SPIROCYCLIC ANDROGEN RECEPTOR PROTEIN DEGRADERS

Information

  • Patent Application
  • 20220380368
  • Publication Number
    20220380368
  • Date Filed
    September 18, 2020
    3 years ago
  • Date Published
    December 01, 2022
    a year ago
Abstract
The present disclosure provides compounds represented by Formula (I) and the salts or solvates thereof, wherein R3a, E, L, A1, B1, X1, X2, Z1, and Z2 are as defined in the specification. Compounds having Formula (I) are androgen receptor degraders useful for the treatment of cancer and other diseases.
Description
BACKGROUND OF THE INVENTION
Field of the Invention

The present disclosure provides heterobifunctional small molecules as androgen receptor (AR) protein degraders. AR degraders useful for the treatment of a variety of diseases including cancer.


Background

Despite improvements in medical treatments over the past three decades, prostate cancer is significant cause of cancer-related death, and is second only to lung cancer among men in developed countries. Hamdy et al., N Engl J Med, 2016, 375, 1415-1424; Litwin and Tan, H. J. JAMA, 2017, 317, 2532-2542. In addition to surgery and radiotherapy, androgen deprivation therapies (ADT) are front-line treatments for prostate cancer patients with high-risk localized disease, and second-generation anti-androgens such as abiraterone and enzalutamide have been shown to benefit patients with advanced prostate cancer. Karantanos et al., Oncogene. 2013, 32, 5501-511; Harris et al., Nat Clin Pract Urol, 2009, 6, 76-85. Nevertheless, patients who progress to metastatic castration-resistant prostate cancer (mCRPC), a hormone-refractory form of the disease, face a high mortality rate and no cure is currently available. Narayanan et al., Oncoscience. 2017, 4, 175-177; Crowder et al., Endocrinology. 2018, 159, 980-993.


The androgen receptor (AR) and its downstream signaling play a critical role in the development and progression of both localized and metastatic prostate cancer. Previous strategies that successfully target AR signaling have focused on blocking androgen synthesis by drugs such as abiraterone and inhibition of AR function by AR antagonists such as enzalutamide and apalutamide (ARN-509). Watson et al., Nat Rev Cancer. 2015, 15, 701-711. However, such agents become ineffective in advanced prostate cancer with AR gene amplification, mutation, and alternate splicing. Balbas et al., Elife. 2013, 2, e00499; Lottrup et al., J Clin Endocrinol Metab. 2013, 98, 2223-2229. But in most patients with CRPC, the AR protein continues to be expressed and tumors are still dependent upon AR signaling. Consequently, AR is an attractive therapeutic target for mCRPC. Zhu et al., Nat Commun. 2018, 9, 500; Munuganti et al., Chem Biol. 2014, 21, 1476-485.


The Proteolysis Targeting Chimera (PROTAC) strategy has gained momentum with its promise in the discovery and development of completely new types of small molecule therapeutics by inducing targeted protein degradation. Raina et al., Proc Natl Acad Sci USA. 2016, 113, 7124-7129; Zhou et al., J. Med. Chem. 2018, 61, 462-481.


A PROTAC molecule is a heterobifunctional small molecule containing one ligand, which binds to the target protein of interest, and a second ligand for an E3 ligase system, tethered together by a chemical linker. Bondeson, D. P.; Crews, C. M. Targeted Protein Degradation by Small Molecules. Annu Rev Pharmacol Toxicol. 2017, 57, 107-123. Because AR protein plays a key role in CRPC, AR degraders designed based upon the PROTAC concept could be effective for the treatment of CRPC when the disease becomes resistant to AR antagonists or to androgen synthesis inhibitors. Salami et al., Commun Biol. 2018, 1, 100; Pal et al., Cancer. 2018, 124, 1216-1224; Wang et al., Clin Cancer Res. 2018, 24, 708-723; Gustafson et al., Angew. Chem. Int. Ed. 2015, 54, 9659-9662. Naito et al. have recently reported AR degraders designed based upon the PROTAC concept, which were named Specific and Nongenetic IAP-dependent Protein Erasers (SNIPERs). Shibata et al., J. Med. Chem. 2018, 61, 543-575.


While SNIPER AR degraders are effective in inducing partial degradation of the AR protein in cells, they also induce the auto-ubiquitylation and proteasomal degradation of the cIAP1 protein, the E3 ligase needed for induced degradation of AR protein, thus limiting their AR degradation efficiency and therapeutic efficacy.


(4R)-1-((S)-2-(2-(4-((4′-(3-(4-cyano-3-(trifluoromethyl)phenyl)-5,5-dimethyl-4-oxo-2-thioxoimidazolidin-1-yl)-[1,1′-biphenyl]-4-yl)oxy)butoxy)acetamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide ((ARCC-4) was recently reported as another PROTAC degrader, which was designed using enzalutamide as the AR antagonist and a von Hippel-Lindau (VHL) ligand. Salami et al., Commun Biol. 2018, 1, 100; US 20170327469. ARCC-4 was shown to be more potent and effective than enzalutamide at inducing apoptosis and inhibiting proliferation of AR-amplified prostate cancer cells. ARD-69 was also recently reported as a PROTAC AR degrader. Han et al., J. Med. Chem. 62:941-964 (2019).


There is a need in the art for additional AR degraders to treat prostate cancer and other diseases.


BRIEF SUMMARY OF THE INVENTION

In one aspect, the present disclosure provides heterobifunctional small molecules represented by any one or more of Formulae I, III-VIII, XV, or XVI, below, and the pharmaceutically acceptable salts and solvates, e.g., hydrates, thereof. These compounds are collectively referred to herein as “Compounds of the Disclosure.” Compounds of the Disclosure are androgen receptor (AR) degraders and are thus useful in treating diseases or conditions wherein degradation of the androgen receptor protein provides a therapeutic benefit to a subject.


In another aspect, the present disclosure provides compounds represented by any one or more of Formulae II or IX-XIV, below, and salts and solvates thereof. These compounds are collectively referred to herein as “Intermediates of the Disclosure.” Intermediates of the Disclosure can be used to make the heterobifunctional Compounds of the Disclosure.


In another aspect, the present disclosure provides methods of treating a condition or disease by administering a therapeutically effective amount of a Compound of the Disclosure to a subject, e.g., a human cancer patient, in need thereof. The disease or condition treatable by degradation of the androgen receptor is, for example, a cancer, e.g., prostate cancer, e.g., metastatic castration-resistant prostate cancer.


In another aspect, the present disclosure provides a method of degrading, e.g., reducing the level of, of androgen receptor protein in a subject in need thereof, comprising administering to the individual an effective amount of at least one Compound of the Disclosure.


In another aspect, the present disclosure provides a pharmaceutical composition comprising a Compound of the Disclosure and an excipient and/or pharmaceutically acceptable carrier.


In another aspect, the present disclosure provides a composition comprising a Compound of the Disclosure and an excipient and/or pharmaceutically acceptable carrier for use treating diseases or conditions wherein degradation of the androgen receptor provides a benefit, e.g., cancer.


In another aspect, the present disclosure provides a composition comprising: (a) a Compound of the Disclosure; (b) a second therapeutically active agent; and (c) optionally an excipient and/or pharmaceutically acceptable carrier.


In another aspect, the present disclosure provides a Compound of the Disclosure for use in treatment of a disease or condition of interest, e.g., cancer.


In another aspect, the present disclosure provides a use of a Compound of the Disclosure for the manufacture of a medicament for treating a disease or condition of interest, e.g., cancer.


In another aspect, the present disclosure provides a kit comprising a Compound of the Disclosure, and, optionally, a packaged composition comprising a second therapeutic agent useful in the treatment of a disease or condition of interest, and a package insert containing directions for use in the treatment of a disease or condition, e.g., cancer.


In another aspect, the present disclosure provides methods of preparing Compounds of the Disclosure.


Additional embodiments and advantages of the disclosure will be set forth, in part, in the description that follows, and will flow from the description, or can be learned by practice of the disclosure. The embodiments and advantages of the disclosure will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing summary and the following detailed description are exemplary and explanatory only, and are not restrictive of the invention as claimed.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 is an image of the Western blotting analysis of AR protein levels in prostate cancer VcaP cells treated with Cpd. No. 307 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 2 is an image of the Western blotting analysis of AR protein levels in prostate cancer VcaP cells treated with Cpd. No. 293 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 3 is an image of the Western blotting analysis of AR protein levels in prostate cancer 22RV1 cells treated with Cpd. No. 307 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 4 is an image of the Western blotting analysis of AR protein levels in prostate cancer 22RV1 cells treated with Cpd. No. 293 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 5 is an image of the Western blotting analysis of AR protein levels in prostate cancer LNCaP cells treated with Cpd. No. 307 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 6 is an image of the Western blotting analysis of AR protein levels in prostate cancer LNCaP cells treated with Cpd. No. 293 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 7 is an image of the Western blotting analysis of AR protein levels in prostate cancer VCaP cells treated with 3 nM and 10 nM of Cpd. No. 307 and Cpd. No. 293 at the time points indicated. GAPDH was used as the loading control.



FIG. 8 is an image of the Western blotting analysis of AR protein levels in prostate cancer 22RV1 cells treated with 3 nM and 10 nM of Cpd. No. 307 and Cpd. No. 293 at the time points indicated. GAPDH was used as the loading control.



FIG. 9 is an image of the Western blotting analysis of AR protein levels in prostate cancer LNCaP cells treated with 3 nM and 10 nM of Cpd. No. 307 and Cpd. No. 293 at the time points indicated. GAPDH was used as the loading control.



FIG. 10 is five images of the Western blotting analysis of AR protein levels in the prostate cancer cell line indicated treated with the Compound of the Disclosure indicated for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 11 is two images of the Western blotting analysis of AR protein levels in the VCaP prostate cancer cell line treated with the Compound of the Disclosure indicated for 24 h at 10 nM and 100 nM. GAPDH was used as the loading control.



FIG. 12 is two images of the Western blotting analysis of AR protein levels in the VCaP prostate cancer cell line treated with the Compound of the Disclosure indicated for 24 h at 10 nM and 100 nM. GAPDH was used as the loading control.



FIG. 13 is an image of the Western blotting analysis of AR protein levels in prostate cancer VcaP cells treated with Cpd. No. 122 for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 14 is four images of the Western blotting analysis of AR protein levels in prostate cancer VcaP cells treated with the Compound of the Disclosure indicated for 24 h at the concentrations indicated. GAPDH was used as the loading control.



FIG. 15 is a line graph showing the antitumor activity of Cpd. No. 307 in the AR-positive VCaP xenograft model in SCID mice. Cpd. No. 307 was administered via oral gavage daily starting at day 18 for three weeks.



FIG. 16 is a line graph showing the antitumor activity of Cpd. No. 293 in the AR-positive VCaP xenograft model in SCID mice. Cpd. No. 293 was administered via oral gavage daily starting at day 18 for three weeks.



FIG. 17 is four images of the Western blotting analysis of AR protein levels in VCaP or MDA-MB-453 cells treated with Cpd. No. 200 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 18 is four images of the Western blotting analysis of AR protein levels in VCaP or MDA-MB-453 cells treated with Cpd. No. 201 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 19 is six images of the Western blotting analysis of AR protein levels in VCaP, LNCaP, or 22RV1 cells treated with Cpd. No. 202 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control



FIG. 20 is four images of the Western blotting analysis of AR protein levels in VCaP or LNCaP cells treated with Cpd. No. 203 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 21 is four images of the Western blotting analysis of AR protein levels in VCaP or LNCaP cells treated with Cpd. No. 206 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 22 is four images of the Western blotting analysis of AR protein levels in VCaP or LNCaP cells treated with Cpd. No. 207 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 23 is four images of the Western blotting analysis of AR protein levels in VCaP or LNCaP cells treated with Cpd. No. 208 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 24 is four images of the Western blotting analysis of AR protein levels in VCaP or LNCaP cells treated with Cpd. No. 209 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 25 is four images of the Western blotting analysis of AR protein levels in LNCaP or MDA-MB-453 cells treated with Cpd. No. 152 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 26 is three images of the Western blotting analysis of AR protein levels in VCaP, LNCaP, or MDA-MB-453 cells treated with Cpd. No. 159 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 27 is three images of the Western blotting analysis of AR protein levels in VCaP, LNCaP, or MDA-MB-453 cells treated with Cpd. No. 160 under the conditions indicated in connection with each analysis. GAPDH was used as the loading control.



FIG. 28 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 305, and Cpd. No. 307 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 21.



FIG. 29 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 444, and Cpd. No. 445 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 21.



FIG. 30 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 443, and Cpd. No. 490 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 21.



FIG. 31 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 497, and Cpd. No. 499 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 21.



FIG. 32 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 498, and Cpd. No. 500 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 21.



FIG. 33 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 302, and Cpd. No. 305 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 16.



FIG. 34 is a line graph showing the antitumor activity of enzalutamide, Cpd. No. 344, and Cpd. No. 540 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 16.



FIG. 35 is a line graph showing the antitumor activity of enzalutamide and Cpd. No. 503 in the AR-positive VCaP xenograft model in SCID mice. The compounds were administered via oral gavage daily starting at day 16.





DETAILED DESCRIPTION OF THE INVENTION
I. Compounds of the Disclosure

Compounds of the Disclosure are heterobifunctional AR degraders. In one embodiment, Compounds of the Disclosure are compounds of Formula I:




embedded image


wherein:


R3a is selected from the group consisting of halo, C1-C4 alkyl, and C1-C4 haloalkyl;


Z1 is selected from the group consisting of ═C(H)— and ═N—;


Z2 is selected from the group consisting of ═C(R3b)— and ═N—;


R3b is selected from the group consisting of hydrogen, halo, C1-C4 alkyl, and C1-C4 haloalkyl;


E is a spiroheterocyclenyl;


X1 is selected from the group consisting of —C(═O)—, —S(═O)2—, and —CR4aR4b—; or


X1 is absent;


R4a and R4b are independently selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl;


X2 is selected from the group consisting of —C(═O)—, —S(═O)2—, —O—, and —CR4cR4d—; or


X2 is absent;


R4c and R4d are independently selected from the group consisting of hydrogen and C1-3 alkyl;


L is -J1-J2-J3-J4-J5-,


wherein J1 is attached to X2.


J1 is selected from the group consisting of cycloalkylenyl and heterocyclenyl; or


J1 is absent;


J2 is selected from the group consisting of —(CH2)m1—, —CH═CH—, and —C≡C—;


m1 is 0, 1, 2, or 3;


J3 is selected from the group consisting of alkylenyl, heteroalkylenyl, cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl; or


J3 is absent;


J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl; or


J4 is absent;


J5 is selected from the group consisting of —(CH2)m2—, —O—, —N(R6)—, and —C(═O)—;


m2 is 0, 1, 2, or 3;


R6 is selected from the group consisting of hydrogen and C1-C3 alkyl;


B1 is selected from the group consisting of:




embedded image


embedded image


embedded image


R2a, R2b, R2c, R2d, R2e, R2f, and R2g are independently selected from the group consisting of hydrogen, halo, C1-C3 alkyl, and C1-C3 alkoxy;


R3 is selected from the group consisting of hydrogen, deuterium, fluoro, and C1-C3 alkyl;


m is 1, 2, or 3;


n is 1, 2, or 3;


o is 1, 2, or 3;


p is 1, 2, or 3;


Z is selected from the group consisting of —CR1jR1k— and —C(═O);


R1j and R1k are independently selected from the group consisting of hydrogen and C1-C3 alkyl; or R1j and R1k taken together with the carbon to which they are attached from a C3-C6 cycloalkyl; and


R8 is selected from the group consisting of hydrogen and C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, below, wherein Z is selected from the group consisting of —CH2— and —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein B1 is selected from the group consisting of B1-1, B1-2, B1-3, B1-4, B1-5, B1-6, and B1-7, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to X1;


o and p are independently 0 or 1;


q and r are independently 0, 1, 2, or 3;


wherein the sum of o, p, q, and r is 2, 3, 4, 5, 6, or 7;


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


t, u, v, w, and x are independently 0, 1, 2, or 3;


R1a and R1b are independently selected from the group consisting of hydrogen, C1-C3 alkyl, C1-C4 haloalkyl, optionally substituted C3-C6 cycloalkyl, and (C3-C6 cycloalkyl)C1-C6 alkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted C3-C6 cycloalkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted 4- to 6-membered heterocyclo;


R1c and R1d are independently selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group;


each R1e is independently C1-C3 alkyl;


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


each R1f is independently C1-C3 alkyl;


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


each R1g is independently C1-C3 alkyl; and


h is 0, 1, 2, 3, or 4, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E is E-1, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E 1 is selected from the group consisting of:




embedded image


or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1; and R1a and R1b are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1; and R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted C3-C6 cycloalkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1; and R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted 4- to 6-membered optionally substituted heterocyclo, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, R1a and R1b are hydrogen, and, q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-A:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, R1a and R1b are hydrogen, and q is 2; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-B:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, R1a and R1b are hydrogen, and q is 1; r is 0; s is 0; and t is 2, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-C:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, R1a and R1b are hydrogen, and q is 0; r is 1; s is 1; and t is 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-D:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1, R1a and R1b are hydrogen, and q is 1; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-E:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1; and R1a and R1b are independently C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E 1 is E-1-1; and R1a and R1b are independently C1-C3 alkyl; and q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-F:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E 1 is E-1-1; and R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted C3-C6 cycloalkyl; and q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-1; R1a is C1-C3 alkyl; and R1b is hydrogen; and q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-G:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula III:




embedded image


wherein R1a, R3a, Z1, Z2, X1, A1, X2, L, and B1 are as defined in connection with Formula I, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula IV:




embedded image


wherein R1a, R3a, Z1, Z2, X1, A1, X2, L, and B1 are as defined in connection with Formula I, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E 1 is E-1-1; and R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E 1 is E-1-1; R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group; and q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-1 is E-1-1-H:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein:


E-1 is E-1-2;


R1c is C1-C3 alkyl;


R1d is selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, E-1 is E-1-2; R1c is C1-C3 alkyl; and R1d is selected from the group consisting of hydrogen and C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R1d is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula V:




embedded image


wherein R1c, R3a, Z1, Z2, X1, A1, X2, L, and B1 are as defined in connection with Formula I, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula VI:




embedded image


wherein R1c, R3a, Z1, Z2, X1, A1, X2, L, and B1 are as defined in connection with Formula I, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E-1 is E-1-2; and R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof, i.e., E-1-2 is E-1-2-A:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E is E-2, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, E-2 is:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of Formula I, and Intermediates of the Disclosure are compounds of Formula II, wherein E is E-3, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, E-3 is:




embedded image


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, below, wherein X1 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X1 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X1 is —CR4aR4b—, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R4a and R4b are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X1 is absent, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV, or XVI, see below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein:


A1 is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to X2;


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen, halo, C1-C3 alkyl, and C1-C3 alkoxy;


e is 0, 1, or 2; and


f is 0, 1, or 2, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-1, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5b, R5c, and R5d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, XV, or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-2, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5b, R5c, and R5d are hydrogen. In another embodiment, R5a is fluoro or chloro; and R5b, R5c, and R5d are hydrogen. In another embodiment, R5b is fluoro or chloro; and R5a, R5c, and R5d are hydrogen. In another embodiment, R5c is fluoro or chloro; and R5a, R5b, and R5d are hydrogen. In another embodiment, R5d is fluoro or chloro; and R5a, R5b, and R5c are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV, or XVI, below, wherein A1 is A1-3, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5b, and R5d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-4, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a and R5b are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-5, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-6, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-7, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-8, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5b, and R5c are hydrogen. In another embodiment, e is 0 or 1; and f is 0 or 1.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV, or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-9, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5c, and R5d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV, or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-10, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a and R5d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV, or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-11, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a and R5b are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-12, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a and R5b are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VI, XV or XVI, below, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein A1 is A1-13, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a and R5b are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X2 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X2 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X2 is —O—, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X2 is —CR4cR4d—, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R4c and R4d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein X2 is absent, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula VII:




embedded image


wherein R1a, R3a, Z1, Z2, R5a, R5b, R5c, R5d, J1, J4, J5, and B1 are as defined in connection with Formula I, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula VII, wherein R1a is selected from the group consisting of hydrogen and C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R1a is methyl or ethyl. In another embodiment, Compounds of the Disclosure are compounds of Formula VII, wherein R1a is methyl.


