 1H), 3.27 (m,

111).2.22-1.20 (m, 18H), 0.93 (s, 3H), 0.43 (s, 311). LCMS m/z 389.48 I M i I ] .

Compound 7: 53% yield. ¾ NMR (500 MHz, DMSO-d6): d 12.91 (s, 1H), 7.97 (s, 1H), 7.46 (s, 1H), 7.31 (d, J= 6Hz, 1 1 1). 7.18 (d, J= 6Hz, H I). 6.39 (s, I l f ). 5.30 (s, I l f ). 4.63 (s, I l f ). 3.27 (m.

1 1 1). 2.22-1.20 (m, 19H), 0.93 (s, 31 1). 0.43 (s, 31 1). MS m/z 389.48 | M I f ] .

Compound 48:

64% yield. J H NMR (500 MHz, DMSO-de): d 9.47 (s, 1H), 8.44 (s, 1H), 8.20 (s, 2H), 8.11 (d, J= 6Hz, 1H), 7.95 (d, J= 61 1;· ' . 1H), 6.40 (s, 1H), 5.30 (s, i l l). 3.29 (m, 1H), 2.97 (1. ./ 10Hz, 1H), 2.30-1.96 (m, 5H), 1.85-1.26 (m, 12H), 0.91 (s. 3H), 0.43 (s, 3H). LCMS m/z 400.56 [M+Hf.

Example 2: (3N,8N,9N,10R,13N,14,S',17,S')-17-(isoqiimolm-7-yI)-10,13~di mefhy]l·

2,3,4,7,8,9,10,ll,12,13,14,15,16,17-tetradecaliydro-li/-e ydopenta[a]plieiiasithre!i-3-ol (8a)

To a solution of (3 S , ,8i?,95',l 0/?,135',l 45)-l 7-(isoquinolin-7-yl)-10,l 3-dimethyi- 2,3,4,7,8,9,10,1 1 ,12, 13, 14,15-dodecahydro-l H-cyclopenta[a]phenanthren-3-ol (1) (10 mg, 0.025 mmol) in DMSO/THF (1 : 1 ) (3 mL) was added potassium diazene-1 ,2-diearboxylate (1 09 rng, 0.50 mmol) and AcOH (64 pL, 1 mmol) in 3 portions over 2 hours. The mixture was stirred at rt overnight. An additional 109 mg of diazene-l,2-dicarboxylate and 64 pL of AcOH was added and the mixture stirred 2 h. The mixture was quenched with sat. aq. NaHCCb and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with H?0, brine, dried over MgSOr, and condensed to give a yellow oil that was purified by reverse phase HPLC using a gradient of 1-80% MeCN in HzO to give the title compound (8a) as a brown oil (6 mg, 60 % yield). ¾ NMR (500 MHz, DMSO-d6): d 9.66 (s, 1H), 8.57 (s, 1H), 8.29 (s, 2H), 8.16 (d, J =

61 Iz. 1H), 7.99 (d, J = 6Hz, 1H), 5.32 (s. 1 1 1). 3.28 (m, 1H), 2.98 (t, J = 10Hz, 1H), 2.35-1.98 (m, 5H), 1.87-1.29 (m, 12H), 0.93 (s, 3H), 0.46 (s, 3H). MS m/z 402.39 [M+H]7

Compounds 8b and 9-14 in Table 1 above were prepared by this preceding procedure from Compounds 2-7 and 1, respectively.

Compound 8b: 71% yield. 'll NMR (500 MHz, DMSO-d6) d 9.68 (s, ill).8.54 (d, J - 51 iz.111).8.37 (d, J zzz 61 iz. Hi).8.31 (s, Hi).8.19 (d, J = 61 iz. ifi).8.04 (d, J = 61 iz. Hi).3.36 (m, lif).3.17 (s, IH), 2.97 (t, J = 10Hz, 1H), 2.41-2.22 (m, 3H), 1.78-1.12 (m, 17H), 0.72 (s, 3H), 0.41 (s, 3H). MS m/z 404.76 i\Ml) .