In another embodiment, Compounds of the Disclosure are compounds of Formula VII, wherein R3a is selected from the group consisting of halo and C1-C4 haloalkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula VII, wherein R5a, R5b, R5c, and R5d are each independently selected from the group consisting of hydrogen and halo, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, R5a, R5b, R5c, and R5d are each hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of Formula VII, wherein Z1 and Z2 are —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, J1 is cycloalkylenyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J1 is a C4-C6 cycloalkylenyl.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J1 is heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J1 is selected from the group consisting of:




embedded image


or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J1 is absent, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J2 is selected from the group consisting of —(CH2)m1— and —C≡C—; and m1 is 0, 1, or 2, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J2 is —(CH2)m1—; and m1 is 0. In another embodiment, J2 is —(CH2)m1—; and m1 is 1. In another embodiment, J2 is —C≡C—.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J3 is selected from the group consisting of cycloalkylenyl and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J3 is a C4-C6 cycloalkylenyl. In another embodiment, J3 is a 4- to 10-membered heterocyclenyl.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I-VI, wherein J3 is absent, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J4 is a C1-C6 alkylenyl. In another embodiment, J4 is a C4-C6 cycloalkylenyl. In another embodiment, J4 is a 4- to 10-membered heterocyclenyl


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J4 is absent, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, and Intermediates of the Disclosure are compounds of Formula II or IX-XII, wherein J5 is selected from the group consisting of —(CH2)m2—, —O—, —N(H)—, and —C(═O)—; m2 is 0 or 1, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, J5 is —(CH2)m2— and m2 is 0.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VI, wherein:


X1 is absent;


A1 is selected from the group consisting phenylenyl and 6-membered heteroarylenyl;


X2 is —C(═O)—;


L is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to X2; and


B1 is B1-2, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-1. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-2. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-3. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-4. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-5. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-6. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-7. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-9. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-10. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-11. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-12. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-13. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-14. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-15. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-16. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-17. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-18. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-19. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-20. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-21. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-22. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-23. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-24. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-25. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XV, below, wherein B1 is B1-26. In another embodiment, R8 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-27. In another embodiment, R8 is hydrogen. In another embodiment, R2f is hydrogen. In another embodiment, R2g is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I, III-VII, or XVI, below, wherein B1 is B1-28. In another embodiment, R8 is hydrogen. In another embodiment, R2f is hydrogen. In another embodiment, R2g is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —O— and —N(H)—; and


B1 is selected from the group consisting of hydrogen, B1-1, B1-2, B1-3, and B1-4, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-1. In another embodiment, B1 is B1-2. In another embodiment, B1 is B1-3. In another embodiment, B1 is B1-1. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, R2a, R2b, and R2c are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —O—;


m2 is 0;


J4 is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to B1;


R7 is selected from the group consisting of hydrogen, halo, cyano, hydroxy, C1-C3 alkyl, and C1-C3 alkoxy; and


B1 is selected from the group consisting of B1-1, B1-2, B1-3, and B1-4, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-1. In another embodiment, B1 is B1-2. In another embodiment, B1 is B1-3. In another embodiment, B1 is B1-1. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, R2a, R2b, and R2c are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen. In another embodiment, J5 is —(CH2)m2— and m2 is 0.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is —(CH2)m2—;


m2 is 0;


J4 is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to B1;


R7 is selected from the group consisting of hydrogen, halo, cyano, hydroxy, C1-C3 alkyl, and C1-C3 alkoxy; and


B1 is selected from the group consisting of B1-15, B1-16, B1-17, B1-18, B1-19, and B1-20, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-15. In another embodiment, B1 is B1-16. In another embodiment, B1 is B1-17. In another embodiment, B1 is B1-18. In another embodiment, B1 is B1-19. In another embodiment, B1 is B1-20. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, R2b and R2c are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is —(CH2)m2—;


m2 is 0;


J4 is selected from the group consisting of:




embedded image


wherein the bond designated with an “*” is attached to B1;


R7 is selected from the group consisting of hydrogen, halo, cyano, hydroxy, C1-C3 alkyl, and C1-C3 alkoxy; and


B1 is selected from the group consisting of B1-21, B1-22, B1-23, B1-24, B1-25, and B1-26, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-21. In another embodiment, B1 is B1-22. In another embodiment, B1 is B1-23. In another embodiment, B1 is B1-24. In another embodiment, B1 is B1-25. In another embodiment, B1 is B1-26. In another embodiment, R2b and R2c are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B1 is selected from the group consisting of B1-5, B1-6, and B1-7, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-5. In another embodiment, B1 is B1-6. In another embodiment, B1 is B1-7. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, m is 1 or 2; and n is 1 or 2. In another embodiment, R2d and R2e are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B1 is selected from the group consisting of B1-27 and B1-28, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-27. In another embodiment, B1 is B1-28. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, R2d and R2e are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R2d and R2e are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R2e and R2f are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R2e and R2f are hydrogen. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B1 is selected from the group consisting of B1-9, B1-10, and B1-11, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-9. In another embodiment, B1 is B1-10. In another embodiment, B1 is B1-11. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, m is 1 or 2; and n is 1 or 2. In another embodiment, o is 1 or 2; and p is 1 or 2. In another embodiment, R2d and R2e are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B1 is selected from the group consisting of B1-12, B1-13, and B1-14, or a pharmaceutically acceptable salt or solvate thereof. In another embodiment, B1 is B1-12. In another embodiment, B1 is B1-13. In another embodiment, B1 is B1-14. In another embodiment, Z is —CH2—. In another embodiment, Z is —C(═O)—. In another embodiment, m is 1 or 2; and n is 1 or 2. In another embodiment, R2d and R2e are independently selected from the group consisting of hydrogen and fluoro. In another embodiment, R3 is hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV:




embedded image


or a pharmaceutically acceptable salt or solvate thereof, wherein:


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of A1-2, A1-3, A1-9, A1-10, A1-11, A1-12, and A1-13;


Z3 and Z4 are independently selected from the group consisting of N and CH;


with the provisos that (i) at least one of Z3 or Z4 is CH; and (ii) y1 and w1 are 1 when Z4 is N;


y, y1, w, and w1 are each independently 0 or 1;


m2 is 0 or 1; and


B1 is selected from the group consisting of B1-1, B1-2, B1-3, B1-4, B1-15, B1-16, B1-17, B1-18, B1-19, B1-20, B1-21, B1-22, B1-23, B1-24, B1-25 and B1-26.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein R1a is methyl.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 0 and w is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 0 and w is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 1 and w is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CH, y1 is 0, and w1 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CH, y1 is 0, and w1 is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein y1 is 1 and w1 is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein m2 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, or a pharmaceutically acceptable salt or solvate thereof, wherein m2 is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI:




embedded image


or a pharmaceutically acceptable salt or solvate thereof, wherein:


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of A1-2, A1-3, A1-9, A1-10, A1-11, A1-12, and A1-13;


y2 and w2 are each independently 0 or 1;


m4 is 0 or 1; and


B1 is selected from the group consisting of B1-5, B1-6, B1-7, B1-9, B1-10, B1-11, B1-12, B1-13, B1-14, B1-27, and B1-28.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein R1a is methyl.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 0 and w2 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 1 and w2 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 1 and w2 is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein m4 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, or a pharmaceutically acceptable salt or solvate thereof, wherein m4 is 1.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein B1 is selected from the group consisting of:




embedded image


or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula XV, wherein B1 is selected from the group consisting of:




embedded image


or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula XVI, wherein B1 is selected from the group consisting of:




embedded image


or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein R3a is halo, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein R3a is C1-C4 alkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein R3a is C1-C4 haloalkyl, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein R3a is selected from the group consisting of —Cl, —CH3, and —CF3, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein Z1 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of any one of Formulae I or III-VII, wherein Z2 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII:




embedded image


or a pharmaceutically acceptable salt or solvate thereof, wherein:


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl;


R2d and R2e are each independently selected from the group consisting of hydrogen and halo;


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen and halo;


w and y are independently 0 or 1;


m1 is 0 or 1; and


Z is selected from the group consisting of —CH2— and —C(═O)—.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein R1a is methyl.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein R2d and R2e are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein R5a, R5b, R5c, and R5d are hydrogen.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein R5b, R5c, and R5d are hydrogen; and R5a is halo.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein R5a, R5b, R5c are hydrogen; and R5d is halo.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein m1 is 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein m1 is 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein w and y are 0.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein w and y are 1.


In another embodiment, Compounds of the Disclosure are compounds of Formula VIII, or a pharmaceutically acceptable salt or solvate thereof, wherein Z is —C(═O)—.


In another embodiment, Compounds of the Disclosure are any one or more of the compounds of Table 1, or a pharmaceutically acceptable salt or solvate thereof.










TABLE 1





Cpd.



No.
Structure







 1


embedded image







 2


embedded image







 3


embedded image







 4


embedded image







 5


embedded image







 6


embedded image







 7


embedded image







 8


embedded image







 9


embedded image







 10


embedded image







 11


embedded image







 12


embedded image







 13


embedded image







 14


embedded image







 15


embedded image







 16


embedded image







 17


embedded image







 18


embedded image







 19


embedded image







 20


embedded image







 21


embedded image







 22


embedded image







 23


embedded image







 24


embedded image







 25


embedded image







 26


embedded image







 27


embedded image







 28


embedded image







 29


embedded image







 30


embedded image







 31


embedded image







 32


embedded image







 33


embedded image







 34


embedded image







 35


embedded image







 36


embedded image







 37


embedded image







 38


embedded image







 39


embedded image







 40


embedded image







 41


embedded image







 42


embedded image







 43


embedded image







 44


embedded image







 45


embedded image







 46


embedded image







 47


embedded image







 48


embedded image







 49


embedded image







 50


embedded image







 51


embedded image







 52


embedded image







 53


embedded image







 54


embedded image







 55


embedded image







 56


embedded image







 57


embedded image







 58


embedded image







 59


embedded image







 60


embedded image







 61


embedded image







 62


embedded image







 63


embedded image







 64


embedded image







 65


embedded image







 66


embedded image







 67


embedded image







 68


embedded image







 69


embedded image







 70


embedded image







 71


embedded image







 72


embedded image







 73


embedded image







 74


embedded image







 75


embedded image







 76


embedded image







 77


embedded image







 78


embedded image







 79


embedded image







 80


embedded image







 81


embedded image







 82


embedded image







 83


embedded image







 84


embedded image







 85


embedded image







 86


embedded image







 87


embedded image







 88


embedded image







 89


embedded image







 90


embedded image







 91


embedded image







 92


embedded image







 93


embedded image







 94


embedded image







 95


embedded image







 96


embedded image







 97


embedded image







 98


embedded image







 99


embedded image







100


embedded image







101


embedded image







102


embedded image







103


embedded image







104


embedded image







105


embedded image







106


embedded image







107


embedded image







108


embedded image







109


embedded image







110


embedded image







111


embedded image







112


embedded image







113


embedded image







114


embedded image







115


embedded image







116


embedded image







117


embedded image







118


embedded image







119


embedded image







120


embedded image







121


embedded image







122


embedded image







123


embedded image







124


embedded image







125


embedded image







126


embedded image







127


embedded image







128


embedded image







129


embedded image







130


embedded image







131


embedded image







132


embedded image







133


embedded image







134


embedded image







135


embedded image







136


embedded image







137


embedded image







138


embedded image







139


embedded image







140


embedded image







141


embedded image







142


embedded image







143


embedded image







144


embedded image







145


embedded image







146


embedded image







147


embedded image







148


embedded image







149


embedded image







150


embedded image







151


embedded image







152


embedded image







153


embedded image







154


embedded image







155


embedded image







156


embedded image







157


embedded image







158


embedded image







159


embedded image







160


embedded image







161


embedded image







162


embedded image







163


embedded image







164


embedded image







165


embedded image







166


embedded image







167


embedded image







168


embedded image







169


embedded image







170


embedded image







171


embedded image







172


embedded image







173


embedded image







174


embedded image







175


embedded image







176


embedded image







177


embedded image







178


embedded image







179


embedded image







180


embedded image







181


embedded image







182


embedded image







183


embedded image







184


embedded image







185


embedded image







186


embedded image







187


embedded image







188


embedded image







189


embedded image







190


embedded image







191


embedded image







192


embedded image







193


embedded image







194


embedded image







195


embedded image







196


embedded image







197


embedded image







198


embedded image







199


embedded image







200


embedded image







201


embedded image







202


embedded image







203


embedded image







204


embedded image







205


embedded image







206


embedded image







207


embedded image







208


embedded image







209


embedded image







210


embedded image







211


embedded image







212


embedded image







213


embedded image







214


embedded image







215


embedded image







216


embedded image







217


embedded image







218


embedded image







219


embedded image







220


embedded image







221


embedded image







222


embedded image







223


embedded image







224


embedded image







225


embedded image







226


embedded image







227


embedded image







228


embedded image







229


embedded image







230


embedded image







231


embedded image







232


embedded image







233


embedded image







234


embedded image







235


embedded image







236


embedded image







237


embedded image







238


embedded image







239


embedded image







240


embedded image







241


embedded image







242


embedded image







243


embedded image







244


embedded image







245


embedded image







246


embedded image







247


embedded image







248


embedded image







249


embedded image







250


embedded image







251


embedded image







252


embedded image







253


embedded image







254


embedded image







255


embedded image







256


embedded image







257


embedded image







258


embedded image







259


embedded image







260


embedded image







261


embedded image







262


embedded image







263


embedded image







264


embedded image







265


embedded image







266


embedded image







267


embedded image







268


embedded image







269


embedded image







270


embedded image







271


embedded image







272


embedded image







273


embedded image







274


embedded image







275


embedded image







276


embedded image







277


embedded image







278


embedded image







279


embedded image







280


embedded image







281


embedded image







282


embedded image







283


embedded image







284


embedded image







285


embedded image







286


embedded image







287


embedded image







288


embedded image







289


embedded image







290


embedded image







291


embedded image







292


embedded image







293


embedded image







294


embedded image







295


embedded image







296


embedded image







297


embedded image







298


embedded image







299


embedded image







300


embedded image







301


embedded image







302


embedded image







303


embedded image







304


embedded image







305


embedded image







306


embedded image







307


embedded image







308


embedded image







309


embedded image







310


embedded image







311


embedded image







312


embedded image







313


embedded image







314


embedded image







315


embedded image







316


embedded image







317


embedded image







318


embedded image







319


embedded image







320


embedded image







321


embedded image







322


embedded image







323


embedded image







324


embedded image







325


embedded image







326


embedded image







327


embedded image







328


embedded image







329


embedded image







330


embedded image







331


embedded image







332


embedded image







333


embedded image







334


embedded image







335


embedded image







336


embedded image







337


embedded image







338


embedded image







339


embedded image







340


embedded image







341


embedded image







342


embedded image







343


embedded image







344


embedded image







345


embedded image







346


embedded image







347


embedded image







348


embedded image







349


embedded image







350


embedded image







351


embedded image







352


embedded image







353


embedded image







354


embedded image







355


embedded image







356


embedded image







357


embedded image







359


embedded image







360


embedded image







361


embedded image







362


embedded image







363


embedded image







364


embedded image







365


embedded image







366


embedded image







367


embedded image







368


embedded image







369


embedded image







370


embedded image







371


embedded image







372


embedded image







373


embedded image







374


embedded image







375


embedded image







376


embedded image







377


embedded image







378


embedded image







379


embedded image







380


embedded image







381


embedded image







382


embedded image







383


embedded image







384


embedded image







385


embedded image







386


embedded image







387


embedded image







388


embedded image







389


embedded image







390


embedded image







391


embedded image







392


embedded image







393


embedded image







394


embedded image







395


embedded image







396


embedded image







397


embedded image







398


embedded image







399


embedded image







400


embedded image







401


embedded image







402


embedded image







403


embedded image







404


embedded image







405


embedded image







406


embedded image







407


embedded image







408


embedded image







409


embedded image







410


embedded image







411


embedded image







412


embedded image







413


embedded image







414


embedded image







415


embedded image







416


embedded image







417


embedded image







418


embedded image







419


embedded image







420


embedded image







421


embedded image







422


embedded image







423


embedded image







424


embedded image







425


embedded image







426


embedded image







427


embedded image







428


embedded image







429


embedded image







430


embedded image







431


embedded image







432


embedded image







433


embedded image







434


embedded image







435


embedded image







436


embedded image







437


embedded image







438


embedded image







439


embedded image







440


embedded image







441


embedded image







442


embedded image







443


embedded image







444


embedded image







445


embedded image







446


embedded image







447


embedded image







448


embedded image







449


embedded image







450


embedded image







451


embedded image







452


embedded image







453


embedded image







454


embedded image







455


embedded image







456


embedded image







457


embedded image







458


embedded image







459


embedded image







460


embedded image







461


embedded image







462


embedded image







463


embedded image







467


embedded image







468


embedded image







469


embedded image







470


embedded image







471


embedded image







472


embedded image







473


embedded image







474


embedded image







475


embedded image







476


embedded image







477


embedded image







478


embedded image







479


embedded image







480


embedded image







481


embedded image







482


embedded image







483


embedded image







484


embedded image







485


embedded image







486


embedded image







487


embedded image







488


embedded image







489


embedded image







490


embedded image







491


embedded image







492


embedded image







493


embedded image







494


embedded image







495


embedded image







496


embedded image







497


embedded image







498


embedded image







499


embedded image







500


embedded image







501


embedded image







502


embedded image







503


embedded image







504


embedded image







505


embedded image







506


embedded image







507


embedded image







508


embedded image







509


embedded image







510


embedded image







511


embedded image







512


embedded image







513


embedded image







514


embedded image







515


embedded image







516


embedded image







517


embedded image







518


embedded image







519


embedded image







520


embedded image







521


embedded image







522


embedded image







523


embedded image







524


embedded image







525


embedded image







526


embedded image







527


embedded image







528


embedded image







529


embedded image







530


embedded image







531


embedded image







532


embedded image







533


embedded image







534


embedded image







535


embedded image







536


embedded image







537


embedded image







538


embedded image







539


embedded image







540


embedded image







541


embedded image







542


embedded image







543


embedded image







544


embedded image







545


embedded image







546


embedded image







547


embedded image







548


embedded image







549


embedded image







550


embedded image







551


embedded image







552


embedded image







553


embedded image







554


embedded image







555


embedded image







556


embedded image







557


embedded image







558


embedded image







559


embedded image







560


embedded image







561


embedded image







562


embedded image







563


embedded image







564


embedded image







565


embedded image







566


embedded image







567


embedded image







568


embedded image







569


embedded image







570


embedded image







571


embedded image







572


embedded image







573


embedded image







574


embedded image







575


embedded image







576


embedded image







577


embedded image







578


embedded image







579


embedded image







580


embedded image







581


embedded image







582


embedded image







583


embedded image







584


embedded image







585


embedded image







586


embedded image







587


embedded image







588


embedded image











In another embodiment, the disclosure provides a pharmaceutical composition comprising a Compound of the Disclosure and a pharmaceutically acceptable carrier or excipient.


Compounds of the Disclosure contain an asymmetric carbon atom. In some embodiments, Compounds of the Disclosure are racemic compounds. In other embodiments, Compounds of the Disclosure are enantiomerically enriched, e.g., the enantiomeric excess or “ee” of the compound is about 5% or more as measured by chiral HPLC. In another embodiment, the ee is about 10%. In another embodiment, the ee is about 20%. In another embodiment, the ee is about 30%. In another embodiment, the ee is about 40%. In another embodiment, the ee is about 50%. In another embodiment, the ee is about 60%. In another embodiment, the ee is about 70%. In another embodiment, the ee is about 80%. In another embodiment, the ee is about 85%. In another embodiment, the ee is about 90%. In another embodiment, the ee is about 91%. In another embodiment, the ee is about 92%. In another embodiment, the ee is about 93%. In another embodiment, the ee is about 94%. In another embodiment, the ee is about 95%. In another embodiment, the ee is about 96%. In another embodiment, the ee is about 97%. In another embodiment, the ee is about 98%. In another embodiment, the ee is about 99%.


In another embodiment, the cereblon binding portion of a Compound of the Disclosure, e.g., B1 is B1-1, B1-2, B1-3, B1-4, B1-5, B1-6, or B1-7, is enantiomerically enriched. In another embodiment, the cereblon binding portion of the molecule is racemic. The present disclosure encompasses all possible stereoisomeric, e.g., diastereomeric, forms of Compounds of the Disclosure. For example, all possible stereoisomers of Compounds of the Disclosure are encompassed when E portion of Formula I is enantiomerically enriched and the cereblon binding portion of the molecule is racemic. When a Compound of the Disclosure is desired as a single enantiomer, it can be obtained either by resolution of the final product or by stereospecific synthesis from either isomerically pure starting material or use of a chiral auxiliary reagent, for example, see Z. Ma et al., Tetrahedron: Asymmetry, 8(6), pages 883-888 (1997). Resolution of the final product, an intermediate, or a starting material can be achieved by any suitable method known in the art. Additionally, in situations where tautomers of the Compounds of the Disclosure are possible, the present disclosure is intended to include all tautomeric forms of the compounds.