Compound 9:

64% yield 'll NMR (500 MHz, DMSO-d6) d 9.86 (s, IH), 9.80 (s, IH), 8.28 (s, IH), 8.24 (d, J = 6Hz, 2H), 5.31 (s, IH), 4.87 (s, IH) 3.26 (m, IH), 3.17 (s, IH), 2.41-1.87 (m, 6H), 1.83-1.54 (m, 10H), 1.41-1.30 (m, 3H), 1.09 (s, 3H), 0.98 (s, 3H). MS m/z 403.38 [M+H] + .

Compound 10:

52% yield 'll NMR (500 MHz, DMSO-d6) d 9.36 (s, IH), 8.14 (br, 2H), 7.91 (s, IH), 7.86 (d, J = 6Hz, IH), 7.57 (d../ 61 Iz. IH), 5.33 (s, IH), 4.72 (s, IH), 3.29 (m, IH), 3.17 (s,

IH), ), 2.97 (t, J = 10Hz, IH), 1.85-1.20 (m, 1811).0.72 (s, 3H), 0.41 (s, 3H). MS m/z 417.96

Compound 11:

48% yield 'll NMR (500 MHz, DMSO-d6): d 1296 (s, 111).794 (s, 111).7.48 (s, 111). 7.40 (d, J = 61 iz.1H), 719 (d, J = 6Hz, ill).5.32 (s, ill).4.66 (s, Hi).3.27 (m, 111).2.22-1.20 (m, 20H), 093 (s, 3H), 043 (s, 3H) MS m/z: 405.83 [M II!

Compound 12:

50% yield. 12.91 (s, 1H), 798 (s, 1H), 7.58 (s, 1H), 7.43 (d, J = 6Hz, HI).721 (d, J - 61 iz. HI).5.31 (s. III).4.67 (s, 111).3.27 (m, 111).2.22-1.20 (m, 2011).092 (s, 311).044 (s.311). MS m/z; 391.28 |M 11!

Compound 13;

53% yield. l U NMR (500 MHz, DMSO-d6): d 12.91 (s, 1H), 797 (s, 1H), 7.46 (s, 1H), 7.31 (d, J = 6 Hz, III).718 HI J - 61 iz. ill).5.32 (s, 111).4.63 (s, 111).3.27 (m, 111).2.22-1.20 (m, 20H), 0.91 (s, 3H), 0.46 (s, 311). MS m/z: 391.34 |M ll| .

Compound 14:

68% yield. ¾ NMR (500 MHz, DMSO-d6): d 9.36 (s, 1H), 8.14 (br, 2H), 7.91 (s, 1H), 7.86 (d, J = 61 Iz. ! i 1 ).7.57 HI J - 61 iz. ill).5.33 (s, 111).3.29 (m, IH), 3.17 (s, 111). ), 2.98 (t, J = ! 01 Iz. HI).1.85-1.20 (m, 19H), 0.72 (s, 311).0.41 (s, 3H). MS m/z: 402.83 i\Ml) ·. Example 3: (3A',8i?,9S',10S',13 ,14M-17-(isoqumolm-7-yl)-N,N,10,13-tetrametliyl-

2,3,4,5,6,7,8,9,10,11 2,13,14,15-tetradecahydro-lH-cycIopenta[a]phenanthren-3-amin e

Compound 22r was prepared according to literature procedure. Czako, J. Am Chem.

Soc. 2009, 131, 9014-9019.

To a solution of (35',8ii,9^,10 , ,l35',l45)-3-(dimethylamino)-l 0,13-dimethyl- 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15-tetradecahydro- l/f-cyclopenta[a]phenanthren-l 7-y 1 trifluoromethanesulfonate (100 mg, 0.22 mmol) in 1,4-dioxane (5 mL) and 2M aq. NaaCOs (0.55 inL, 1.1 mmol) was added isoquinolin-7-yl boronic acid (46 mg, 0.27 mmol). The mixture was thoroughly degassed and Pd(PPJi3)2Ch (8 mg, 0.011 mmol) was added. The mixture was stirred at 90 °C for 1 hour. The reaction was quenched with H2O and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with H2O, brine, dried over MgSQ 4 , and condensed to give a yellow oil that was purified by reverse phase HPLC using a gradient of 1 - 70% MeCN in H2O to give the title compound as a brown oil (38 mg, 40 % yield). MS m/z 429.39 ( M i l )

tetramethyihexadecahydro-li/-cydopesitafa]phesiaiithreii- 3-amiiie (23)