The present disclosure encompasses the preparation and use of salts of Compounds of the Disclosure, including pharmaceutically acceptable salts. As used herein, the “pharmaceutically acceptable salt” refers to non-toxic salt forms of Compounds of the Disclosure. See e.g., Gupta et al., Molecules 23:1719 (2018). Salts of Compounds of the Disclosure can be prepared during the final isolation and purification of the compounds or separately by reacting the compound with an acid having a suitable cation. The pharmaceutically acceptable salts of Compounds of the Disclosure can be acid addition salts formed with pharmaceutically acceptable acids. Examples of acids which can be employed to form pharmaceutically acceptable salts include inorganic acids such as nitric, boric, hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Nonlimiting examples of salts of compounds of the disclosure include, but are not limited to, the hydrochloride, hydrobromide, hydroiodide, sulfate, bisulfate, 2-hydroxyethansulfonate, phosphate, hydrogen phosphate, acetate, adipate, alginate, aspartate, benzoate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerolphosphate, hemisulfate, heptanoate, hexanoate, formate, succinate, fumarate, maleate, ascorbate, isethionate, salicylate, methanesulfonate, mesitylenesulfonate, naphthylenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproprionate, picrate, pivalate, propionate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, paratoluenesulfonate, undecanoate, lactate, citrate, tartrate, gluconate, methanesulfonate, ethanedisulfonate, benzene sulfonate, and p-toluenesulfonate salts. In addition, available amino groups present in the compounds of the disclosure can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides; and benzyl and phenethyl bromides. In light of the foregoing, any reference Compounds of the Disclosure appearing herein is intended to include the actual compound as well as pharmaceutically acceptable salts, hydrates, or solvates thereof.


The present disclosure also encompasses the preparation and use of solvates of Compounds of the Disclosure. Solvates typically do not significantly alter the physiological activity or toxicity of the compounds, and as such may function as pharmacological equivalents. The term “solvate” as used herein is a combination, physical association and/or solvation of a compound of the present disclosure with a solvent molecule such as, e.g. a disolvate, monosolvate or hemisolvate, where the ratio of solvent molecule to compound of the present disclosure is about 2:1, about 1:1 or about 1:2, respectively. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances, the solvate can be isolated, such as when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid. Thus, “solvate” encompasses both solution-phase and isolatable solvates. Compounds of the Disclosure can be present as solvated forms with a pharmaceutically acceptable solvent, such as water, methanol, and ethanol, and it is intended that the disclosure includes both solvated and unsolvated forms of Compounds of the Disclosure. One type of solvate is a hydrate. A “hydrate” relates to a particular subgroup of solvates where the solvent molecule is water. Solvates typically can function as pharmacological equivalents. Preparation of solvates is known in the art. See, for example, M. Caira et al, J. Pharmaceut. Sci., 93(3):601-611 (2004), which describes the preparation of solvates of fluconazole with ethyl acetate and with water. Similar preparation of solvates, hemisolvates, hydrates, and the like are described by E. C. van Tonder et al., AAPS Pharm. Sci. Tech., 5(1):Article 12 (2004), and A. L. Bingham et al., Chem. Commun. 603-604 (2001). A typical, non-limiting, process of preparing a solvate would involve dissolving a Compound of the Disclosure in a desired solvent (organic, water, or a mixture thereof) at temperatures above 20° C. to about 25° C., then cooling the solution at a rate sufficient to form crystals, and isolating the crystals by known methods, e.g., filtration. Analytical techniques such as infrared spectroscopy can be used to confirm the presence of the solvent in a crystal of the solvate.


In another aspect, the present disclosure provides the following particular embodiments drawn to Compounds of the Disclosure, referred to as “CD Embodiment 1,” “CD Embodiment 2,” “CD Embodiment 3,” and so on.


CD Embodiment 1. A compound of Formula I, see above, wherein:


R3a is selected from the group consisting of halo, C1-C4 alkyl, and C1-C4 haloalkyl;


Z1 is selected from the group consisting of ═C(H)— and ═N—;


Z2 is selected from the group consisting of ═C(R3b)— and ═N—;


R3b is selected from the group consisting of hydrogen, halo, C1-C4 alkyl, and C1-C4 haloalkyl;


E is a spiroheterocyclenyl;


X1 is selected from the group consisting of —C(═O)—, —S(═O)2—, and —CR4aR4b—; or


X1 is absent;


R4a and R4b are independently selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl;


X2 is selected from the group consisting of —C(═O)—, —S(═O)2—, —O—, and —CR4cR4d—; or


X2 is absent;


R4c and R4d are independently selected from the group consisting of hydrogen and C1-3 alkyl;


L is -J1-J2-J3-J4-J5-,


wherein J1 is attached to X2;


J1 is selected from the group consisting of cycloalkylenyl and heterocyclenyl; or


J1 is absent;


J2 is selected from the group consisting of —(CH2)m1—, —CH═CH—, and —C≡C—;


m1 is 0, 1, 2, or 3;


J3 is selected from the group consisting of alkylenyl, heteroalkylenyl, cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl; or


J3 is absent;


J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl; or


J4 is absent;


J5 is selected from the group consisting of —(CH2)m2—, —O—, —N(R6)—, and —C(═O)—;


m2 is 0, 1, 2, or 3;


R6 is selected from the group consisting of hydrogen and C1-C3 alkyl;


B1 is selected from the group consisting of B1-1, B1-2, B1-3, B1-4, B1-5, B1-6, B1-7, B1-9, B1-10, B1-1, B1-12, B1-13, B1-14, B1-15, B1-16, B1-17, B1-18, B1-19, B1-20, B1-21, B1-22, B1-23, B1-24, B1-25, and B1-26, see above;


R2a, R2b, R2c, R2d, and R2e are independently selected from the group consisting of hydrogen, halo, C1-C3 alkyl, and C1-C3 alkoxy; or


R3 is selected from the group consisting of hydrogen, deuterium, fluoro, and C1-C3alkyl;


m is 1, 2, or 3;


n is 1, 2, or 3;


o is 1, 2, or 3;


p is 1, 2, or 3;


Z is selected from the group consisting of —CR1jR1k— and —C(═O)—;


R1j and R1k are independently selected from the group consisting of hydrogen and C1-C3 alkyl; or R1j and R1k taken together with the carbon to which they are attached from a C3-C6 cycloalkyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 2. The compound of CD Embodiment 1, wherein E is selected from the group consisting of E-1, E-2, and E-3, see above;


wherein the bond designated with an “*” is attached to X1;


o and p are independently 0 or 1;


q and r are independently 0, 1, 2, or 3;


wherein the sum of o, p, q, and r is 2, 3, 4, 5, 6, or 7;


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


t, u, v, w, and x are independently 0, 1, 2, or 3;


R1a and R1b are independently selected from the group consisting of hydrogen, C1-C3 alkyl, C1-C4 haloalkyl, optionally substituted C3-C6 cycloalkyl, and (C3-C6 cycloalkyl)C1-C6 alkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted C3-C6 cycloalkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted 4- to 6-membered heterocyclo;


R1c and R1d are independently selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group;


each R1f is independently C1-C3 alkyl;


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


each R1f is independently C1-C3 alkyl;


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


each R1g is independently C1-C3 alkyl; and


h is 0, 1, 2, 3, or 4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 3. The compound of CD Embodiment 2, wherein E is E-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 4. The compound of CD Embodiment 3, wherein E-1 is selected from the group consisting of E-1-1 and E-1-2, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 5. The compound of CD Embodiment 4, wherein E-1 is E-1-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 6. The compound of CD Embodiment 5, wherein R1a and R1b are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 7. The compound of CD Embodiment 6, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 8. The compound of CD Embodiment 6, wherein q is 2; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 9. The compound of CD Embodiment 6, wherein q is 1; r is 0; s is 0; and t is 2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 10. The compound of CD Embodiment 6, wherein q is 0; r is 1; s is 1; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 11. The compound of CD Embodiment 6, wherein q is 1; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 12. The compound of CD Embodiment 5, wherein R1a and R1b are independently C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 13. The compound of CD Embodiment 12, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 14. The compound of CD Embodiment 5, wherein R1a is C1-C3 alkyl; and R1b is hydrogen or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 15. The compound of CD Embodiment 14, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 16. The compound of CD Embodiment 15 of Formula III, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 17. The compound of CD Embodiment 15 of Formula IV, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 18. The compound of CD Embodiment 5, wherein R1a and R1b taken together with the carbon atom to which they are attached form a —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 19. The compound of CD Embodiment 18, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 20. The compound of CD Embodiment 4, wherein:


E-1 is E-1-2;


R1c is C1-C3 alkyl;


R1d is selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form a —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 21. The compound of CD Embodiment 20, wherein R1d is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 22. The compound of CD Embodiment 21 of Formula V, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 23. The compound of CD Embodiment 21 of Formula VI, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 24. The compound of CD Embodiment 20, wherein R1c and R1d taken together with the carbon atom to which they are attached form a —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 25. The compound of CD Embodiment 2, wherein E is E-2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 26. The compound of CD Embodiment 25, wherein E-2 is E-2-1, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 27. The compound of CD Embodiment 2, wherein E is E-3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 28. The compound of CD Embodiment 27, wherein E-3 is E-3-1, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 29. The compound of any one of CD Embodiments 1-26, wherein X1 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 30. The compound of any one of CD Embodiments 1-26, wherein X1 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 31. The compound of any one of CD Embodiments 1-26, wherein X1 is —CR4aR4b—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 32. The compound of CD Embodiments 31, wherein R4a and R4b are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 33. The compound of any one of CD Embodiments 1-28, wherein X1 is absent, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 34. The compound of any one of CD Embodiments 1-33, wherein:


A1 is selected from the group consisting of A1-1, A1-2, A1-3, A1-4, A1-5, A1-6, A1-7, A1-8, A1-9, A1-10, A1-11, A1-12, and A1-13 see above, wherein the bond designated with an “*” is attached to X2.


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen, halo, C1-C3 alkyl, and C1-C3 alkoxy


e is 0, 1, or 2; and


f is 0, 1, or 2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 35. The compound of CD Embodiment 34, wherein A1 is A1-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 36. The compound of CD Embodiment 34, wherein A1 is A1-2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 37. The compound of CD Embodiment 34, wherein A1 is A1-3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 38. The compound of CD Embodiment 34, wherein A1 is A1-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 39. The compound of CD Embodiment 34, wherein A1 is A1-5, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 40. The compound of CD Embodiment 34, wherein A1 is A1-6, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 41. The compound of CD Embodiment 34, wherein A1 is A1-7, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 42. The compound of any one of CD Embodiments 34-36, wherein R5a, R5b, R5c, and R5d are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 43. The compound of any one of CD Embodiments 1-42, wherein X2 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 44. The compound of any one of CD Embodiments 1-42, wherein X2 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 45. The compound of any one of CD Embodiments 1-42, wherein X2 is —O—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 46. The compound of any one of CD Embodiments 1-42, wherein X2 is —CR4cR4d—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 47. The compound of CD Embodiment 46, wherein R4c and R4d are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 48. The compound of any one of CD Embodiment 1-42, wherein X2 is absent, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 49. The compound of any one of CD Embodiments 1-48, wherein J1 is cycloalkylenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 50. The compound of any one of CD Embodiments 1-48, wherein J1 is heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 51. The compound of CD Embodiment 20, wherein J1 is selected from the group consisting of J1-1, J1-2, J1-3, J1-4, J1-5, J1-6, J1-7, J1-8, J1-9, J1-10, J1-11, J1-12, and J1-13, see above, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 52. The compound of CD Embodiment 51, wherein J1 is J1-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 53. The compound of CD Embodiment 51, wherein J1 is J1-2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 54. The compound of CD Embodiment 51, wherein J1 is J1-3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 55. The compound of CD Embodiment 51, wherein J1 is J1-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 56. The compound of CD Embodiment 51, wherein J1 is J1-5, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 57. The compound of CD Embodiment 51, wherein J1 is J1-6, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 58. The compound of CD Embodiment 51, wherein J1 is J1-7, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 59. The compound of CD Embodiment 51, wherein J1 is J1-8, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 60. The compound of CD Embodiment 51, wherein J1 is J1-9, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 61. The compound of CD Embodiment 51, wherein J1 is J1-10, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 62. The compound of CD Embodiment 51, wherein J1 is J1-11, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 63. The compound of CD Embodiment 51, wherein J1 is J1-12, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 64. The compound of CD Embodiment 51, wherein J1 is J1-13, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 65. The compound of any one of CD Embodiments, 1-48, wherein J1 is absent, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 66. The compound of any one of CD Embodiments 1-65, wherein J2 is selected from the group consisting of —(CH2)m1— and —C≡C—; and m1 is 0, 1, or 2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 67. The compound of CD Embodiment 66, wherein J2 is —(CH2)m1—; and m1 is 0, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 68. The compound of CD Embodiment 66, wherein J2 is —(CH2)m1—; and m1 is 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 69. The compound of CD Embodiment 66, wherein J2 is —C≡C—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 70. The compound of any one of CD Embodiments 1-69, wherein J3 is selected from the group consisting of cycloalkylenyl and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 71. The compound of CD Embodiment 70, wherein J3 is cycloalkylenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 72. The compound of CD Embodiment 70, wherein J3 is heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 73. The compound of any one of CD Embodiments 1-69, wherein J3 is absent, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 74. The compound of any one of CD Embodiments 1-73, wherein J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 75. The compound of CD Embodiment 74, wherein J4 is alkylenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 76. The compound of CD Embodiment 74, wherein J4 is cycloalkylenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 77. The compound of CD Embodiment 74, wherein J4 is heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 78. The compound of any one of CD Embodiments 1-73, wherein J4 is absent, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 79. The compound of any one of CD Embodiments 1-78, wherein:


J5 is selected from the group consisting of —O— and —N(H)—; and


B1 is selected from the group consisting of B1-1, B1-2, B1-3, and B1-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 80. The compound of any one of CD Embodiments 1-77, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —O—;


m2 is 0;


J4 is selected from the group consisting of J4-1, J4-2, J4-3, J4-4, J4-5, and J4-6, see above, wherein the bond designated with an “*” is attached to B1;


R7 is selected from the group consisting of hydrogen, halo, cyano, hydroxy, C1-C3 alkyl, and C1-C3 alkoxy; and


B1 is selected from the group consisting of B1-1, B1-2, B1-3, and B1-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 81. The compound of CD Embodiment 80, wherein J4 is J4-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 82. The compound of CD Embodiment 80, wherein J4 is J4-2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 83. The compound of CD Embodiment 80, wherein J4 is J4-3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 84. The compound of CD Embodiment 80, wherein J4 is J4-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 85. The compound of CD Embodiment 80, wherein J4 is J4-5, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 86. The compound of CD Embodiment 80, wherein J4 is J4-6, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 87. The compound of any one of CD Embodiments 79-86, wherein B1 is B1-1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 88. The compound of CD Embodiment 87, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 89. The compound of CD Embodiment 87, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 90. The compound of any one of CD Embodiments 79-86, wherein B1 is B1-2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 91. The compound of CD Embodiment 90, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 92. The compound of CD Embodiment 90, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof


CD Embodiment 93. The compound of any one of CD Embodiments 79-86, wherein B1 is B1-3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 94. The compound of any one of CD Embodiments 79-86, wherein B1 is B1-4, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 95. The compound of any one of CD Embodiments 79-94, wherein R2a, R2b, and R2c are independently selected from the group consisting of hydrogen and fluoro, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 96. The compound of CD Embodiment 95, wherein R2a, R2b, and R2c are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 97. The compound of any one of CD Embodiments 1-78, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B1 is selected from the group consisting of B1-5, B1-6, B1-7, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 98. The compound of CD Embodiment 97, wherein B1 is B1-5, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 99. The compound of CD Embodiment 98, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 100. The compound of CD Embodiment 98, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 101. The compound of any one of CD Embodiments 98-100, wherein m is 1 or 2; and n is 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 102. The compound of CD Embodiment 97, wherein B1 is B1-6, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 103. The compound of CD Embodiment 102, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 104. The compound of CD Embodiment 102, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 105. The compound of any one of CD Embodiment 102-104, wherein m is 1 or 2; and n is 1 or 2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 106. The compound of CD Embodiment 97, wherein B1 is B1-7, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 107. The compound of CD Embodiment 106, wherein m is 1 or 2; and n is 1 or 2, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 108. The compound of any one of CD Embodiments 97-107, wherein R2d and R2e are independently selected from the group consisting of hydrogen and fluoro, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 109. The compound of CD Embodiment 108, wherein R2d and R2e are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 110. The compound of any one of CD Embodiments 79-108, wherein R3 is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 111. The compound of any one of CD Embodiments 1-110, wherein R3a is halo, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 112. The compound of any one of CD Embodiments 1-110, wherein R3a is C1-C4 alkyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 113. The compound of any one of CD Embodiments 1-110, wherein R3a is C1-C4 haloalkyl, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 114 The compound of any one of CD Embodiments 1-110, wherein R3a is selected from the group consisting of —Cl, —CH3, and —CF3, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 115. The compound of any one of CD Embodiments 1-114, wherein Z1 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 116. The compound of any one of CD Embodiment 1-115, wherein Z2 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 117. The compound of CD Embodiment 1 that is any one or more of the compounds of Table 1, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 118. The compound of CD Embodiment 34, wherein A1 is A1-8, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 119. The compound of CD Embodiment 34, wherein A1 is A1-9, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 120. The compound of CD Embodiment 34, wherein A1 is A1-10, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 121. The compound of CD Embodiment 34, wherein A1 is A1-11, or a pharmaceutically acceptable salt or solvate thereof. CD Embodiment 122.


The compound of CD Embodiment 34, wherein A1 is A1-12, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 123. The compound of CD Embodiment 34, wherein A1 is A1-13, or a pharmaceutically acceptable salt or solvate thereof.


CD Embodiment 124. The compound of CD Embodiment 1 of Formula XV, see above, or a pharmaceutically acceptable salt or solvate thereof, wherein:


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of A1-2, A1-3, A1-9, A1-10, A1-11, A1-12, and A1-13 see above;


Z3 and Z4 are independently selected from the group consisting of N and CH;


with the provisos that (i) at least one of Z3 or Z4 is CH; and (ii) y1 and w1 are 1 when Z4 is N;


y, y1, w, and w1 are each independently 0 or 1;


m2 is 0 or 1; and


B1 is selected from the group consisting of B1-1, B1-2, B1-3, B1-4, B1-15, B1-16, B1-17, B1-18, B1-19, B1-20, B1-21, B1-22, B1-23, B1-24, B1-25 and B1-26, see above.


CD Embodiment 125. The compound of CD Embodiment 124, or a pharmaceutically acceptable salt or solvate thereof, wherein R1a is methyl.


CD Embodiment 126. The compound of CD Embodiments 124 or 125, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 0 and w is 0.


CD Embodiment 127. The compound of CD Embodiments 124 or 125, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 0 and w is 1.


CD Embodiment 128. The compound of CD Embodiments 124 or 125, or a pharmaceutically acceptable salt or solvate thereof, wherein y is 1 and w is 1.


CD Embodiment 129. The compound of any one of CD Embodiments 124-128, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CH, y1 is 0, and w1 is 0.


CD Embodiment 130. The compound of any one of CD Embodiments 124-128, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CH, y1 is 0, and w1 is 1.


CD Embodiment 131. The compound of any one of CD Embodiments 124-128, or a pharmaceutically acceptable salt or solvate thereof, wherein Z4 is CH, y1 is 1, and w1 is 1.


CD Embodiment 132. The compound of any one of CD Embodiments 124-131, or a pharmaceutically acceptable salt or solvate thereof, wherein m2 is 0.


CD Embodiment 133. The compound of any one of CD Embodiments 124-131, or a pharmaceutically acceptable salt or solvate thereof, wherein m2 is 1.


CD Embodiment 134. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-1.


CD Embodiment 135. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-2.


CD Embodiment 136. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-3.


CD Embodiment 137. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-9.


CD Embodiment 138. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-10.


CD Embodiment 139. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-11.


CD Embodiment 140. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-12.


CD Embodiment 141. The compound of any one of CD Embodiments 124-133, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-13.


CD Embodiment 142. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-1.