Compound 22r was prepared according to literature procedure. Czako B., et ai,J. Am. Chem. Soc.2009, 131, 9014-9019. Compound 23 was prepared from compound 22r according to the reductive diimide procedure described above in the preparation of compound 8a. MS m/z 431.31 (M il)

Compounds 24 and 25 in Table 1 above were prepared in a similar manner as

Compounds 22r and 23, respectively.

Compound 24: LCMS m/z 429.47 |\Mlj .

Compound 25: LCMS m/z 43182 [M+H]7

Step 1: synthesis of intermediates 26-28

Intermediates 26-28 were prepared in a similar manner according to literature procedure, beginning with cfr-dehydroepiadrosterone, as shown in Scheme 4. Czako, B , et al. J. Am. Chent. Soc. 2009, 131, 9014-9019. Intermediate 29a was prepared as shown in Scheme 3.

Step 2: synthesis of intermediate 29a

(3. ', SR,9S, 1 OR, 135, 14,S)-3 -azido- 10, 13 -dimethyl- 1 ,2, 3 ,4, 7, 8, 9, 10, 11,12, 13 , 14, 15 , 16- tetradecahydrospiro[cyclopenta[a]phenanthrene- 17,2’-[ 1 ,3]dioxolane] (28) (2 5 59 mmol) was dissolved in anhydrous THF (50 rnL). ThO (503 pL, 28 mmol) was added after PMes (1.15 mL, 11.2 mmol) was added. The mixture was stirred for 6 h. The reaction was quenched with H2O and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with H2O, brine, dried over MgSC and condensed to give a yellow oil that was purified by flash chromatography using a gradient of 1-10% MeOH in DCM to give intermediate 29a as a white solid (1.58 g, 85% yield). l H NMR (500 MHz, CDCb): d 5.39 (s, 1H), 3.97-3.86 (m, 4H), 3.47 (br, 2H), 2.81 (m, 4H), 2.28 (m, IH), 2.05-1.98 (m, 2H), 1.92-1.78 (m, 3H), 1.74- 1.67 (m, 1H), 1.64-1.38 (m, 9H), 1.32-1.24 (m, IH), 1.15-1.07 (m, IH), 1.03 (s, 3H), 0.88 (s, 3H). MS m/z 332.47 ( M i l l

Steps 3 and 4\ synthesis of compounds 32 and 33

Compounds 32 and 33 w r ere prepared from intermediate 29a, as shown above in Scheme 4.

Compound 32:

i l NMR (500 MHz, DMSO-d6): d 9.65 (s, IH), 8.57 (d, J = 6Hz, IH), 8.33 (s, IH), 8.17 (d, J = 6Hz, IH), 8.12 (s, 2H), 6.39 (s, IH), 5.49 (s, IH), 3 17 (s, IH), 3 04 (m, 4H), 2.78 (s, 61 1). 2.45-2.26 (m, 5H), 1.98-1.88 (m, 3H), 1.81 -1.55 (m, 6H), 1.49-1.41 (m, IH), 1.17 (s, 3H), 1 06 (s, 3H). MS m/z: 427.29 [ M i l ]

Compound 33:

¾ NMR (500 MHz, DMSO-d6): d 9 72 (s, IH), 8 61 (d, J = 6Hz, IH), 8.35 (d, J = 6Hz, IH), 8.33 (s, IH), 8.20 (d, J = 6Hz, IH), 8.03 (d, J = 61 1/. IH), 5.47 (s, IH), 3.44 (m, IH), 3.0 (t, J = 10Hz, IH), 2 77 (s, 6H), 2.44-2.27 (m, 5H), 1.68-1.28 (m, 121 !). 1.16-1.00 (m, 4H), 0.96 (s, 3H), 0.47 (s, 31 1) MS m/z: 429 58 [ M i l l .