CD Embodiment 143. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-2.


CD Embodiment 144. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-3.


CD Embodiment 145. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-4.


CD Embodiment 146. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-15.


CD Embodiment 147. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-16.


CD Embodiment 148. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-17.


CD Embodiment 149. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-18.


CD Embodiment 150. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-19.


CD Embodiment 151. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-20.


CD Embodiment 152. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-21.


CD Embodiment 153. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-22.


CD Embodiment 154. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-23.


CD Embodiment 155. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-24.


CD Embodiment 156. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-25.


CD Embodiment 157. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-26.


CD Embodiment 158. The compound of any one of CD Embodiments 124-141, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is selected from the group consisting of:




embedded image


CD Embodiment 159. The compound of CD Embodiment 1 of Formula XVI, see above, or a pharmaceutically acceptable salt or solvate thereof, wherein:


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of A1-2, A1-3, A1-9, A1-10, A1-11, A1-12, and A1-13, see above;


y2 and w2 are each independently 0 or 1;


m4 is 0 or 1; and


B1 is selected from the group consisting of B1-5, B1-6, B1-7, B1-9, B1-10, B1-11, B1-12, B1-13, B1-14, B1-27, and B1-28, see above.


CD Embodiment 160. The compound of CD Embodiment 159, or a pharmaceutically acceptable salt or solvate thereof, wherein R1a is methyl.


CD Embodiment 161. The compound of CD Embodiments 159 or 160, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 0 and w2 is 0.


CD Embodiment 162. The compound of CD Embodiments 159 or 160, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 0 and w2 is 1.


CD Embodiment 163. The compound of CD Embodiments 159 or 160, or a pharmaceutically acceptable salt or solvate thereof, wherein y2 is 1 and w2 is 1.


CD Embodiment 164. The compound of any one of CD Embodiments 159-163, or a pharmaceutically acceptable salt or solvate thereof, wherein m4 is 0.


CD Embodiment 165. The compound of any one of CD Embodiments 159-163, or a pharmaceutically acceptable salt or solvate thereof, wherein m4 is 1.


CD Embodiment 166. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-1.


CD Embodiment 167. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-2.


CD Embodiment 168. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-3.


CD Embodiment 169. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-9.


CD Embodiment 170. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-10.


CD Embodiment 171. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-11.


CD Embodiment 172. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-12.


CD Embodiment 173. The compound of any one of CD Embodiments 159-165, or a pharmaceutically acceptable salt or solvate thereof, wherein A1 is A1-13.


CD Embodiment 174. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-5.


CD Embodiment 175. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-6.


CD Embodiment 176. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-7.


CD Embodiment 177. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-8.


CD Embodiment 178. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-9.


CD Embodiment 179. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-10.


CD Embodiment 180. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-11.


CD Embodiment 181. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-12.


CD Embodiment 182. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-13.


CD Embodiment 183. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-14.


CD Embodiment 184. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-27.


CD Embodiment 185. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is B1-28.


CD Embodiment 186. The compound of any one of CD Embodiments 159-173, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is selected from the group consisting of:




embedded image


II. Therapeutic Methods of the Disclosure

Compounds of the Disclosure degrade AR protein and are thus useful in the treatment of a variety of diseases and conditions. In particular, Compounds of the Disclosure are useful in methods of treating a disease or condition wherein degradation AR proteins provides a benefit, for example, cancers and proliferative diseases. The therapeutic methods of the disclosure comprise administering a therapeutically effective amount of a Compound of the Disclosure to a subject, e.g., a cancer patient, in need thereof. The present methods also encompass administering a second therapeutic agent to the subject in combination with the Compound of the Disclosure. The second therapeutic agent is selected from drugs known as useful in treating the disease or condition afflicting the individual in need thereof, e.g., a chemotherapeutic agent and/or radiation known as useful in treating a particular cancer.


The present disclosure provides Compounds of the Disclosure as AR protein degraders for the treatment of a variety of diseases and conditions wherein degradation of AR proteins has a beneficial effect. Compounds of the Disclosure typically have DC50 (the drug concentration that results in 50% AR protein degradation) values of less than 100 μM, e.g., less than 50 μM, less than 25 μM, and less than 5 μM, less than about 1 μM, less than about 0.5 μM, or less than about 0.1 μM. In some embodiments, Compounds of the Disclosure typically have DC50 values of less than about 0.01 μM. In some embodiments, Compounds of the Disclosure typically have DC50 values of less than about 0.001 μM. In one embodiment, the present disclosure relates to a method of treating an individual suffering from a disease or condition wherein degradation of AR proteins provides a benefit comprising administering a therapeutically effective amount of a Compound of the Disclosure to an individual in need thereof.


Since Compounds of the Disclosure are degraders of AR protein, a number of diseases and conditions mediated by AR can be treated by employing these compounds. The present disclosure is thus directed generally to a method for treating a condition or disorder responsive to degradation of AR in an animal, e.g., a human, suffering from, or at risk of suffering from, the condition or disorder, the method comprising administering to the animal an effective amount of one or more Compounds of the Disclosure.


The present disclosure is further directed to a method of degrading AR protein in a subject in need thereof, said method comprising administering to the subject an effective amount of at least one Compound of the Disclosure.


In another aspect, the present disclosure provides a method of treating cancer in a subject comprising administering a therapeutically effective amount of a Compound of the Disclosure. While not being limited to a specific mechanism, in some embodiments, Compounds of the Disclosure treat cancer by degrading AR. Examples of treatable cancers include, but are not limited to, any one or more of the cancers of Table 2.












TABLE 2







adrenal cancer
acinic cell carcinoma
acoustic neuroma
acral lentigious





melanoma


acrospiroma
acute eosinophilic
acute erythroid
acute lymphoblastic



leukemia
leukemia
leukemia


acute
acute monocytic
acute promyelocytic
adenocarcinoma


megakaryoblastic
leukemia
leukemia



leukemia





adenoid cystic
adenoma
adenomatoid
adenosquamous


carcinoma

odontogenic tumor
carcinoma


adipose tissue
adrenocortical
adult T-cell
aggressive NK-cell


neoplasm
carcinoma
leukemia/lymphoma
leukemia


AIDS-related
alveolar
alveolar soft part
ameloblastic


lymphoma
rhabdomyosarcoma
sarcoma
fibroma


anaplastic large cell
anaplastic thyroid
angioimmunoblastic
angiomyolipoma


lymphoma
cancer
T-cell lymphoma



angiosarcoma
astrocytoma
atypical teratoid
B-cell chronic




rhabdoid tumor
lymphocytic





leukemia


B-cell
B-cell lymphoma
basal cell carcinoma
biliary tract cancer


prolymphocytic





leukemia





bladder cancer
blastoma
bone cancer
Brenner tumor


Brown tumor
Burkitt's lymphoma
breast cancer
brain cancer


carcinoma
carcinoma in situ
carcinosarcoma
cartilage tumor


cementoma
myeloid sarcoma
chondroma
chordoma


choriocarcinoma
choroid plexus
clear-cell sarcoma of
craniopharyngioma



papilloma
the kidney



cutaneous T-cell
cervical cancer
colorectal cancer
Degos disease


lymphoma





desmoplastic small
diffuse large B-cell
dysembryoplastic
dysgerminoma


round cell tumor
lymphoma
neuroepithelial





tumor



embryonal
endocrine gland
endodermal sinus
enteropathy-


carcinoma
neoplasm
tumor
associated T-cell





lymphoma


esophageal cancer
fetus in fetu
fibroma
fibrosarcoma


follicular
follicular thyroid
ganglioneuroma
gastrointestinal


lymphoma
cancer

cancer


germ cell tumor
gestational
giant cell
giant cell tumor of



choriocarcinoma
fibroblastoma
the bone


glial tumor
glioblastoma
glioma
gliomatosis cerebri



multiforme




glucagonoma
gonadoblastoma
granulosa cell tumor
gynandroblastoma


gallbladder cancer
gastric cancer
hairy cell leukemia
hemangioblastoma


head and neck
hemangiopericytoma
hematological
hepatoblastoma


cancer

cancer



hepatosplenic T-cell
Hodgkin's
non-Hodgkin's
invasive lobular


lymphoma
lymphoma
lymphoma
carcinoma


intestinal cancer
kidney cancer
laryngeal cancer
lentigo maligna


lethal midline
leukemia
leydig cell tumor
liposarcoma


carcinoma





lung cancer
lymphangioma
lymphangiosarcoma
lymphoepithelioma


lymphoma
acute lymphocytic
acute myelogeous
chronic



leukemia
leukemia
lymphocytic





leukemia


liver cancer
small cell lung
non-small cell lung
MALT lymphoma



cancer
cancer



malignant fibrous
malignant peripheral
malignant triton
mantle cell


histiocytoma
nerve sheath tumor
tumor
lymphoma


marginal zone B-
mast cell leukemia
mediastinal germ
medullary


cell lymphoma

cell tumor
carcinoma of the





breast


medullary thyroid
medulloblastoma
melanoma
meningioma


cancer





merkel cell cancer
mesothelioma
metastatic urothelial
mixed Mullerian




carcinoma
tumor


mucinous tumor
multiple myeloma
muscle tissue
mycosis fungoides




neoplasm



myxoid
myxoma
myxosarcoma
nasopharyngeal


liposarcoma


carcinoma


neurinoma
neuroblastoma
neurofibroma
neuroma


nodular melanoma
ocular cancer
oligoastrocytoma
oligodendroglioma


oncocytoma
optic nerve sheath
optic nerve tumor
oral cancer



meningioma




osteosarcoma
ovarian cancer
Pancoast tumor
papillary thyroid





cancer


paraganglioma
pinealoblastoma
pineocytoma
pituicytoma


pituitary adenoma
pituitary tumor
plasmacytoma
polyembryoma


precursor T-
primary central
primary effusion
preimary peritoneal


lymphoblastic
nervous system
lymphoma
cancer


lymphoma
lymphoma




prostate cancer
pancreatic cancer
pharyngeal cancer
pseudomyxoma





periotonei


renal cell carcinoma
renal medullary
retinoblastoma
rhabdomyoma



carcinoma




rhabdomyosarcoma
Richter's
rectal cancer
sarcoma



transformation




Schwannomatosis
seminoma
Sertoli cell tumor
sex cord-gonadal





stromal tumor


signet ring cell
skin cancer
small blue round cell
small cell


carcinoma

tumors
carcinoma


soft tissue sarcoma
somatostatinoma
soot wart
spinal tumor


splenic marginal
squamous cell
synovial sarcoma
Sezary's disease


zone lymphoma
carcinoma




small intestine
squamous carcinoma
stomach cancer
T-cell lymphoma


cancer





testicular cancer
thecoma
thyroid cancer
transitional cell





carcinoma


throat cancer
urachal cancer
urogenital cancer
urothelial





carcinoma


uveal melanoma
uterine cancer
verrucous carcinoma
visual pathway





glioma


vulvar cancer
vaginal cancer
Waldenstrom's
Warthin's tumor




macroglobulinemia



Wilms' tumor









In another embodiment, the cancer is a solid tumor. In another embodiment, the cancer a hematological cancer. Exemplary hematological cancers include, but are not limited to, the cancers listed in Table 3. In another embodiment, the hematological cancer is acute lymphocytic leukemia, chronic lymphocytic leukemia (including B-cell chronic lymphocytic leukemia), or acute myeloid leukemia.










TABLE 3







acute lymphocytic leukemia (ALL)
acute eosinophilic leukemia


acute myeloid leukemia (AML)
acute erythroid leukemia


chronic lymphocytic leukemia (CLL)
acute lymphoblastic leukemia


small lymphocytic lymphoma (SLL)
acute megakaryoblastic leukemia


multiple myeloma (MM)
acute monocytic leukemia


Hodgkins lymphoma (HL)
acute promyelocytic leukemia


non-Hodgkin's lymphoma (NHL)
acute myelogeous leukemia


mantle cell lymphoma (MCL)
B-cell prolymphocytic leukemia


marginal zone B-cell lymphoma
B-cell lymphoma


splenic marginal zone lymphoma
MALT lymphoma


follicular lymphoma (FL)
precursor T-lymphoblastic lymphoma


Waldenstrom's macroglobulinemia (WM)
T-cell lymphoma


diffuse large B-cell lymphoma (DLBCL)
mast cell leukemia


marginal zone lymphoma (MZL)
adult T cell leukemia/lymphoma


hairy cell leukemia (HCL)
aggressive NK-cell leukemia


Burkitt's lymphoma (BL)
angioimmunoblastic T-cell lymphoma


Richter's transformation









In another embodiment, the cancer is a leukemia, for example a leukemia selected from acute monocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia and mixed lineage leukemia (MLL). In another embodiment the cancer is NUT-midline carcinoma. In another embodiment the cancer is multiple myeloma. In another embodiment the cancer is a lung cancer such as small cell lung cancer (SCLC). In another embodiment the cancer is a neuroblastoma. In another embodiment the cancer is Burkitt's lymphoma. In another embodiment the cancer is cervical cancer. In another embodiment the cancer is esophageal cancer. In another embodiment the cancer is ovarian cancer. In another embodiment the cancer is colorectal cancer. In another embodiment, the cancer is prostate cancer. In another embodiment, the cancer is breast cancer.


In another embodiment, the cancer is selected from the group consisting of acute monocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia mixed lineage leukemia, NUT-midline carcinoma, multiple myeloma, small cell lung cancer, non-small cell lung cancer, neuroblastoma, Burkitt's lymphoma, cervical cancer, esophageal cancer, ovarian cancer, colorectal cancer, prostate cancer, breast cancer, bladder cancer, ovary cancer, glioma, sarcoma, esophageal squamous cell carcinoma, and papillary thyroid carcinoma.


In another embodiment, Compounds of the Disclosure are administered to a subject in need thereof to treat breast cancer or prostate cancer. In another embodiment, the cancer is breast cancer. In another embodiment, the cancer is prostate cancer. In another embodiment, the cancer is metastatic castration-resistant prostate cancer.


The methods of the present disclosure can be accomplished by administering a Compound of the Disclosure as the neat compound or as a pharmaceutical composition. Administration of a pharmaceutical composition, or neat Compound of the Disclosure, can be performed during or after the onset of the disease or condition of interest. Typically, the pharmaceutical compositions are sterile, and contain no toxic, carcinogenic, or mutagenic compounds that would cause an adverse reaction when administered.


In one embodiment, a Compound of the Disclosure is administered as a single agent to treat a disease or condition wherein degradation of AR protein provides a benefit. In another embodiment, a Compound of the Disclosure is administered in conjunction with a second therapeutic agent useful in the treatment of a disease or condition wherein degradation of AR protein provides a benefit. The second therapeutic agent is different from the Compound of the Disclosure. A Compound of the Disclosure and the second therapeutic agent can be administered simultaneously or sequentially to achieve the desired effect. In addition, the Compound of the Disclosure and second therapeutic agent can be administered as a single pharmaceutical composition or two separate pharmaceutical compositions.


The second therapeutic agent is administered in an amount to provide its desired therapeutic effect. The effective dosage range for each second therapeutic agent is known in the art, and the second therapeutic agent is administered to an individual in need thereof within such established ranges.


A Compound of the Disclosure and the second therapeutic agent can be administered together as a single-unit dose or separately as multi-unit doses, wherein the Compound of the Disclosure is administered before the second therapeutic agent or vice versa. One or more doses of the Compound of the Disclosure and/or one or more doses of the second therapeutic agent can be administered. The Compound of the Disclosure therefore can be used in conjunction with one or more second therapeutic agents, for example, but not limited to, anticancer agents.


In methods of the present disclosure, a therapeutically effective amount of a Compound of the Disclosure, typically formulated in accordance with pharmaceutical practice, is administered to a subject, e.g., a human cancer patient, in need thereof. Whether such a treatment is indicated depends on the individual case and is subject to medical assessment (diagnosis) that takes into consideration signs, symptoms, and/or malfunctions that are present, the risks of developing particular signs, symptoms and/or malfunctions, and other factors.


A Compound of the Disclosure can be administered by any suitable route, for example by oral, buccal, inhalation, sublingual, rectal, vaginal, intracisternal or intrathecal through lumbar puncture, transurethral, nasal, percutaneous, i.e., transdermal, or parenteral (including intravenous, intramuscular, subcutaneous, intracoronary, intradermal, intramammary, intraperitoneal, intraarticular, intrathecal, retrobulbar, intrapulmonary injection and/or surgical implantation at a particular site) administration. Parenteral administration can be accomplished using a needle and syringe or using a high pressure technique.


Pharmaceutical compositions include those wherein a Compound of the Disclosure is administered in an effective amount to achieve its intended purpose. The exact formulation, route of administration, and dosage is determined by an individual physician in view of the diagnosed condition or disease. Dosage amount and interval can be adjusted individually to provide levels of a Compound of the Disclosure that is sufficient to maintain therapeutic effects.


Toxicity and therapeutic efficacy of the Compounds of the Disclosure can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the maximum tolerated dose (MTD) of a compound, which defines as the highest dose that causes no toxicity in animals. The dose ratio between the maximum tolerated dose and therapeutic effects (e.g. inhibiting of tumor growth) is the therapeutic index. The dosage can vary within this range depending upon the dosage form employed, and the route of administration utilized. Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.


A therapeutically effective amount of a Compound of the Disclosure required for use in therapy varies with the nature of the condition being treated, the length of time that activity is desired, and the age and the condition of the patient, and ultimately is determined by the attendant physician. Dosage amounts and intervals can be adjusted individually to provide plasma levels of the AR protein degrader that are sufficient to maintain the desired therapeutic effects. The desired dose conveniently can be administered in a single dose, or as multiple doses administered at appropriate intervals, for example as one, two, three, four or more subdoses per day. Multiple doses often are desired, or required. For example, a Compound of the Disclosure can be administered at a frequency of: four doses delivered as one dose per day at four-day intervals (q4d×4); four doses delivered as one dose per day at three-day intervals (q3d×4); one dose delivered per day at five-day intervals (qd×5); one dose per week for three weeks (qwk3); five daily doses, with two days rest, and another five daily doses (5/2/5); or, any dose regimen determined to be appropriate for the circumstance.


A Compound of the Disclosure used in a method of the present disclosure can be administered in an amount of about 0.005 to about 500 milligrams per dose, about 0.05 to about 250 milligrams per dose, or about 0.5 to about 100 milligrams per dose. For example, a Compound of the Disclosure can be administered, per dose, in an amount of about 0.005, 0.05, 0.5, 5, 10, 20, 30, 40, 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 milligrams, including all doses between 0.005 and 500 milligrams.


The dosage of a composition containing a Compound of the Disclosure, or a composition containing the same, can be from about 1 ng/kg to about 200 mg/kg, about 1 μg/kg to about 100 mg/kg, or about 1 mg/kg to about 50 mg/kg. The dosage of a composition can be at any dosage including, but not limited to, about 1 μg/kg. The dosage of a composition may be at any dosage including, but not limited to, about 1 μg/kg, about 10 μg/kg, about 25 μg/kg, about 50 μg/kg, about 75 μg/kg, about 100 μg/kg, about 125 μg/kg, about 150 μg/kg, about 175 μg/kg, about 200 μg/kg, about 225 μg/kg, about 250 μg/kg, about 275 μg/kg, about 300 μg/kg, about 325 μg/kg, about 350 μg/kg, about 375 μg/kg, about 400 μg/kg, about 425 μg/kg, about 450 μg/kg, about 475 μg/kg, about 500 μg/kg, about 525 μg/kg, about 550 μg/kg, about 575 μg/kg, about 600 μg/kg, about 625 μg/kg, about 650 μg/kg, about 675 μg/kg, about 700 μg/kg, about 725 μg/kg, about 750 μg/kg, about 775 μg/kg, about 800 μg/kg, about 825 μg/kg, about 850 μg/kg, about 875 μg/kg, about 900 μg/kg, about 925 μg/kg, about 950 μg/kg, about 975 μg/kg, about 1 mg/kg, about 5 mg/kg, about 10 mg/kg, about 15 mg/kg, about 20 mg/kg, about 25 mg/kg, about 30 mg/kg, about 35 mg/kg, about 40 mg/kg, about 45 mg/kg, about 50 mg/kg, about 60 mg/kg, about 70 mg/kg, about 80 mg/kg, about 90 mg/kg, about 100 mg/kg, about 125 mg/kg, about 150 mg/kg, about 175 mg/kg, about 200 mg/kg, or more. The above dosages are exemplary of the average case, but there can be individual instances in which higher or lower dosages are merited, and such are within the scope of this disclosure. In practice, the physician determines the actual dosing regimen that is most suitable for an individual patient, which can vary with the age, weight, and response of the particular patient.


As stated above, a Compound of the Disclosure can be administered in combination with a second therapeutically active agent. In some embodiments, the second therapeutic agent is an epigenetic drug. As used herein, the term “epigenetic drug” refers to a therapeutic agent that targets an epigenetic regulator. Examples of epigenetic regulators include the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases. Histone deacetylase inhibitors include, but are not limited to, vorinostat.


In another embodiment, chemotherapeutic agents or other anti-proliferative agents can be combined with Compound of the Disclosure to treat proliferative diseases and cancer. Examples of therapies and anticancer agents that can be used in combination with Compounds of the Disclosure include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, a biologic response modifier (e.g., an interferon, an interleukin, tumor necrosis factor (TNF), hyperthermia and cryotherapy, an agent to attenuate any adverse effect (e.g., an antiemetic), and any other approved chemotherapeutic drug.


Examples of antiproliferative compounds include, but are not limited to, an aromatase inhibitor; an anti-estrogen; an anti-androgen; a gonadorelin agonist; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active agent; an alkylating agent; a retinoid, a carontenoid, or a tocopherol; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an antimetabolite; a platin compound; a methionine aminopeptidase inhibitor; a bisphosphonate; an antiproliferative antibody; a heparanase inhibitor; an inhibitor of Ras oncogenic isoforms; a telomerase inhibitor; a proteasome inhibitor; a compound used in the treatment of hematologic malignancies; a Flt-3 inhibitor; an Hsp90 inhibitor; a kinesin spindle protein inhibitor; a MEK inhibitor; an antitumor antibiotic; a nitrosourea; a compound targeting/decreasing protein or lipid kinase activity, a compound targeting/decreasing protein or lipid phosphatase activity, or any further anti-angiogenic compound.


Nonlimiting exemplary aromatase inhibitors include, but are not limited to, steroids, such as atamestane, exemestane, and formestane, and non-steroids, such as aminoglutethimide, rogletimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole, and letrozole.


Nonlimiting anti-estrogens include, but are not limited to, tamoxifen, fulvestrant, raloxifene, and raloxifene hydrochloride. Anti-androgens include, but are not limited to, bicalutamide. Gonadorelin agonists include, but are not limited to, abarelix, goserelin, and goserelin acetate.


Exemplary topoisomerase I inhibitors include, but are not limited to, topotecan, gimatecan, irinotecan, camptothecin and its analogues, 9-nitrocamptothecin, and the macromolecular camptothecin conjugate PNU-166148. Topoisomerase II inhibitors include, but are not limited to, anthracyclines, such as doxorubicin, daunorubicin, epirubicin, idarubicin, and nemorubicin; anthraquinones, such as mitoxantrone and losoxantrone; and podophillotoxines, such as etoposide and teniposide.


Microtubule active agents include microtubule stabilizing, microtubule destabilizing compounds, and microtubulin polymerization inhibitors including, but not limited to, taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine, vinblastine sulfate, vincristine, and vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof.


Exemplary nonlimiting alkylating agents include cyclophosphamide, ifosfamide, melphalan, and nitrosoureas, such as carmustine and lomustine.


Exemplary nonlimiting cyclooxygenase inhibitors include Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib, rofecoxib, etoricoxib, valdecoxib, or a 5-alkyl-2-arylaminophenylacetic acid, such as lumiracoxib.


Exemplary nonlimiting matrix metalloproteinase inhibitors (“MMP inhibitors”) include collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, batimastat, marimastat, prinomastat, metastat, BMS-279251, BAY 12-9566, TAA211, MMI270B, and AAJ996.


Exemplary nonlimiting mTOR inhibitors include compounds that inhibit the mammalian target of rapamycin (mTOR) and possess antiproliferative activity such as sirolimus, everolimus, CCI-779, and ABT578.


Exemplary nonlimiting antimetabolites include 5-fluorouracil (5-FU), capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists, such as pemetrexed.


Exemplary nonlimiting platin compounds include carboplatin, cis-platin, cisplatinum, and oxaliplatin.


Exemplary nonlimiting methionine aminopeptidase inhibitors include bengamide or a derivative thereof and PPI-2458.


Exemplary nonlimiting bisphosphonates include etridonic acid, clodronic acid, tiludronic acid, pamidronic acid, alendronic acid, ibandronic acid, risedronic acid, and zoledronic acid.


Exemplary nonlimiting antiproliferative antibodies include trastuzumab, trastuzumab-DM1, cetuximab, bevacizumab, rituximab, PR064553, and 2C4. The term “antibody” is meant to include intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity.


Exemplary nonlimiting heparanase inhibitors include compounds that target, decrease, or inhibit heparin sulfate degradation, such as PI-88 and OGT2115.


The term “an inhibitor of Ras oncogenic isoforms,” such as H-Ras, K-Ras, or N-Ras, as used herein refers to a compound which targets, decreases, or inhibits the oncogenic activity of Ras, for example, a farnesyl transferase inhibitor, such as L-744832, DK8G557, tipifarnib, and lonafarnib.


Exemplary nonlimiting telomerase inhibitors include compounds that target, decrease, or inhibit the activity of telomerase, such as compounds that inhibit the telomerase receptor, such as telomestatin.


Exemplary nonlimiting proteasome inhibitors include compounds that target, decrease, or inhibit the activity of the proteasome including, but not limited to, bortezomid.


The phrase “compounds used in the treatment of hematologic malignancies” as used herein includes FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, I-β-D-arabinofuransylcytosine (ara-c), and bisulfan; and ALK inhibitors, which are compounds which target, decrease, or inhibit anaplastic lymphoma kinase.


Exemplary nonlimiting Flt-3 inhibitors include PKC412, midostaurin, a staurosporine derivative, SU11248, and MLN518.


Exemplary nonlimiting HSP90 inhibitors include compounds targeting, decreasing, or inhibiting the intrinsic ATPase activity of HSP90; or degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins, or antibodies that inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.


The phrase “a compound targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or any further anti-angiogenic compound” as used herein includes a protein tyrosine kinase and/or serine and/or threonine kinase inhibitor or lipid kinase inhibitor, such as a) a compound targeting, decreasing, or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as a compound that targets, decreases, or inhibits the activity of PDGFR, such as an N-phenyl-2-pyrimidine-amine derivatives, such as imatinib, SUlOl, SU6668, and GFB-111; b) a compound targeting, decreasing, or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); c) a compound targeting, decreasing, or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as a compound that targets, decreases, or inhibits the activity of IGF-IR; d) a compound targeting, decreasing, or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) a compound targeting, decreasing, or inhibiting the activity of the Ax1 receptor tyrosine kinase family; f) a compound targeting, decreasing, or inhibiting the activity of the Ret receptor tyrosine kinase; g) a compound targeting, decreasing, or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) a compound targeting, decreasing, or inhibiting the activity of the c-Kit receptor tyrosine kinases, such as imatinib; i) a compound targeting, decreasing, or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. Bcr-Abl kinase) and mutants, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib; PD180970; AG957; NSC 680410; PD173955; or dasatinib; j) a compound targeting, decreasing, or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK1, PKB/Akt, and Ras/MAPK family members, and/or members of the cyclin-dependent kinase family (CDK), such as a staurosporine derivative disclosed in U.S. Pat. No. 5,093,330, such as midostaurin; examples of further compounds include UCN-01, safingol, BAY 43-9006, bryostatin 1, perifosine; ilmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; LY333531/LY379196; a isochinoline compound; a farnesyl transferase inhibitor; PD184352 or QAN697, or AT7519; k) a compound targeting, decreasing or inhibiting the activity of a protein-tyrosine kinase, such as imatinib mesylate or a tyrphostin, such as Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin); 1) a compound targeting, decreasing, or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as CP 358774, ZD 1839, ZM 105180; trastuzumab, cetuximab, gefitinib, erlotinib, OSI-774, Cl-1033, EKB-569, GW-2016, antibodies E.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 and E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives; and m) a compound targeting, decreasing, or inhibiting the activity of the c-Met receptor.


Exemplary compounds that target, decrease, or inhibit the activity of a protein or lipid phosphatase include inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.


Further anti-angiogenic compounds include compounds having another mechanism for their activity unrelated to protein or lipid kinase inhibition, e.g., thalidomide and TNP-470.


Additional, nonlimiting, exemplary chemotherapeutic compounds, one or more of which may be used in combination with a Compound of the Disclosure, include: daunorubicin, adriamycin, Ara-C, VP-16, teniposide, mitoxantrone, idarubicin, carboplatinum, PKC412, 6-mercaptopurine (6-MP), fludarabine phosphate, octreotide, SOM230, FTY720, 6-thioguanine, cladribine, 6-mercaptopurine, pentostatin, hydroxyurea, 2-hydroxy-1H-isoindole-1,3-dione derivatives, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate, angiostatin, endostatin, anthranilic acid amides, ZD4190, ZD6474, SU5416, SU6668, bevacizumab, rhuMAb, rhuFab, macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, RPI 4610, bevacizumab, porfimer sodium, anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortex olone, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone, dexamethasone, fluocinolone, a plant alkaloid, a hormonal compound and/or antagonist, a biological response modifier, such as a lymphokine or interferon, an antisense oligonucleotide or oligonucleotide derivative, shRNA, and siRNA.


Other examples of second therapeutic agents, one or more of which a Compound of the Disclosure also can be combined, include, but are not limited to: a treatment for Alzheimer's Disease, such as donepezil and rivastigmine; a treatment for Parkinson's Disease, such as L-DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; an agent for treating multiple sclerosis (MS) such as beta interferon (e.g., AVONEX® and REBIF®), glatiramer acetate, and mitoxantrone; a treatment for asthma, such as albuterol and montelukast; an agent for treating schizophrenia, such as zyprexa, risperdal, seroquel, and haloperidol; an anti-inflammatory agent, such as a corticosteroid, a TNF blocker, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; an immunomodulatory agent, including immunosuppressive agents, such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, an interferon, a corticosteroid, cyclophosphamide, azathioprine, and sulfasalazine; a neurotrophic factor, such as an acetylcholinesterase inhibitor, an MAO inhibitor, an interferon, an anti-convulsant, an ion channel blocker, riluzole, or an anti-Parkinson's agent; an agent for treating cardiovascular disease, such as a beta-blocker, an ACE inhibitor, a diuretic, a nitrate, a calcium channel blocker, or a statin; an agent for treating liver disease, such as a corticosteroid, cholestyramine, an interferon, and an anti-viral agent; an agent for treating blood disorders, such as a corticosteroid, an anti-leukemic agent, or a growth factor; or an agent for treating immunodeficiency disorders, such as gamma globulin.


In another embodiment, the second therapeutically active agent is an immune checkpoint inhibitor. Examples of immune checkpoint inhibitors include PD-1 inhibitors, PD-L1 inhibitors, CTLA-4 inhibitors, LAG3 inhibitors, TIM3 inhibitors, cd47 inhibitors, and B7-H1 inhibitors. Thus, in one embodiment, a Compound of the Disclosure is administered in combination with an immune checkpoint inhibitor is selected from the group consisting of a PD-1 inhibitor, a PD-L1 inhibitor, a CTLA-4 inhibitor, a LAG3 inhibitor, a TIM3 inhibitor, and a cd47 inhibitor.


In another embodiment, the immune checkpoint inhibitor is a programmed cell death (PD-1) inhibitor. PD-1 is a T-cell coinhibitory receptor that plays a pivotal role in the ability of tumor cells to evade the host's immune system. Blockage of interactions between PD-1 and PD-L1, a ligand of PD-1, enhances immune function and mediates antitumor activity. Examples of PD-1 inhibitors include antibodies that specifically bind to PD-1. Particular anti-PD-1 antibodies include, but are not limited to nivolumab, pembrolizumab, STI-A1014, and pidilzumab. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies of anti-PD-1 antibodies, see U.S. 2013/0309250, U.S. Pat. Nos. 6,808,710, 7,595,048, 8,008,449, 8,728,474, 8,779,105, 8,952,136, 8,900,587, 9,073,994, 9,084,776, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a PD-L1 (also known as B7-H1 or CD274) inhibitor. Examples of PD-L1 inhibitors include antibodies that specifically bind to PD-L1. Particular anti-PD-L1 antibodies include, but are not limited to, avelumab, atezolizumab, durvalumab, and BMS-936559. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies, see U.S. Pat. No. 8,217,149, U.S. 2014/0341917, U.S. 2013/0071403, WO 2015036499, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a CTLA-4 inhibitor. CTLA-4, also known as cytotoxic T-lymphocyte antigen 4, is a protein receptor that downregulates the immune system. CTLA-4 is characterized as a “brake” that binds costimulatory molecules on antigen-presenting cells, which prevents interaction with CD28 on T cells and also generates an overtly inhibitory signal that constrains T cell activation. Examples of CTLA-4 inhibitors include antibodies that specifically bind to CTLA-4. Particular anti-CTLA-4 antibodies include, but are not limited to, ipilimumab and tremelimumab. For a general discussion of the availability, methods of production, mechanism of action, and clinical studies, see U.S. Pat. Nos. 6,984,720, 6,207,156, and Naido et al., British Journal of Cancer 111:2214-19 (2014).


In another embodiment, the immune checkpoint inhibitor is a LAG3 inhibitor. LAG3, Lymphocyte Activation Gene 3, is a negative co-stimulatory receptor that modulates T cell homeostatis, proliferation, and activation. In addition, LAG3 has been reported to participate in regulatory T cells (Tregs) suppressive function. A large proportion of LAG3 molecules are retained in the cell close to the microtubule-organizing center, and only induced following antigen specific T cell activation. U.S. 2014/0286935. Examples of LAG3 inhibitors include antibodies that specifically bind to LAG3. Particular anti-LAG3 antibodies include, but are not limited to, GSK2831781. For a general discussion of the availability, methods of production, mechanism of action, and studies, see, U.S. 2011/0150892, U.S. 2014/0093511, U.S. 20150259420, and Huang et al., Immunity 21:503-13 (2004).


In another embodiment, the immune checkpoint inhibitor is a TIM3 inhibitor. TIM3, T-cell immunoglobulin and mucin domain 3, is an immune checkpoint receptor that functions to limit the duration and magnitude of TH1 and TC1 T-cell responses. The TIM3 pathway is considered a target for anticancer immunotherapy due to its expression on dysfunctional CD8+ T cells and Tregs, which are two reported immune cell populations that constitute immunosuppression in tumor tissue. Anderson, Cancer Immunology Research 2:393-98 (2014). Examples of TIM3 inhibitors include antibodies that specifically bind to TIM3. For a general discussion of the availability, methods of production, mechanism of action, and studies of TIM3 inhibitors, see U.S. 20150225457, U.S. 20130022623, U.S. Pat. No. 8,522,156, Ngiow et al., Cancer Res 71: 6567-71 (2011), Ngiow, et al., Cancer Res 71:3540-51 (2011), and Anderson, Cancer Immunology Res 2:393-98 (2014).


In another embodiment, the immune checkpoint inhibitor is a cd47 inhibitor. See Unanue, E. R., PNAS 110:10886-87 (2013).


The term “antibody” is meant to include intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity. In another embodiment, “antibody” is meant to include soluble receptors that do not possess the Fc portion of the antibody. In one embodiment, the antibodies are humanized monoclonal antibodies and fragments thereof made by means of recombinant genetic engineering.


Another class of immune checkpoint inhibitors include polypeptides that bind to and block PD-1 receptors on T-cells without triggering inhibitor signal transduction. Such peptides include B7-DC polypeptides, B7-H1 polypeptides, B7-1 polypeptides and B7-2 polypeptides, and soluble fragments thereof, as disclosed in U.S. Pat. No. 8,114,845.


Another class of immune checkpoint inhibitors include compounds with peptide moieties that inhibit PD-1 signaling. Examples of such compounds are disclosed in U.S. Pat. No. 8,907,053.


Another class of immune checkpoint inhibitors include inhibitors of certain metabolic enzymes, such as indoleamine 2,3 dioxygenase (IDO), which is expressed by infiltrating myeloid cells and tumor cells. The IDO enzyme inhibits immune responses by depleting amino acids that are necessary for anabolic functions in T cells or through the synthesis of particular natural ligands for cytosolic receptors that are able to alter lymphocyte functions. Pardoll, Nature Reviews. Cancer 12:252-64 (2012); Löb, Cancer Immunol Immunother 58:153-57 (2009). Particular IDO blocking agents include, but are not limited to levo-1-methyl typtophan (L-1MT) and 1-methyl-tryptophan (1MT). Qian et al., Cancer Res 69:5498-504 (2009); and Löb et al., Cancer Immunol Immunother 58:153-7 (2009).


In one embodiment, the immune checkpoint inhibitor is nivolumab, pembrolizumab, pidilizumab, STI-A1110, avelumab, atezolizumab, durvalumab, STI-A1014, ipilimumab, tremelimumab, GSK2831781, BMS-936559 or MED14736


The above-mentioned second therapeutically active agents, one or more of which can be used in combination with a Compound of the Disclosure, are prepared and administered as described in the art.


Compounds of the Disclosure typically are administered in admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice. Pharmaceutical compositions for use in accordance with the present disclosure are formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and/or auxiliaries that facilitate processing of Compound of the Disclosure.


These pharmaceutical compositions can be manufactured, for example, by conventional mixing, dissolving, granulating, dragee-making, emulsifying, encapsulating, entrapping, or lyophilizing processes. Proper formulation is dependent upon the route of administration chosen. When a therapeutically effective amount of the Compound of the Disclosure is administered orally, the composition typically is in the form of a tablet, capsule, powder, solution, or elixir. When administered in tablet form, the composition additionally can contain a solid carrier, such as a gelatin or an adjuvant. The tablet, capsule, and powder contain about 0.01% to about 95%, and preferably from about 1% to about 50%, of a Compound of the Disclosure. When administered in liquid form, a liquid carrier, such as water, petroleum, or oils of animal or plant origin, can be added. The liquid form of the composition can further contain physiological saline solution, dextrose or other saccharide solutions, or glycols. When administered in liquid form, the composition contains about 0.1% to about 90%, and preferably about 1% to about 50%, by weight, of a Compound of the Disclosure.


When a therapeutically effective amount of a Compound of the Disclosure is administered by intravenous, cutaneous, or subcutaneous injection, the composition is in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred composition for intravenous, cutaneous, or subcutaneous injection typically contains, an isotonic vehicle.


Compounds of the Disclosure can be readily combined with pharmaceutically acceptable carriers well-known in the art. Standard pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 19th ed. 1995. Such carriers enable the active agents to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding the Compound of the Disclosure to a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.


Suitable excipients include fillers such as saccharides (for example, lactose, sucrose, mannitol or sorbitol), cellulose preparations, calcium phosphates (for example, tricalcium phosphate or calcium hydrogen phosphate), as well as binders such as starch paste (using, for example, maize starch, wheat starch, rice starch, or potato starch), gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone. If desired, one or more disintegrating agents can be added, such as the above-mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate. Buffers and pH modifiers can also be added to stabilize the pharmaceutical composition.


Auxiliaries are typically flow-regulating agents and lubricants such as, for example, silica, talc, stearic acid or salts thereof (e.g., magnesium stearate or calcium stearate), and polyethylene glycol. Dragee cores are provided with suitable coatings that are resistant to gastric juices. For this purpose, concentrated saccharide solutions can be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. In order to produce coatings resistant to gastric juices, solutions of suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropylmethyl-cellulose phthalate can be used. Dye stuffs or pigments can be added to the tablets or dragee coatings, for example, for identification or in order to characterize combinations of active compound doses.


Compound of the Disclosure can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, e.g., in ampules or in multidose containers, with an added preservative. The compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.


Pharmaceutical compositions for parenteral administration include aqueous solutions of the active agent in water-soluble form. Additionally, suspensions of a Compound of the Disclosure can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils or synthetic fatty acid esters. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension. Optionally, the suspension also can contain suitable stabilizers or agents that increase the solubility of the compounds and allow for the preparation of highly concentrated solutions. Alternatively, a present composition can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.


Compounds of the Disclosure also can be formulated in rectal compositions, such as suppositories or retention enemas, e.g., containing conventional suppository bases. In addition to the formulations described previously, the Compound of the Disclosure also can be formulated as a depot preparation. Such long-acting formulations can be administered by implantation (for example, subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the Compound of the Disclosure can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins.