Compound 34 was prepared in an analogous manner as demonstrated in Czako, B., et al. J Am. Ghent. Soc. 2009, 131, 9014-9019, i.e., the unsaturated equivalent of intermediate 29a was treated with 2-bromoethyl ether and NaHCCb m toluene to the corresponding morpholine compound; the ketal was removed with para-toluene sulfonic acid (PTSA) in acetone and the resulting ketone was converted to the vinyl triflate with potassium hexamethyldisilazide

(KHMDS) and PhN(S02CF 3 )2 in THF. The vinyl triflate was subsequently coupled with isoquinolme boronic acid under the Suzuki conditions described throughout.

yl)hexadecahydro-l//-cyclopenta[a|phenanthren-17-yl)isoqu inoIine (39)

Scheme 6.

Step 1: synthesis of intermediate 35

To a solution of (3S SR,9S,K)S,l 3S',14V)-3-azido-10,13-dimethylhexadecahydrospiro-

[cyclopenta[a]phenanthrene-l 7,2'-[l ,3]dioxolane] (500 mg, 1.39 mmol) in t-BuOH (10 mL) and H2O (3 mL) was added CuS04,5H2.0 (18 mg, 0.07 rnmol) and sodium ascorbate (6 mg, 0.03 mmol) followed by TMS-acetylene (231 mE, 1.67 mmol). The reaction mixture was heated to 70 °C for 1 h. The reaction was quenched with H2O and extracted with EtOAe (3 x 50 mL). The combined organic extracts were washed with H2O, brine, dried over MgSCti, and condensed to give a yellow oil that was dissolved in THF (20 mL). TBAF 1M in THF (6.95 mL, 6.95 mmol) was added and stirred for 30 minutes. The reaction was quenched with H2O and extracted with EtOAe (3 x 50 mL). The combined organic extracts were washed with II2O, brine, dried over MgSCti, and condensed to give a brown oil that was purified by flash chromatography using a gradient of 10-60% EtOAe in hexanes to give intermediate 35 as a white solid (348 mg, 65 % yield). Ή NMR (500 MHz, CDCb): 6 8.01 (q, J= 10 Hz, 2H), 4.31 (m, 1H), 3.95-3.82 (m, 4H), 2.83 (m, 3H), 2.27 (m, i l l). 2.03-1.95 (m, 2H), 1.90-1.75 (m, 3H), 1.71-1.63 (m, 1H), 1.67-1.39 (m, 10H), 1.35-1.26 (m, 1H), 1.13-1.07 (m, IH), 0.97 (s, 3H), 0.78 (s, 3H). MS m/z 386.76 i \ l · i f j .

Steps 2 and 3: synthesis of intermediates 36 and 37

Intermediate 36 (MS m/z 341.86 [M+H] + ) was prepared using the procedure used to prepare intermediates 20 and 30 above. Intermediate 37 (MS m/z 474.45 [M+H] ÷ ) was prepared using the procedure used to prepare intermediates 21 and 31 above.

Step 4: synthesis of compound 38

Compound 38 was prepared from intermediate 37 using the procedure used to prepare compound 1 above. Ή NMR (500 MHz, DMSO-de): d 9.74 (s, 1H), 8.53 (d, J= 6Hz, 1H), 8.17 (s, 2H), 8.03 (q, J= 10Hz, 2H), 7.90 (s, 1H), 7.69 (s, 1H), 6.38 (s, 1H), 4.52 (m, 1H), 2.33-2.11 (m, 3H), 2.12-1.85 (m, 5H), 1.72-1.40 (m, 8H), 1.30-1.20 (m, 2H), 1.15-1.08 (m, 1H), 0.97 (s, 3H), 0.92-0.87 (m, IH), 0.51 (s. 3H). MS m/z 453.51 [M+Hf

Step 5: synthesis of compound 39

Compound 39 was prepared from compound 38 using the procedure used to prepare compounds 8a. l H NMR (500 MHz, DMSO-d ): d 9.76 (s, IH), 8.57 (d, J= 6Hz, IH), 8.21 (s, 2H), 8.06 (q, ./= iOHz, 2H), 7.92 (s, IH), 7.72 (s, IH), 4 58 (m, IH), 3.02 (t, ,/= 10Hz, IH), 2.33-2.11 (m, 3H), 2.06-1.82 (m, 5H), 1.69-1.37 (m, 10H), 1.33-1.22 (m, 2H), 1.13-1.04 (m, IH), 0.95 (s, 3H), 0.92-0 87 (m, IH), 0.53 (s, 3H). MS m/z 455 68 [M+Hf.