In particular, the Compounds of the Disclosure can be administered orally, buccally, or sublingually in the form of tablets containing excipients, such as starch or lactose, or in capsules or ovules, either alone or in admixture with excipients, or in the form of elixirs or suspensions containing flavoring or coloring agents. Such liquid preparations can be prepared with pharmaceutically acceptable additives, such as suspending agents. Compound of the Disclosure also can be injected parenterally, for example, intravenously, intramuscularly, subcutaneously, or intracoronarily. For parenteral administration, the Compound of the Disclosure are typically used in the form of a sterile aqueous solution which can contain other substances, for example, salts or monosaccharides, such as mannitol or glucose, to make the solution isotonic with blood.


The disclosure provides the following particular embodiments in connection with treating a disease in a subject.


Embodiment I. A method of treating a subject, the method comprising administering to the subject a therapeutically effective amount of a Compound of the Disclosure, e.g., a compound of any one of CD Embodiments 1-117, wherein the subject has cancer, a chronic autoimmune disorder, an inflammatory condition, a proliferative disorder, sepsis, or a viral infection.


Embodiment II. The method Embodiment I, wherein the subject has cancer, e.g., any one of more of the cancers of Table 2 or Table 3.


Embodiment III. The method of Embodiment II, wherein the cancer is prostate cancer or breast cancer.


Embodiment IV. The method of Embodiment II, wherein the cancer is breast cancer.


Embodiment V. The method of Embodiment II, wherein the cancer is prostate cancer, e.g., metastatic castration-resistant prostate cancer.


Embodiment VI. The method of any one of Embodiments I-V further comprising administering a therapeutically effective amount of a second therapeutic agent useful in the treatment of the disease or condition, e.g., an immune checkpoint inhibitor or other anticancer agent.


Embodiment VII. A pharmaceutical composition comprising a Compound of the Disclosure, e.g., a compound of any one of CD Embodiments 1-117, and a pharmaceutically acceptable excipient for use in treating cancer, a chronic autoimmune disorder, an inflammatory condition, a proliferative disorder, sepsis, or a viral infection.


Embodiment VIII. The pharmaceutical composition of Embodiment VII for use in treating cancer.


Embodiment IX. The pharmaceutical composition of Embodiment VIII, wherein the cancer is prostate cancer or breast cancer.


Embodiment X. The pharmaceutical composition of Embodiment VIII, wherein the cancer is breast cancer.


Embodiment XI. The pharmaceutical composition of Embodiment VIII, wherein the cancer is prostate cancer, e.g., metastatic castration-resistant prostate cancer.


Embodiment XII. A Compound of the Disclosure, e.g., a compound of any one of CD Embodiments 1-117, for use in treatment of cancer, a chronic autoimmune disorder, an inflammatory condition, a proliferative disorder, sepsis, or a viral infection.


Embodiment XIII. The compound of Embodiment XIII for use in treating cancer.


Embodiment XIV. The compound of Embodiment XIII, wherein the cancer is breast cancer.


Embodiment XV. The compound of Embodiment XIII, wherein the cancer is prostate cancer, e.g., metastatic castration-resistant prostate cancer.


Embodiment XVI. Use of a Compound of the Disclosure, e.g., a compound of any one of CD Embodiments 1-117, for the manufacture of a medicament for treatment of cancer, a chronic autoimmune disorder, an inflammatory condition, a proliferative disorder, sepsis, or a viral infection.


Embodiment XVII. The use of Embodiment XVI for the treatment of cancer.


Embodiment XVIII. The use of Embodiment XVII, wherein the cancer is prostate cancer or breast cancer.


Embodiment XIV. The use of Embodiment XVII, wherein the cancer is breast cancer.


Embodiment XX. The use of Embodiment XVII, wherein the cancer is prostate cancer, e.g., metastatic castration-resistant prostate cancer.


Embodiment XXI. A method of reducing AR protein within a cell of a subject in need thereof, the method comprising administering to the patient a Compound of the Disclosure, e.g., a compound of any one of CD Embodiments 1-117. In one embodiment, the AR protein is reduced by about 50% or less, e.g., 1%, about 2%, about 3%, about 4%, about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, or about 45%. In one embodiment, the AR protein is reduced by about 51% or more, e.g., about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95%.


III. Intermediates of the Disclosure

In another aspect, the present disclosure provides Intermediates of the Disclosure. Intermediates of the Disclosure are compounds that can be used to prepare the heterobifunctional Compounds of the Disclosure.


In another aspect, the present disclosure provides the following particular embodiments drawn to Intermediates of the Disclosure referred to as “ID Embodiment 1,” “ID Embodiment 2,” “ID Embodiment 3,” and so on.


ID Embodiment 1. A compound of Formula II:




embedded image


wherein:


R3a is selected from the group consisting of halo, C1-C4 alkyl, and C1-C4 haloalkyl;


Z1 is selected from the group consisting of ═C(H)— and ═N—;


Z2 is selected from the group consisting of ═C(R3b)— and ═N—;


R3b is selected from the group consisting of hydrogen, halo, C1-C4 alkyl, and C1-C4 haloalkyl;


E is a spiroheterocyclenyl;


X1 is selected from the group consisting of —C(═O)—, —S(═O)2—, and —CR4aR4b—; or


X1 is absent;


R4a and R4b are independently selected from the group consisting of hydrogen and C1-C3 alkyl;


A1 is selected from the group consisting of cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl;


X2 is selected from the group consisting of —C(═O)—, —S(═O)2—, —O—, and —CR4cR4d—; or


X2 is absent;


R4c and R4d are independently selected from the group consisting of hydrogen and C1-3 alkyl;


L is -J1-J2-J3-J4-J5-,


wherein J1 is attached to X2;


J1 is selected from the group consisting of cycloalkylenyl and heterocyclenyl; or


J1 is absent;


J2 is selected from the group consisting of —(CH2)m1—, —CH═CH—, and —C≡C—;


m1 is 0, 1, 2, or 3;


J3 is selected from the group consisting of alkylenyl, heteroalkylenyl, cycloalkylenyl, heterocyclenyl, phenylenyl, and heteroarylenyl; or


J3 is absent;


J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl; or


J4 is absent;


J5 is selected from the group consisting of —(CH2)m2—, —O—, —N(R6)—, and —C(═O)—;


m2 is 0, 1, 2, or 3;


R6 is selected from the group consisting of hydrogen and C1-C3 alkyl;


B2 is selected from the group consisting of hydrogen, —CHO, and B2-1:




embedded image


m3 is 0, 1, or 2;


n3 is 0, 1, or 2;


each R1h is independently C1-C3 alkyl; and


k1 is 0, 1, or 2, or a salt or solvate thereof.


ID Embodiment 2. The compound of ID Embodiment 1, wherein E is selected from the group consisting of E-1, E-2, and E-3, see above;


wherein the bond designated with an “*” is attached to X1;


o and p are independently 0 or 1;


q and r are independently 0, 1, 2, or 3;


wherein the sum of o, p, q, and r is 2, 3, 4, 5, 6, or 7;


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


t, u, v, w, and x are independently 0, 1, 2, or 3;


R1a and R1b are independently selected from the group consisting of hydrogen, C1-C3 alkyl, C1-C4 haloalkyl, optionally substituted C3-C6 cycloalkyl, and (C3-C6 cycloalkyl)C1-C6 alkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted C3-C6 cycloalkyl; or


R1a and R1b taken together with the carbon atom to which they are attached form an optionally substituted 4- to 6-membered heterocyclo;


R1c and R1d are independently selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group;


each R1e is independently C1-C3 alkyl;


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


each R1f is independently C1-C3 alkyl;


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


each R1g is independently C1-C3 alkyl; and


h is 0, 1, 2, 3, or 4, or a salt or solvate thereof.


ID Embodiment 3. The compound of ID Embodiment 2, wherein E is E-1, or a salt or solvate thereof.


ID Embodiment 4. The compound of ID Embodiment 3, wherein E-1 is selected from the group consisting of E-1-1 and E-1-2, see above, or a salt or solvate thereof.


ID Embodiment 5. The compound of ID Embodiment 4, wherein E-1 is E-1-1, or a salt or solvate thereof.


ID Embodiment 6. The compound of ID Embodiment 5, wherein R1a and R1b are hydrogen, or a salt or solvate thereof.


ID Embodiment 7. The compound of ID Embodiment 6, wherein q, r, s, and t are 1, or a salt or solvate thereof.


ID Embodiment 8. The compound of ID Embodiment 6, wherein q is 2; r is 1; s is 0; and t is 1, or a salt or solvate thereof.


ID Embodiment 9. The compound of ID Embodiment 6, wherein q is 1; r is 0; s is 0; and t is 2, or a salt or solvate thereof.


ID Embodiment 10. The compound of ID Embodiment 6, wherein q is 0; r is 1; s is 1; and t is 1, or a salt or solvate thereof.


ID Embodiment 11. The compound of ID Embodiment 6, wherein q is 1; r is 1; s is 0; and t is 1, or a salt or solvate thereof


ID Embodiment 12. The compound of ID Embodiment 5, wherein R1a and R1b are independently C1-C3 alkyl, or a salt or solvate thereof.


ID Embodiment 13. The compound of ID Embodiment 12, wherein q, r, s, and t are 1, or a salt or solvate thereof.


ID Embodiment 14. The compound of ID Embodiment 5, wherein R1a is C1-C3 alkyl; and R1b is hydrogen or a salt or solvate thereof.


ID Embodiment 15. The compound of ID Embodiment 14, wherein q, r, s, and t are 1, or a salt or solvate thereof.


ID Embodiment 16. The compound of ID Embodiment 15 of Formula IX:




embedded image


or a salt or solvate thereof.


ID Embodiment 17. The compound of ID Embodiment 15 of Formula X:




embedded image


or a salt or solvate thereof.


ID Embodiment 18. The compound of ID Embodiment 5, wherein R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group, or a salt or solvate thereof.


ID Embodiment 19. The compound of ID Embodiment 18, wherein q, r, s, and t are 1, or a salt or solvate thereof.


ID Embodiment 20. The compound of ID Embodiment 4, wherein:


E-1 is E-1-2;


R1c is C1-C3 alkyl;


R1d is selected from the group consisting of hydrogen and C1-C3 alkyl; or


R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group, or a salt or solvate thereof.


ID Embodiment 21. The compound of ID Embodiment 20, wherein R1d is hydrogen, or a salt or solvate thereof.


ID Embodiment 22. The compound of ID Embodiment 21 of Formula XI:




embedded image


or a salt or solvate thereof.


ID Embodiment 23. The compound of ID Embodiment 21 of Formula XII:




embedded image


or a salt or solvate thereof.


ID Embodiment 24. The compound of ID Embodiment 20, wherein R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group, or a salt or solvate thereof.


ID Embodiment 25. The compound of ID Embodiment 2, wherein E is E-2, or a salt or solvate thereof.


ID Embodiment 26. The compound of ID Embodiment 25, wherein E-2 is E-2-1, see above, or a salt or solvate thereof.


ID Embodiment 27. The compound of ID Embodiment 2, wherein E is E-3, or a salt or solvate thereof.


ID Embodiment 28. The compound of ID Embodiment 27, wherein E-3 is E-3-1, see above, or a salt or solvate thereof.


ID Embodiment 29. The compound of any one of ID Embodiments 1-26, wherein X1 is —C(═O)—, or a salt or solvate thereof.


ID Embodiment 30. The compound of any one of ID Embodiments 1-26, wherein X1 is —S(═O)2—, or a salt or solvate thereof.


ID Embodiment 31. The compound of any one of ID Embodiments 1-26, wherein X1 is —CR4aR4b—, or a salt or solvate thereof.


ID Embodiment 32. The compound of ID Embodiments 31, wherein R4a and R4b are hydrogen, or a salt or solvate thereof.


ID Embodiment 33. The compound of any one of ID Embodiments 1-28, wherein X1 is absent, or a salt or solvate thereof.


ID Embodiment 34. The compound of any one of ID Embodiments 1-33, wherein:


A1 is selected from the group consisting of A1-1, A1-2, A1-3, A1-4, A1-5, A1-6, A1-7, and A1-8, see above, wherein the bond designated with an “*” is attached to X2;


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen, halo, C1-C3 alkyl, and C1-C3 alkoxy


e is 0, 1, or 2; and


f is 0, 1, or 2, or a salt or solvate thereof.


ID Embodiment 35. The compound of ID Embodiment 34, wherein A1 is A1-1, or a salt or solvate thereof.


ID Embodiment 36. The compound of ID Embodiment 34, wherein A1 is A1-2, or a salt or solvate thereof.


ID Embodiment 37. The compound of ID Embodiment 34, wherein A1 is A1-3, or a salt or solvate thereof.


ID Embodiment 38. The compound of ID Embodiment 34, wherein A1 is A1-4, or a salt or solvate thereof.


ID Embodiment 39. The compound of ID Embodiment 34, wherein A1 is A1-5, or a salt or solvate thereof.


ID Embodiment 40. The compound of ID Embodiment 34, wherein A1 is A1-6, or a salt or solvate thereof.


ID Embodiment 41. The compound of ID Embodiment 34, wherein A1 is A1-7, or a salt or solvate thereof.


ID Embodiment 42. The compound of any one of ID Embodiments 34-41, wherein R5a, R5b, R5c, and R5d are hydrogen, or a salt or solvate thereof.


ID Embodiment 43. The compound of any one of ID Embodiments 1-42, wherein X2 is —C(═O)—, or a salt or solvate thereof.


ID Embodiment 44. The compound of any one of ID Embodiments 1-42, wherein X2 is —S(═O)2—, or a salt or solvate thereof.


ID Embodiment 45. The compound of any one of ID Embodiments 1-42, wherein X2 is —O—, or a salt or solvate thereof.


ID Embodiment 46. The compound of any one of ID Embodiments 1-42, wherein X2 is —CR4cR4d—, or a salt or solvate thereof.


ID Embodiment 47. The compound of ID Embodiment 46, wherein R4c and R4d are hydrogen, or a salt or solvate thereof.


ID Embodiment 48. The compound of any one of ID Embodiments 1-42, wherein X2 is absent, or a salt or solvate thereof.


ID Embodiment 49. The compound of any one of ID Embodiments 1-48, wherein J1 is cycloalkylenyl, or a salt or solvate thereof.


ID Embodiment 50. The compound of any one of ID Embodiments 1-48, wherein J1 is heterocyclenyl, or a salt or solvate thereof.


ID Embodiment 51. The compound of ID Embodiment 20, wherein J1 is selected from the group consisting of J1-1, J1-2, J1-3, J1-4, J1-5, J1-6, J1-7, J1-8, J1-9, J1-10, J1-11, J1-12, and J1-13, see above, or a salt or solvate thereof.


ID Embodiment 52. The compound of ID Embodiment 51, wherein J1 is J1-1, or a salt or solvate thereof.


ID Embodiment 53. The compound of ID Embodiment 51, wherein J1 is J1-2, or a salt or solvate thereof.


ID Embodiment 54. The compound of ID Embodiment 51, wherein J1 is J1-3, or a salt or solvate thereof.


ID Embodiment 55. The compound of ID Embodiment 51, wherein J1 is J1-4, or a salt or solvate thereof.


ID Embodiment 56. The compound of ID Embodiment 51, wherein J1 is J1-5, or a salt or solvate thereof.


ID Embodiment 57. The compound of ID Embodiment 51, wherein J1 is J1-6, or a salt or solvate thereof.


ID Embodiment 58. The compound of ID Embodiment 51, wherein J1 is J1-7, or a salt or solvate thereof.


ID Embodiment 59. The compound of ID Embodiment 51, wherein J1 is J1-8, or a salt or solvate thereof.


ID Embodiment 60. The compound of ID Embodiment 51, wherein J1 is J1-9, or a salt or solvate thereof.


ID Embodiment 61. The compound of ID Embodiment 51, wherein J1 is J1-10, or a salt or solvate thereof.


ID Embodiment 62. The compound of ID Embodiment 51, wherein J1 is J1-11, or a salt or solvate thereof.


ID Embodiment 63. The compound of ID Embodiment 51, wherein J1 is J1-12, or a salt or solvate thereof.


ID Embodiment 64. The compound of ID Embodiment 51, wherein J1 is J1-13, or a salt or solvate thereof.


ID Embodiment 65. The compound of any one of ID Embodiments, 1-48, wherein J1 is absent, or a salt or solvate thereof.


ID Embodiment 66. The compound of any one of ID Embodiments 1-65, wherein J2 is selected from the group consisting of —(CH2)m1— and —C≡C—; and m1 is 0, 1, or 2, or a salt or solvate thereof.


ID Embodiment 67. The compound of ID Embodiment 66, wherein J2 is —(CH2)m1—; and m1 is 0, or a salt or solvate thereof.


ID Embodiment 68. The compound of ID Embodiment 66, wherein J2 is —(CH2)m1—; and m1 is 1, or a salt or solvate thereof.


ID Embodiment 69. The compound of ID Embodiment 66, wherein J2 is —C≡C—, or a salt or solvate thereof.


ID Embodiment 70. The compound of any one of ID Embodiments 1-69, wherein J3 is selected from the group consisting of cycloalkylenyl and heterocyclenyl, or a salt or solvate thereof.


ID Embodiment 71. The compound of ID Embodiment 70, wherein J3 is cycloalkylenyl, or a salt or solvate thereof.


ID Embodiment 72. The compound of ID Embodiment 70, wherein J3 is heterocyclenyl, or a salt or solvate thereof.


ID Embodiment 73. The compound of any one of ID Embodiments 1-69, wherein J3 is absent, or a salt or solvate thereof.


ID Embodiment 74. The compound of any one of ID Embodiments 1-73, wherein J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl, or a salt or solvate thereof.


ID Embodiment 75. The compound of ID Embodiment 74, wherein J4 is alkylenyl, or a salt or solvate thereof.


ID Embodiment 76. The compound of ID Embodiment 74, wherein J4 is cycloalkylenyl, or a salt or solvate thereof.


ID Embodiment 77. The compound of ID Embodiment 74, wherein J4 is heterocyclenyl, or a salt or solvate thereof.


ID Embodiment 78. The compound of any one of ID Embodiments 1-73, wherein J4 is absent, or a salt or solvate thereof.


ID Embodiment 79. The compound of any one of ID Embodiments 1-78, wherein:


J5 is selected from the group consisting of —O— and —N(H)—; and


B2 is hydrogen, or salt or solvate thereof.


ID Embodiment 80. The compound of any one of ID Embodiments 1-77, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —O—;


m2 is 0;


J4 is selected from the group consisting of J4-1, J4-2, J4-3, J4-4, J4-5, and J4-6, see above, wherein the bond designated with an “*” is attached to B2;


R7 is selected from the group consisting of hydrogen, halo, cyano, hydroxy, C1-C3 alkyl, and C1-C3 alkoxy; and


B2 is hydrogen, or a salt or solvate thereof.


ID Embodiment 81. The compound of ID Embodiment 80, wherein J4 is J4-1, or a salt or solvate thereof.


ID Embodiment 82. The compound of ID Embodiment 80, wherein J4 is J4-2, or a salt or solvate thereof.


ID Embodiment 83. The compound of ID Embodiment 80, wherein J4 is J4-3, or a salt or solvate thereof.


ID Embodiment 84. The compound of ID Embodiment 80, wherein J4 is J4-4, or a salt or solvate thereof.


ID Embodiment 85. The compound of ID Embodiment 80, wherein J4 is J4-5, or a salt or solvate thereof.


ID Embodiment 86. The compound of ID Embodiment 80, wherein J4 is J4-6, or a salt or solvate thereof.


ID Embodiment 87. The compound of any one of ID Embodiments 1-86, wherein:


J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;


m2 is 0, 1, 2, or 3; and


B2 is B1-1, or a salt or solvate thereof.


ID Embodiment 88. The compound of ID Embodiment 87, wherein k1 is 0, or a salt or solvate thereof.


ID Embodiment 89. The compound of ID Embodiments 87 or 88, wherein m3 and n3 are 1, or a salt or solvate thereof.


ID Embodiment 90. The compound of any one of ID Embodiments 1-43, wherein:


J1 is selected from the group consisting of:




embedded image


wherein the bond marked with “*” is attached to X2;


J3 and J4 are absent;


J2 is —(CH2)m1—;


m1 is 0;


J5 is —(CH2)m2—;


m2 is 0; and


B2 is —CHO, or a salt or solvate thereof.


ID Embodiment 91. The compound of any one of ID Embodiments 1-43, wherein:


J1, J3, and J4 are absent;


J2 is —(CH2)m1—;


m1 is 0;


J5 is —(CH2)m2—;


m2 is 0; and


B2 is B2-1, or a salt or solvate thereof.


ID Embodiment 92. The compound of any one of ID Embodiments 1-91, wherein R3a is halo, or a salt or solvate thereof.