Example 8: (3 i S’,8i?,9A ,10A ,13S T ,14S T ,17N)-17~(isoqiimolm-7-y!)-A r , 10,13- trimethyIhexadeeahydro-li/-eydopei8ta[a]pheiiaislhren-3-amin e (46)

Scheme 7.

Step 1: synthesis of intermediate 40

To a solution of (3481?, 94,104, 134,144)-10,l 3-dimethylhexadecahydrospiro- [cyciopen†a[a]phenanthrene-17,2'-[l,3]dioxolan]-3-amine (500 mg, 1.50 mmol) in THF (25 rnL) was added B0C2O (393 mg, 1 80 mmol) followed by DMAP (37 mg, 0 3 mmol). The mixture was stirred for 3 h. The reaction was quenched with H2O and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with H2O, brine, dried over MgSC>4, and condensed to give a yellow oil that was purified by flash chromatography using a gradient of 5-20% EtOAc m Hexanes to give intermediate 40 as a white solid (602 mg, 93% yield). ¾ NMR (500 MHz, CDCb): 5 6.42 (br, 1H), 3.92-3.81 (m, 4H), 2.06-1.89 (m, 1H), 1.80-1.52 (m, ! OH), 1.46 (s, 9H), 1.41-1.18 (m, 1 OH), 1.08-0.90 (m, 2H), 0.81 (s, 3H), 0.79 (s, 3H). MS m/z 434.58 [M+Hf.

Step 2: synthesis of intermediate 41

To a solution of tert-butyi ((3, 9,87?, 94,105', 13, S’, 145)- 10, 13- dimethy lliexadecahy drospiro [cyclopenta[a] phenanthrene- 17,2'- [ 1 ,3 ] dioxolan] -3 -yl)carbamate

(602 mg, 1.39 mmol) in DMF (10 mL) was added NaH 60% in mineral oil (83 mg, 2.08 mmol). The mixture was stirred for 30 minutes, then Mel (208 pL, 3.34 mmol) was added and stirred overnight. The reaction was quenched with H2O and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with H2O, brine, dried over MgSQ 4 , and condensed to give a yellow oil that was purified by flash chromatography using a gradient of 5-30% EtOAc in Hexanes to give intermediate 41 as a white solid (602 mg, 93% yield). l H NMR (500 MHz, CDCb): 5 3.95-3.82 (m, 4H), 2.75 (s, 3H), 2.02-1.93 (m, 1 1 1 ). 1.84-1.50 (m, 10H), 1.48 (s, 9H), 1.45-1.18 (m, 10H), 1.10-0.90 (m, 2H), 0.86 (s, 311), 0.82 (s, 3H). MS m/z 448.83 i \ M ! j .

Steps 3 and 4: synthesis of intermediates 42 and 43

Intermediate 42 (MS m/z 404.31 [M+H] + ) was prepared from intermediate 41 using the procedure used to prepare intermediates 20 and 30 above. Intermediate 43 (MS m/z 536.78 [M+H ) was prepared from intermediate 42 using the procedure used to prepare intermediates 21 and 31 above.

Step 5: synthesis of intermediate 44

Intermediate 44 was prepared from intermediate 43 using the procedure used to prepare Compound 1 above. 4 1 NMR (500 MHz, DMSO-de): d 9.70 (s, 1H), 8.50 (d, J = 6 Hz, 1H),

8.25 (s, 1H), 8.19 (d, J = 6 Hz, 1H), 8.14 (d, J= 6 Hz, 1H), 8.04 (d, J= 8 Hz, i l l). 6.32 s, 1H), 2.77 (s, 3H), 2.40-2.33 (m, H I). 2.21-2.10 (m, 2H), 1.82-1.59 (m, 8H), 1.49 (s, 9H), 1.44-1.24 (m, 8H), 1.15 (s, 3H), 1.12-1.05 (m, 2H), 0 90 (s, 3H) MS m/z 515.63 j M ! i ] .

Step 6: synthesis of compound 45

To a solution of tert-butyl ((36/8i?,9S,106/136yi45)-17-(isoquinolm~7~yl)-10,13- dimethyl-2,3,4,5,6,7,8,9, 10, 11,12, 13 , 14, 15-tetradecahydro-l//-cyclopenta[a]phenanthren-3- yl)(methyl)carbamate (50 mg, 0.1 mmol) in DCM (10 mL) was added TFA (1 ml). The mixture was stirred for 30 min and the solvent removed in vacuo to give compound 45 as a beige solid that was used without further purification (40 mg, 98% yield). MS m/z 41 5.57 [M+H]T

Step 7: synthesis of compound 46

Compound 46 was prepared from compound 45 using the procedure used to prepare compound 8a above. 91% yield. MS m/z 417.37 [M+H] +

dimethylhexadecahydro-li-cyclopenta[a]phenaiithren-3-yl)-iV- methylacetamide (47)

Scheme 8,

To a solution of (35, 8i?, 05, 135, 145, 175)- i 7-(isoquinolin-7-yl)-JV, 10, 13- trimethylhexadecahydro~l/7-cyclopenta[a]phenanthren-3-amine 46 (20 mg, 0.048 mmol) in THF (3 niL) and sat aq. NaHCCb solution (2mL) was added acetyl chloride (4gL, 0.058 mmol). The mixture was stirred for 30 minutes and then the reaction was quenched with H2O and extracted with EtOAc (3 X 50 ml). The combined organic extracts were washed with H2O, brine, dried over MgSOi and condensed to give a yellow oil that was purified by reverse phase HPLC using a gradient of 1-80% MeCN in H20 to give the title compound 47 as a brown solid (12 mg, 57% yield). Ή NMR (500 MHz DMSO-de): d 9.70 is.111).8.62 (d, ./= 6Hz, 111).8.33 (d, J = 6Hz,

111).8.31 is.111).8.22 (d, J = 61 Iz. ill).8.01 (d, J = 6Hz, 111).3.54 (m, 111).3.21 (s, 311).3.1 (t, j 10! Iz.111).2.70 (s, 311).2.44-227 (m, 611).1.68-1.28 (m, 1211).1.16-1.00 (m.4H), 0.94 (s. 311).0.44 is.311). MS rri/z: 45972 [M+H]+.

To a solution of compound 46 (20 mg, 0.048 mmol) m DMF was added HATU (37 mg,

0.096 mmol) and l-((2~(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin~4-yl)ami no)~3, 6,9,12- tetraoxapentadecan-15-oic acid (28 mg, 0.053 mmol) followed by DXEA (42 pL, 0.24 mmol).

The reaction was stirred for 30 minutes and then injected directly onto the HPLC and purified using a gradient of 1-90% MeCM in H2O to give the title compound 1-1 as a yellow solid (8 mg, 18% yield) ¾ NMR (500 MHz, DMSO): d 11.09 (s, 1H), 9.71 (s, 1H), 8.60 (s, 1H), 8.36-829 (m, 3H), 819-8.16 (m, 111).799 (t ../ 5 Hz, HI).7.60-754 (m, HI).7.17-713 (dd ,./= 5 Hz, 8

Hz, Hi).7.04 (d, J= 6 Hz, 1H), 5.12- 5.03 (m, 1H), 427 (m, 1H), 3.64-359 (m, 4H), 3.54-3.45 (m, 14H), 299-2.84 (m, 2H), 279 (s, 311).2.06-1.95 (m, 311).1.83-091 (m, 2411).0.77 (s, 311). 0.43 (s, 311). MS m/z 921.491M 1 S ) . Example 11: Synthesis

yl)amino)ethoxy)ethoxy)-iV-((3^,10S',175 17 7-vl)-10,

dimethylhexadecahydro-l/I-cydopeiita en-3-yl)-/V-methylpropananiide (1-2)