ID Embodiment 93. The compound of any one of ID Embodiments 1-91, wherein R3a is C1-C4 alkyl, or a salt or solvate thereof.


ID Embodiment 94. The compound of any one of ID Embodiments 1-91, wherein R3a is C1-C4 haloalkyl, or a salt or solvate thereof.


ID Embodiment 95. The compound of any one of ID Embodiments 1-91, wherein R3a is selected from the group consisting of —Cl, —CH3, and —CF3, or a salt or solvate thereof.


ID Embodiment 96. The compound of any one of ID Embodiments 1-95, wherein Z1 is —C(H)═, or a salt or solvate thereof.


ID Embodiment 97. The compound of any one of ID Embodiment 1-96, wherein Z2 is —C(H)═, or a salt or solvate thereof.


ID Embodiment 98. The compound of ID Embodiment 1 of Formula XIII:




embedded image


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl; and


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen and halo.


ID Embodiment 99. The compound of ID Embodiment 1 of Formula XIV:




embedded image


R1a is selected from the group consisting of hydrogen and C1-C3 alkyl; and


R5a, R5b, R5c and R5d are each independently selected from the group consisting of hydrogen and halo.


ID Embodiment 100. The compound of ID Embodiments 98 or 99, wherein R1a is methyl and R5a, R5b, R5c, and R5d are hydrogen.


IV. Kits of the Disclosure

In another embodiment, the present disclosure provides kits which comprise a Compound of the Disclosure (or a composition comprising a Compound of the Disclosure) packaged in a manner that facilitates its use to practice methods of the present disclosure. In one embodiment, the kit includes a Compound of the Disclosure (or a composition comprising a Compound of the Disclosure) packaged in a container, such as a sealed bottle or vessel, with a label affixed to the container or included in the kit that describes use of the compound or composition to practice the method of the disclosure. In one embodiment, the compound or composition is packaged in a unit dosage form. The kit further can include a device suitable for administering the composition according to the intended route of administration.


V. Definitions

The term “a disease or condition wherein degradation of androgen receptor (AR) provides a benefit” and the like pertains to a disease or condition in which the androgen receptor is important or necessary, e.g., for the onset, progress, expression of that disease or condition, or a disease or a condition which is known to be treated by an AR degrader. Examples of such conditions include, but are not limited to, a cancer. One of ordinary skill in the art is readily able to determine whether a compound treats a disease or condition mediated by an AR degrader for any particular cell type, for example, by assays which conveniently can be used to assess the activity of particular compounds.


The term “androgen receptor degrader,” “AR degrader,” and the like refer to a heterobifunctional small molecule that degrades AR protein. AR degraders contain a first ligand which binds to AR protein, a second ligand for an E3 ligase system, and a chemical linker that tethers the first and second ligands. Representative Compounds of the Disclosure that degrade AR protein are disclosed in Table 1.


The term “second therapeutic agent” refers to a therapeutic agent different from a Compound of the Disclosure and that is known to treat the disease or condition of interest. For example when a cancer is the disease or condition of interest, the second therapeutic agent can be a known chemotherapeutic drug, like taxol, or radiation, for example.


The term “disease” or “condition” denotes disturbances and/or anomalies that as a rule are regarded as being pathological conditions or functions, and that can manifest themselves in the form of particular signs, symptoms, and/or malfunctions. Compounds of the Disclosure are degraders of AR and can be used in treating or preventing diseases and conditions wherein degradation of AR provides a benefit.


As used herein, the terms “treat,” “treating,” “treatment,” and the like refer to eliminating, reducing, or ameliorating a disease or condition, and/or symptoms associated therewith. Although not precluded, treating a disease or condition does not require that the disease, condition, or symptoms associated therewith be completely eliminated. The term “treat” and synonyms contemplate administering a therapeutically effective amount of a Compound of the Disclosure to a subject in need of such treatment. The treatment can be orientated symptomatically, for example, to suppress symptoms. It can be effected over a short period, be oriented over a medium term, or can be a long-term treatment, for example within the context of a maintenance therapy.


As used herein, the terms “prevent,” “preventing,” and “prevention” refer to a method of preventing the onset of a disease or condition and/or its attendant symptoms or barring a subject from acquiring a disease. As used herein, “prevent,” “preventing,” and “prevention” also include delaying the onset of a disease and/or its attendant symptoms and reducing a subject's risk of acquiring a disease. The terms “prevent,” “preventing” and “prevention” may include “prophylactic treatment,” which refers to reducing the probability of redeveloping a disease or condition, or of a recurrence of a previously-controlled disease or condition, in a subject who does not have, but is at risk of or is susceptible to, redeveloping a disease or condition or a recurrence of the disease or condition.


The term “therapeutically effective amount” or “effective dose” as used herein refers to an amount of the active ingredient(s) that is(are) sufficient, when administered by a method of the disclosure, to efficaciously deliver the active ingredient(s) for the treatment of condition or disease of interest to a subject in need thereof. In the case of a cancer or other proliferation disorder, the therapeutically effective amount of the agent may reduce (i.e., retard to some extent or stop) unwanted cellular proliferation; reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., retard to some extent or stop) cancer cell infiltration into peripheral organs; inhibit (i.e., retard to some extent or stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve, to some extent, one or more of the symptoms associated with the cancer. To the extent the administered compound or composition prevents growth and/or kills existing cancer cells, it may be cytostatic and/or cytotoxic.


The term “container” means any receptacle and closure therefore suitable for storing, shipping, dispensing, and/or handling a pharmaceutical product.


The term “insert” means information accompanying a pharmaceutical product that provides a description of how to administer the product, along with the safety and efficacy data required to allow the physician, pharmacist, and patient to make an informed decision regarding use of the product. The package insert generally is regarded as the “label” for a pharmaceutical product.


“Concurrent administration,” “administered in combination,” “simultaneous administration,” and similar phrases mean that two or more agents are administered concurrently to the subject being treated. By “concurrently,” it is meant that each agent is administered either simultaneously or sequentially in any order at different points in time. However, if not administered simultaneously, it is meant that they are administered to a subject in a sequence and sufficiently close in time so as to provide the desired therapeutic effect and can act in concert. For example, a Compound of the Disclosure can be administered at the same time or sequentially in any order at different points in time as a second therapeutic agent. A Compound of the Disclosure and the second therapeutic agent can be administered separately, in any appropriate form and by any suitable route. When a Compound of the Disclosure and the second therapeutic agent are not administered concurrently, it is understood that they can be administered in any order to a subject in need thereof. For example, a Compound of the Disclosure can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapeutic agent treatment modality (e.g., radiotherapy), to a subject in need thereof. In various embodiments, a Compound of the Disclosure and the second therapeutic agent are administered 1 minute apart, 10 minutes apart, 30 minutes apart, less than 1 hour apart, 1 hour apart, 1 hour to 2 hours apart, 2 hours to 3 hours apart, 3 hours to 4 hours apart, 4 hours to 5 hours apart, 5 hours to 6 hours apart, 6 hours to 7 hours apart, 7 hours to 8 hours apart, 8 hours to 9 hours apart, 9 hours to 10 hours apart, 10 hours to 11 hours apart, 11 hours to 12 hours apart, no more than 24 hours apart or no more than 48 hours apart. In one embodiment, the components of the combination therapies are administered at about 1 minute to about 24 hours apart.


The use of the terms “a”, “an”, “the”, and similar referents in the context of describing the disclosure (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated. Recitation of ranges of values herein merely are intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended to better illustrate the disclosure and is not a limitation on the scope of the disclosure unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the disclosure.


The term “halo” as used herein by itself or as part of another group refers to —Cl, —F, —Br, or —I.


The term “nitro” as used herein by itself or as part of another group refers to —NO2.


The term “cyano” as used herein by itself or as part of another group refers to —CN.


The term “hydroxy” as herein used by itself or as part of another group refers to —OH.


The term “alkyl” as used herein by itself or as part of another group refers to a straight- or branched-chain aliphatic hydrocarbon containing one to twelve carbon atoms, i.e., a C1-C12 alkyl, or the number of carbon atoms designated, e.g., a C1 alkyl such as methyl, a C2 alkyl such as ethyl, etc. In one embodiment, the alkyl is a C1-C10 alkyl. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1-C3 alkyl, i.e., methyl, ethyl, propyl, or isopropyl. Non-limiting exemplary C1-C12 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, iso-butyl, 3-pentyl, hexyl, heptyl, octyl, nonyl, and decyl.


The term “optionally substituted alkyl” as used herein by itself or as part of another group refers to an alkyl group that is either unsubstituted or substituted with one, two, or three substituents, wherein each substituent is independently nitro, haloalkoxy, aryloxy, aralkyloxy, alkylthio, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carbamate, carboxy, alkoxycarbonyl, carboxyalkyl, —N(R56a)C(═O)R56b, —N(R56c)S(═O)2R56d, —C(═O)R57, —S(═O)R56e, or —S(═O)2R58; wherein:


R56a is hydrogen or alkyl;


R56b is alkyl, haloalkyl, optionally substituted cycloalkyl, alkoxy, (alkoxy)alkyl, (aryl)alkyl, (heteroaryl)alkyl, (amino)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycle, optionally substituted C6-C10 aryl, or optionally substituted heteroaryl;


R56c is hydrogen or alkyl;


R56d is alkyl, haloalkyl, optionally substituted cycloalkyl, alkoxy, (alkoxy)alkyl, (aryl)alkyl, (heteroaryl)alkyl, (amino)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycle, optionally substituted C6-C10 aryl, or optionally substituted heteroaryl;


R56e is alkyl, haloalkyl, optionally substituted cycloalkyl, alkoxy, (alkoxy)alkyl, (aryl)alkyl, (heteroaryl)alkyl, (amino)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycle, optionally substituted C6-C10 aryl, or optionally substituted heteroaryl;


R57 is haloalkyl, optionally substituted cycloalkyl, alkoxy, (alkoxy)alkyl, (aryl)alkyl, (heteroaryl)alkyl, (amino)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycle, or optionally substituted heteroaryl; and


R58 is haloalkyl, optionally substituted cycloalkyl, alkoxy, (alkoxy)alkyl, (aryl)alkyl, (heteroaryl)alkyl, (amino)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted cycloalkyl, optionally substituted heterocycle, or optionally substituted heteroaryl. Non-limiting exemplary optionally substituted alkyl groups include —CH(CO2Me)CH2CO2Me and —CH(CH3)CH2N(H)C(═O)O(CH3)3.


The term “alkenyl” as used herein by itself or as part of another group refers to an alkyl group containing one, two, or three carbon-to-carbon double bonds. In one embodiment, the alkenyl group is a C2-C6 alkenyl group. In another embodiment, the alkenyl group is a C2-C4 alkenyl group. In another embodiment, the alkenyl group has one carbon-to-carbon double bond. Non-limiting exemplary alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, sec-butenyl, pentenyl, and hexenyl.


The term “optionally substituted alkenyl” as used herein by itself or as part of another refers to an alkenyl group that is either unsubstituted or substituted with one, two or three substituents, wherein each substituent is independently halo, nitro, cyano, hydroxy, amino (e.g., alkylamino, dialkylamino), haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, or optionally substituted heterocyclo. Non-limiting exemplary optionally substituted alkenyl groups include —CH═CHPh.


The term “alkynyl” as used herein by itself or as part of another group refers to an alkyl group containing one, two, or three carbon-to-carbon triple bonds. In one embodiment, the alkynyl is a C2-C6 alkynyl. In another embodiment, the alkynyl is a C2-C4 alkynyl. In another embodiment, the alkynyl has one carbon-to-carbon triple bond. Non-limiting exemplary alkynyl groups include ethynyl, propynyl, butynyl, 2-butynyl, pentynyl, and hexynyl groups.


The term “optionally substituted alkynyl” as used herein by itself or as part of another group refers to an alkynyl group that is either unsubstituted or substituted with one, two or three substituents, wherein each substituent is independently halo, nitro, cyano, hydroxy, amino, e.g., alkylamino, dialkylamino, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, or optionally substituted heterocyclo. Non-limiting exemplary optionally substituted alkynyl groups include —C≡CPh and —CH(Ph)C≡CH.


The term “haloalkyl” as used herein by itself or as part of another group refers to an alkyl group substituted by one or more fluorine, chlorine, bromine, and/or iodine atoms. In one embodiment, the alkyl is substituted by one, two, or three fluorine and/or chlorine atoms. In another embodiment, the alkyl is substituted by one, two, or three fluorine atoms. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl group is a C1 or C2 alkyl. Non-limiting exemplary haloalkyl groups include fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, 2,2-difluoroethyl, 2,2,2-trifluoroethyl, 3,3,3-trifluoropropyl, 4,4,4-trifluorobutyl, and trichloromethyl groups.


The terms “hydroxyalkyl” or “(hydroxy)alkyl” as used herein by themselves or as part of another group refer to an alkyl group substituted with one, two, or three hydroxy groups. In one embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1 or C2 alkyl. In another embodiment, the hydroxyalkyl is a monohydroxyalkyl group, i.e., substituted with one hydroxy group. In another embodiment, the hydroxyalkyl group is a dihydroxyalkyl group, i.e., substituted with two hydroxy groups. Non-limiting exemplary (hydroxyl)alkyl groups include hydroxymethyl, hydroxyethyl, hydroxypropyl and hydroxybutyl groups, such as 1-hydroxyethyl, 2-hydroxyethyl, 1,2-dihydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 3-hydroxybutyl, 4-hydroxybutyl, 2-hydroxy-1-methylpropyl, and 1,3-dihydroxyprop-2-yl.


The term “alkoxy” as used herein by itself or as part of another group refers to an alkyl group attached to a terminal oxygen atom. In one embodiment, the alkyl is a C1-C6 alkyl and resulting alkoxy is thus referred to as a “C1-C6 alkoxy.” In another embodiment, the alkyl is a C1-C4 alkyl group. Non-limiting exemplary alkoxy groups include methoxy, ethoxy, and tert-butoxy.


The term “haloalkoxy” as used herein by itself or as part of another group refers to a haloalkyl group attached to a terminal oxygen atom. In one embodiment, the haloalkyl group is a C1-C6 haloalkyl. In another embodiment, the haloalkyl group is a C1-C4 haloalkyl group. Non-limiting exemplary haloalkoxy groups include fluoromethoxy, difluoromethoxy, trifluoromethoxy, and 2,2,2-trifluoroethoxy.


The term “alkylthio” as used herein by itself or as part of another group refers to an alkyl group attached to a terminal sulfur atom. In one embodiment, the alkyl group is a C1-C4 alkyl group. Non-limiting exemplary alkylthio groups include —SCH3, and —SCH2CH3.


The terms “alkoxyalkyl” or “(alkoxy)alkyl” as used herein by themselves or as part of another group refers to an alkyl group substituted with one alkoxy group. In one embodiment, the alkoxy is a C1-C6 alkoxy. In another embodiment, the alkoxy is a C1-C4 alkoxy. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. Non-limiting exemplary alkoxyalkyl groups include methoxymethyl, methoxyethyl, methoxypropyl, methoxybutyl, ethoxymethyl, ethoxyethyl, ethoxypropyl, ethoxybutyl, propoxymethyl, iso-propoxymethyl, propoxyethyl, propoxypropyl, butoxymethyl, tert-butoxymethyl, isobutoxymethyl, sec-butoxymethyl, and pentyloxymethyl.


The term “heteroalkyl” as used by itself or part of another group refers to unsubstituted straight- or branched-chain aliphatic hydrocarbons containing from three to twenty chain atoms, i.e., 3- to 20-membered heteroalkyl, or the number of chain atoms designated, wherein at least one —CH2— is replaced with at least one of —O—, —N(H)—, —N(C1-C4 alkyl)-, or —S—. The —O—, —N(H)—, —N(C1-C4 alkyl)-, or —S— can independently be placed at any interior position of the aliphatic hydrocarbon chain so long as each —O—, —N(H)—, —N(C1-C4 alkyl)-, and —S— group is separated by at least two —CH2— groups. In one embodiment, one —CH2— group is replaced with one —O— group. In another embodiment, two —CH2— groups are replaced with two —O— groups. In another embodiment, three —CH2— groups are replaced with three —O— groups. In another embodiment, four —CH2— groups are replaced with four —O— groups. Non-limiting exemplary heteroalkyl groups include —CH2OCH3, —CH2OCH2CH2CH3, —CH2CH2CH2OCH3, —CH2CH2OCH2CH2OCH2CH3, —CH2CH2OCH2CH2OCH2CH2OCH2CH3.


The term “cycloalkyl” as used herein by itself or as part of another group refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic aliphatic hydrocarbons containing three to twelve carbon atoms, i.e., a C3-12 cycloalkyl, or the number of carbons designated, e.g., a C3 cycloalkyl such a cyclopropyl, a C4 cycloalkyl such as cyclobutyl, etc. In one embodiment, the cycloalkyl is bicyclic, i.e., it has two rings. In another embodiment, the cycloalkyl is monocyclic, i.e., it has one ring. In another embodiment, the cycloalkyl is a C3-8 cycloalkyl. In another embodiment, the cycloalkyl is a C3-6 cycloalkyl, i.e., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In another embodiment, the cycloalkyl is a C5 cycloalkyl, i.e., cyclopentyl. In another embodiment, the cycloalkyl is a C6 cycloalkyl, i.e., cyclohexyl. Non-limiting exemplary C3-12 cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, decalin, adamantyl, cyclohexenyl, and spiro[3.3]heptane.


The term “optionally substituted cycloalkyl” as used herein by itself or as part of another group refers to a cycloalkyl group that is either unsubstituted or substituted with one, two, or three substituents, wherein each substituent is independently halo, nitro, cyano, hydroxy, amino (e.g., —NH2, alkylamino, dialkylamino, aralkylamino, hydroxyalkylamino, or (heterocyclo)alkylamino), heteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyl, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxyalkyl, (amino)alkyl, (cyano)alkyl, (carboxamido)alkyl, mercaptoalkyl, (heterocyclo)alkyl, (heteroaryl)alkyl, —N(R56a)C(═O)R56b, —N(R56c)S(═O)2R56d, —C(═O)R57, —S(═O)R56e, —S(═O)2R58, or —OR59, wherein R56a, R56b, R56c, R56d, R56e, R57, and R58 are as defined in connection with the term “optionally substituted alkyl” and R59 is (hydroxy)alkyl or (amino)alkyl. The term optionally substituted cycloalkyl also includes cycloalkyl groups having fused optionally substituted aryl or optionally substituted heteroaryl groups such as




embedded image


Non-limiting exemplary optionally substituted cycloalkyl groups include:




embedded image


The term “heterocyclo” as used herein by itself or as part of another group refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic groups containing three to eighteen ring members, i.e., a 3- to 18-membered heterocyclo, comprising one, two, three, or four heteroatoms. Each heteroatom is independently oxygen, sulfur, or nitrogen. Each sulfur atom is independently oxidized to give a sulfoxide, i.e., S(═O), or sulfone, i.e., S(═O)2. The term heterocyclo includes groups wherein one or more —CH2— groups is replaced with one or more —C(═O)— groups, including cyclic ureido groups such as imidazolidinyl-2-one, cyclic amide groups such as pyrrolidin-2-one or piperidin-2-one, and cyclic carbamate groups such as oxazolidinyl-2-one. The term heterocyclo also includes groups having fused optionally substituted aryl or optionally substituted heteroaryl groups such as indoline, indolin-2-one, 2,3-dihydro-1H-pyrrolo[2,3-c]pyridine, 2,3,4,5-tetrahydro-1H-benzo[d]azepine, or 1,3,4,5-tetrahydro-2H-benzo[d]azepin-2-one.