Compound 1-2 was prepared from compound 46 and 3-(2-(2-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)amino)ethoxy)ethoxy)propanoic acid as described above in

Example 10. ¾NMR (500 MHz, DMSO): d 11.10 (s, 1H), 9.67 (s, 1H), 8.59 (s, 1H), 8.31-8.24 (m, 311 ).8.15 (d, J= 7 Hz, 111}.7.96 (t, J= 511/.. HI).7.76 (dd, J= 5 Hz, 81 fz.1H), 7.66-7.52 (m, 111).7.24-7.12 (m, 1H), 5.12 (m, 1H), 4.25 (m, 111).3.68 (m, 8H), 3.00-2.84 (m, 4H), 2.77 (s, 3H), 2.26 (m, 2H), 1.71-0.91 (m, 2511;·.0.79 (s, 3H), 0.46 (s, 3H). MS m/z 832.79 [M ill .

Compound 1-3 was prepared from compound 46 and 3-(2-(2-((l,3-dioxo-2-(2- oxopiperidin-3-yl)isoindoiin-4-yl)amino)ethoxy)ethoxy)propan oic acid as described above in Example 10.

Compound 1-4 was prepared by treating compound 46 and 2-(2,6-dioxopiperidin-3-yl)-4-((2-(2- hydroxyethoxy)ethyl)amino)isoindoline-l,3-dione with 1,6-dibromo hexane in the presence of a base, e.g., potassium carbonate, DIEA, TEA, or the like, in a solvent, e.g., polar aprotic solvent such as DMF.

Example 14. Synthesis of N-((3S,5S,10S,13S,14S,17S)-17-(isoqumolm-7-yI)-10,13- dimethylhexadecahydro-lH-cyclopenta|a]phenanthren-3-yl)-N-me thyl-l-((2-(l-methyl-2,6- dioxopiperidin-3-yI)-l,3-dioxoisoindolin-4-yI)asnino)-3,6,9, 12-tetraoxapentadecan-15-an ide

(1-5)

Compound 1-5 was prepared from compound 46 and l-((2-(l -methyl-2, 6-dioxopiperidin- 3-yl)-! ,3-dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxapentadecan-1 5-oic acid as described above in Example 10. ] H NMR (500 MHz, DMSO): d 9.71 (s, 1H), 8.60 (s, 1H), 8.35-8.29 (m, 3H), 8.19-8.16 (m, 1H), 7.99 (t, J= 5Hz, 1H), 7.61-7.57 (m, 1H), 7.16 (dd , J = 5Hz, 8Hz, i l l ). 7.04 (d, J = 6Hz, 1H), 5.12- 5.03 (m, 1H), 4.27 (m, 1H), 3.64-3.45 (m, 18H), 3.01 (s, 3H), 2.99- 2.91 (m, 2H), 2.80 (s, 3H), 2.06-1.95 (m, 3H), 1.83-0.91 (m, 24H), 1.16 (s, 3H), 1.06 (s, 3H), 0.76 (s, 3H). MS / ' z: 935.71 [M+Hf.

Example 15: Biological activities of the compounds of the application

Compounds of the present application were tested in a competitive kinase binding assay by using commercially available assay system. For example, LanthaScreen ® Eu Kinase Binding Assay (Invitrogen™) was used to measure the ICso of the compounds of the present application in displacing the Tracer from binding to the target kinase, by following the assay protocol from Invitrogen™.

Biological activities of the Targeting Ligands of Table 1 are shown in Table 2 below. Biological activities of the bifunctional compounds are shown in Table 3 below. in Table 2,

ICso ratings: “A” <100 nM;“B” = 101 -200 nM;“C” = 201 -300 nM;“D” = 301-700 nM;“E” = 701-2,000 nM;“F”

=== 2,001-6,000 nM; G " > 6,001 nM. Table 3,

ICso ratings: “A” <100 nM;“B” - 101-200 nM;“C” - 201-300 nM;“D” - 301-700 nM; Έ” - 701-2,000 nM;“F”

- 2,001-6,000 nM;“G” > 6,001 nM.

EQUIVALENTS

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

All patents, patent applications, and literature references cited herein are hereby expressly incorporated by reference.