In one embodiment, the heterocyclo group is a 4- to 8-membered cyclic group containing one ring and one or two oxygen atoms, e.g., tetrahydrofuran or tetrahydropyran, or one or two nitrogen atoms, e.g., pyrrolidine, piperidine, or piperazine, or one oxygen and one nitrogen atom, e.g., morpholine, and, optionally, one —CH2— group is replaced with one —C(═O)— group, e.g., pyrrolidin-2-one or piperazin-2-one. In another embodiment, the heterocyclo group is a 5- to 8-membered cyclic group containing one ring and one or two nitrogen atoms and, optionally, one —CH2— group is replaced with one —C(═O)— group. In another embodiment, the heterocyclo group is a 5- or 6-membered cyclic group containing one ring and one or two nitrogen atoms and, optionally, one —CH2— group is replaced with one —C(═O)— group. In another embodiment, the heterocyclo group is a 8- to 12-membered cyclic group containing two rings and one or two nitrogen atoms. The heterocyclo can be linked to the rest of the molecule through any available carbon or nitrogen atom. Non-limiting exemplary heterocyclo groups include:




embedded image


The term “optionally substituted heterocyclo” as used herein by itself or part of another group refers to a heterocyclo group that is either unsubstituted or substituted with one to four substituents, wherein each substituent is independently halo, nitro, cyano, hydroxy, amino, (e.g., —NH2, alkylamino, dialkylamino, aralkylamino, hydroxyalkylamino, or (heterocyclo)alkylamino), heteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyl, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxyalkyl, (amino)alkyl, (cyano)alkyl, (carboxamido)alkyl, mercaptoalkyl, (heterocyclo)alkyl, (heteroaryl)alkyl, —N(R56a)C(═O)R56b, —N(R56c)S(═O)2R56d, —C(═O)R57, —S(═O)R56e, —S(═O)2R58, or —OR59, wherein R56a, R56b, R56c, R56d, R56e, R57, R58, and R59 are as defined in connection with the term “optionally substituted cycloalkyl.” Substitution may occur on any available carbon or nitrogen atom of the heterocyclo group. Non-limiting exemplary optionally substituted heterocyclo groups include:




embedded image


In one embodiment, the heterocyclo group is a spiroheterocyclo. The term “spiroheterocyclo” as used herein by itself or part of another group refers to an optionally substituted heterocyclo group containing seven to eighteen ring members, wherein:


(i) a first and second ring are connected through a quaternary carbon atom, i.e., a spirocarbon;


(ii) the first ring is an optionally substituted mono- or bicyclic heterocyclo containing a nitrogen atom; and


(iii) the second ring is either:


(a) an optionally substituted mono- or bicyclic cycloalkyl; or


(b) an optionally substituted mono- or bicyclic heterocyclo containing a nitrogen atom.


In one embodiment, the first ring is an optionally substituted monocyclic 4- to 9-membered heterocyclo containing a nitrogen atom. In another embodiment, the second ring is an optionally substituted monocyclic C3-8 cycloalkyl. In another embodiment, the second ring is a monocyclic C3-8 cycloalkyl substituted with a hydroxy group. In another embodiment, the second ring is an optionally substituted monocyclic 4- to 9-membered heterocyclo containing a nitrogen atom. Non-limiting exemplary spiroheterocyclo groups include:




embedded image


The term “aryl” as used herein by itself or as part of another group refers to an aromatic ring system having six to fourteen carbon atoms, i.e., C6-C14 aryl. Non-limiting exemplary aryl groups include phenyl (abbreviated as “Ph”), naphthyl, phenanthryl, anthracyl, indenyl, azulenyl, biphenyl, biphenylenyl, and fluorenyl groups. In one embodiment, the aryl group is phenyl or naphthyl. In another embodiment, the aryl group is phenyl.


The term “optionally substituted aryl” as used herein by itself or as part of another group refers to aryl that is either unsubstituted or substituted with one to five substituents, wherein the substituents are each independently halo, nitro, cyano, hydroxy, amino, (e.g., —NH2, alkylamino, dialkylamino, aralkylamino, hydroxyalkylamino, or (heterocyclo)alkylamino), heteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyl, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxyalkyl, (amino)alkyl, (cyano)alkyl, (carboxamido)alkyl, mercaptoalkyl, (heterocyclo)alkyl, (heteroaryl)alkyl, —N(R56a)C(═O)R56b, —N(R56c)S(═O)2R56d, —C(═O)R57, —S(═O)R56e, —S(═O)2R58, or —OR59, wherein R56a, R56b, R56c, R56d, R56e, R57, R58, and R59 are as defined in connection with the term “optionally substituted cycloalkyl.”


In one embodiment, the optionally substituted aryl is an optionally substituted phenyl. In another embodiment, the optionally substituted phenyl has four substituents. In another embodiment, the optionally substituted phenyl has three substituents. In another embodiment, the optionally substituted phenyl has two substituents. In another embodiment, the optionally substituted phenyl has one substituent. Non-limiting exemplary optionally substituted aryl groups include 2-methylphenyl, 2-methoxyphenyl, 2-fluorophenyl, 2-chlorophenyl, 2-bromophenyl, 3-methylphenyl, 3-methoxyphenyl, 3-fluorophenyl, 3-chlorophenyl, 4-methylphenyl, 4-ethylphenyl, 4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 2,6-di-fluorophenyl, 2,6-di-chlorophenyl, 2-methyl, 3-methoxyphenyl, 2-ethyl, 3-methoxyphenyl, 3,4-di-methoxyphenyl, 3,5-di-fluorophenyl 3,5-di-methylphenyl, 3,5-dimethoxy, 4-methylphenyl, 2-fluoro-3-chlorophenyl, 3-chloro-4-fluorophenyl, and 2-phenylpropan-2-amine. The term optionally substituted aryl includes aryl groups having fused optionally substituted cycloalkyl groups and fused optionally substituted heterocyclo groups. Non-limiting examples include: 2,3-dihydro-1H-inden-1-yl, 1,2,3,4-tetrahydronaphthalen-1-yl, 1,3,4,5-tetrahydro-2H-benzo[c]azepin-2-yl, 1,2,3,4-tetrahydroisoquinolin-1-yl, and 2-oxo-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-yl.


The term “heteroaryl” as used herein by itself or as part of another group refers to monocyclic and bicyclic aromatic ring systems having five to 14 fourteen ring members, i.e., a 5- to 14-membered heteroaryl, comprising one, two, three, or four heteroatoms. Each heteroatom is independently oxygen, sulfur, or nitrogen. In one embodiment, the heteroaryl has three heteroatoms. In another embodiment, the heteroaryl has two heteroatoms. In another embodiment, the heteroaryl has one heteroatom. In another embodiment, the heteroaryl is a 5- to 10-membered heteroaryl. In another embodiment, the heteroaryl has 5 ring atoms, e.g., thienyl, a 5-membered heteroaryl having four carbon atoms and one sulfur atom. In another embodiment, the heteroaryl has 6 ring atoms, e.g., pyridyl, a 6-membered heteroaryl having five carbon atoms and one nitrogen atom. Non-limiting exemplary heteroaryl groups include thienyl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl, benzofuryl, pyranyl, isobenzofuranyl, benzooxazonyl, chromenyl, xanthenyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, cinnolinyl, quinazolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, β-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, thiazolyl, isothiazolyl, phenothiazolyl, isoxazolyl, furazanyl, and phenoxazinyl. In one embodiment, the heteroaryl is chosen from thienyl (e.g., thien-2-yl and thien-3-yl), furyl (e.g., 2-furyl and 3-furyl), pyrrolyl (e.g., 1H-pyrrol-2-yl and 1H-pyrrol-3-yl), imidazolyl (e.g., 2H-imidazol-2-yl and 2H-imidazol-4-yl), pyrazolyl (e.g., 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, and 1H-pyrazol-5-yl), pyridyl (e.g., pyridin-2-yl, pyridin-3-yl, and pyridin-4-yl), pyrimidinyl (e.g., pyrimidin-2-yl, pyrimidin-4-yl, and pyrimidin-5-yl), thiazolyl (e.g., thiazol-2-yl, thiazol-4-yl, and thiazol-5-yl), isothiazolyl (e.g., isothiazol-3-yl, isothiazol-4-yl, and isothiazol-5-yl), oxazolyl (e.g., oxazol-2-yl, oxazol-4-yl, and oxazol-5-yl) and isoxazolyl (e.g., isoxazol-3-yl, isoxazol-4-yl, and isoxazol-5-yl). The term heteroaryl also includes N-oxides. A non-limiting exemplary N-oxide is pyridyl N-oxide.


The term “optionally substituted heteroaryl” as used herein by itself or as part of another group refers to a heteroaryl that is either unsubstituted or substituted with one to four substituents, wherein the substituents are independently halo, nitro, cyano, hydroxy, amino, (e.g., —NH2, alkylamino, dialkylamino, aralkylamino, hydroxyalkylamino, or (heterocyclo)alkylamino), heteroalkyl, haloalkyl, hydroxyalkyl, alkoxy, haloalkoxy, aryloxy, aralkyl, aralkyloxy, alkylthio, carboxamido, sulfonamido, alkylcarbonyl, arylcarbonyl, alkylsulfonyl, arylsulfonyl, ureido, guanidino, carboxy, carboxyalkyl, optionally substituted alkyl, optionally substituted cycloalkyl, alkenyl, alkynyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocyclo, alkoxyalkyl, (amino)alkyl, (cyano)alkyl, (carboxamido)alkyl, mercaptoalkyl, (heterocyclo)alkyl, (heteroaryl)alkyl, —N(R56a)C(═O)R56b, —N(R56c)S(═O)2R56d, —C(═O)R57, —S(═O)R56e, —S(═O)2R58, or —OR59, wherein R56a, R56b, —R56c, R56d, R56, R57, R58, and R59 are as defined in connection with the term “optionally substituted cycloalkyl.”


In one embodiment, the optionally substituted heteroaryl has two substituents. In another embodiment, the optionally substituted heteroaryl has one substituent. Any available carbon or nitrogen atom can be substituted.


The term “aryloxy” as used herein by itself or as part of another group refers to an optionally substituted aryl attached to a terminal oxygen atom. A non-limiting exemplary aryloxy group is PhO—.


The term “heteroaryloxy” as used herein by itself or as part of another group refers to an optionally substituted heteroaryl attached to a terminal oxygen atom. A non-limiting exemplary aryloxy group is pyridyl-O—.


The term “aralkyloxy” as used herein by itself or as part of another group refers to an aralkyl attached to a terminal oxygen atom. A non-limiting exemplary aralkyloxy group is PhCH2O—.


The term “(cyano)alkyl” as used herein by itself or as part of another group refers to an alkyl substituted with one, two, or three cyano groups. In one embodiment, the alkyl is substituted with one cyano group. In another embodiment, the alkyl is a C1-C6 alkyl In another embodiment, the alkyl is a C1-C4 alkyl. Non-limiting exemplary (cyano)alkyl groups include —CH2CH2CN and —CH2CH2CH2CN.


The term “(cycloalkyl)alkyl” as used herein by itself or as part of another group refers to an alkyl substituted with one or two optionally substituted cycloalkyl groups. In one embodiment, the cycloalkyl group(s) is an optionally substituted C3-C6 cycloalkyl. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1 or C2 alkyl. In another embodiment, the alkyl is substituted with one optionally substituted cycloalkyl group. In another embodiment, the alkyl is substituted with two optionally substituted cycloalkyl groups. Non-limiting exemplary (cycloalkyl)alkyl groups include:




embedded image


The term “sulfonamido” as used herein by itself or as part of another group refers to a radical of the formula —SO2NR50aR50b, wherein R50a and R50b are each independently hydrogen, alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, or optionally substituted heteroaryl; or R50a and R50b taken together with the nitrogen to which they are attached form a 3- to 8-membered optionally substituted heterocyclo group. Non-limiting exemplary sulfonamido groups include —SO2NH2, —SO2N(H)CH3, and —SO2N(H)Ph.


The term “alkylcarbonyl” as used herein by itself or as part of another group refers to a carbonyl group, i.e., —C(═O)—, substituted by an alkyl group. In one embodiment, the alkyl is a C1-C4 alkyl. A non-limiting exemplary alkylcarbonyl group is —COCH3.


The term “arylcarbonyl” as used herein by itself or as part of another group refers to a carbonyl group, i.e., —C(═O)—, substituted by an optionally substituted aryl group. A non-limiting exemplary arylcarbonyl group is —COPh.


The term “alkylsulfonyl” as used herein by itself or as part of another group refers to a sulfonyl group, i.e., —SO2—, substituted by an alkyl group. A non-limiting exemplary alkylsulfonyl group is —SO2CH3.


The term “arylsulfonyl” as used herein by itself or as part of another group refers to a sulfonyl group, i.e., —SO2—, substituted by an optionally substituted aryl group. A non-limiting exemplary arylsulfonyl group is —SO2Ph.


The term “mercaptoalkyl” as used herein by itself or as part of another group refers to an alkyl substituted by a —SH group.


The term “carboxy” as used by itself or as part of another group refers to a radical of the formula —C(═O)OH.


The term “ureido” as used herein by itself or as part of another group refers to a radical of the formula —NR51a—C(═O)—NR51bR51c, wherein R51a is hydrogen or alkyl; and R51b and R51c are each independently hydrogen, alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, or optionally substituted heteroaryl, or R51b and R51c taken together with the nitrogen to which they are attached form a 4- to 8-membered optionally substituted heterocyclo group. Non-limiting exemplary ureido groups include —NH—C(C═O)—NH2 and —NH—C(C═O)—NHCH3.


The term “guanidino” as used herein by itself or as part of another group refers to a radical of the formula —NR52a—C(═NR53)—NR52bR52c, wherein R52a is hydrogen or alkyl; R52b and R53c are each independently hydrogen, alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, or optionally substituted heteroaryl; or R52b and R52c taken together with the nitrogen to which they are attached form a 4- to 8-membered optionally substituted heterocyclo group; and R53 is hydrogen, alkyl, cyano, alkylsulfonyl, alkylcarbonyl, carboxamido, or sulfonamido. Non-limiting exemplary guanidino groups include —NH—C(C═NH)—NH2, —NH—C(C═NCN)—NH2, and —NH—C(C═NH)—NHCH3.


The term “(heterocyclo)alkyl” as used herein by itself or as part of another group refers to an alkyl substituted with one, two, or three optionally substituted heterocyclo groups. In one embodiment, the alkyl is substituted with one optionally substituted 5- to 8-membered heterocyclo group. In another embodiment, alkyl is a C1-C6 alkyl. In another embodiment, alkyl is a C1-C4 alkyl. The heterocyclo group can be linked to the alkyl group through a carbon or nitrogen atom. Non-limiting exemplary (heterocyclo)alkyl groups include:




embedded image


The term “carbamate” as used herein by itself or as part of another group refers to a radical of the formula —NR54a—C(═O)—OR54b, wherein R54a is hydrogen or alkyl, and R54b is hydrogen, alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, or optionally substituted heteroaryl. A non-limiting exemplary carbamate group is —NH—(C═O)—OtBu.


The term “(heteroaryl)alkyl” as used herein by itself or as part of another group refers to an alkyl substituted with one or two optionally substituted heteroaryl groups. In one embodiment, the alkyl group is substituted with one optionally substituted 5- to 14-membered heteroaryl group. In another embodiment, the alkyl group is substituted with two optionally substituted 5- to 14-membered heteroaryl groups. In another embodiment, the alkyl group is substituted with one optionally substituted 5- to 9-membered heteroaryl group. In another embodiment, the alkyl group is substituted with two optionally substituted 5- to 9-membered heteroaryl groups. In another embodiment, the alkyl group is substituted with one optionally substituted 5- or 6-membered heteroaryl group. In another embodiment, the alkyl group is substituted with two optionally substituted 5- or 6-membered heteroaryl groups. In one embodiment, the alkyl group is a C1-C6 alkyl. In another embodiment, the alkyl group is a C1-C4 alkyl. In another embodiment, the alkyl group is a C1 or C2 alkyl. Non-limiting exemplary (heteroaryl)alkyl groups include:




embedded image


The term “(amino)(heteroaryl)alkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with one optionally substituted heteroaryl group and one amino group. In one embodiment, the heteroaryl is an optionally substituted 5- to 9-membered heteroaryl group. In another embodiment, the heteroaryl is an optionally substituted 5- or 6-membered heteroaryl group. In one embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1 or C2 alkyl. A non-limiting exemplary (amino)(heteroaryl)alkyl group is:




embedded image


The terms “aralkyl” or “(aryl)alkyl” as used herein by themselves or as part of another group refers to an alkyl substituted with one, two, or three optionally substituted aryl groups. In one embodiment, the alkyl is substituted with one optionally substituted aryl group. In another embodiment, the alkyl is substituted with two optionally substituted aryl groups. In one embodiment, the aryl is an optionally substituted phenyl or optionally substituted naphthyl. In another embodiment, the aryl is an optionally substituted phenyl. In one embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. In another embodiment, the alkyl is a C1 or C2 alkyl. Non-limiting exemplary (aryl)alkyl groups include benzyl, phenethyl, —CHPh2, and —CH(4-F-Ph)2.


The term “amido” as used herein by itself or as part of another group refers to a radical of formula —C(═O)NR60aR60b, wherein R60a and R60b are each independently hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, haloalkyl, (alkoxy)alkyl, (hydroxy)alkyl, (cyano)alkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, (aryl)alkyl, (cycloalkyl)alkyl, (heterocyclo)alkyl, or (heteroaryl)alkyl; or R60a and R60b taken together with the nitrogen to which they are attached from a 4- to 8-membered optionally substituted heterocyclo group. In one embodiment, R60a and R60b are each independently hydrogen or C1-C6 alkyl.


The term “(amido)(aryl)alkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with one amido group and one optionally substituted aryl group. In one embodiment, the aryl group is an optionally substituted phenyl. In one embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. Non-limiting exemplary (amido)(aryl)alkyl groups include:




embedded image


The term “(amino)(aryl)alkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with one amino group and one optionally substituted aryl group. In one embodiment, the amino group is —NH2, alkylamino, or dialkylamino. In one embodiment, the aryl group is an optionally substituted phenyl. In one embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. Non-limiting exemplary (amino)(aryl)alkyl groups include:




embedded image


The term “amino” as used by itself or as part of another group refers to a radical of the formula —NR55aR55b, wherein R55a and R55b are independently hydrogen, optionally substituted alkyl, haloalkyl, (hydroxy)alkyl, (alkoxy)alkyl, (amino)alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, (aryl)alkyl, (cycloalkyl)alkyl, (heterocyclo)alkyl, or (heteroaryl)alkyl.


In one embodiment, the amino is —NH2.


In another embodiment, the amino is an “alkylamino,” i.e., an amino group wherein R55a is C1-6 alkyl and R55b is hydrogen. In one embodiment, R55a is C1-C4 alkyl. Non-limiting exemplary alkylamino groups include —N(H)CH3 and —N(H)CH2CH3.


In another embodiment, the amino is a “dialkylamino,” i.e., an amino group wherein R55a and R55b are each independently C1-6 alkyl. In one embodiment, R55a and R55b are each independently C1-C4 alkyl. Non-limiting exemplary dialkylamino groups include —N(CH3)2 and —N(CH3)CH2CH(CH3)2.


In another embodiment, the amino is a “hydroxyalkylamino,” i.e., an amino group wherein R55a is (hydroxyl)alkyl and R55b is hydrogen or C1-C4 alkyl.


In another embodiment, the amino is a “cycloalkylamino,” i.e., an amino group wherein R55a is optionally substituted cycloalkyl and R55b is hydrogen or C1-C4 alkyl.


In another embodiment, the amino is a “aralkylamino,” i.e., an amino group wherein R55a is aralkyl and R55b is hydrogen or C1-C4 alkyl. Non-limiting exemplary aralkylamino groups include —N(H)CH2Ph, —N(H)CHPh2, and —N(CH3)CH2Ph.


In another embodiment, the amino is a “(cycloalkyl)alkylamino,” i.e., an amino group wherein R55a is (cycloalkyl)alkyl and R55b is hydrogen or C1-C4 alkyl. Non-limiting exemplary (cycloalkyl)alkylamino groups include:




embedded image


In another embodiment, the amino is a “(heterocyclo)alkylamino,” i.e., an amino group wherein R55a is (heterocyclo)alkyl and R55b is hydrogen or C1-C4 alkyl. Non-limiting exemplary (heterocyclo)alkylamino groups include:




embedded image


The term “(amino)alkyl” as used herein by itself or as part of another group refers to an alkyl substituted with one amino group. In one embodiment, the amino group is —NH2. In one embodiment, the amino group is an alkylamino. In another embodiment, the amino group is a dialkylamino. In another embodiment, the alkyl is a C1-C6 alkyl. In another embodiment, the alkyl is a C1-C4 alkyl. Non-limiting exemplary (amino)alkyl groups include —CH2NH2, —CH2CH2N(H)CH3, —CH2CH2N(CH3)2, CH2N(H)cyclopropyl, —CH2N(H)cyclobutyl, and —CH2N(H)cyclohexyl, and —CH2CH2CH2N(H)CH2Ph and —CH2CH2CH2N(H)CH2(4-CF3-Ph).


The term “heteroarylenyl” as used herein by itself or part of another group refers to a divalent form of an optionally substituted heteroaryl group, e.g., a 5- to 9-membered heteroarylenyl. In one embodiment, the heteroarylenyl is a 6-membered heteroarylenyl, e.g., heteroarylenyl derived from pyridine. In one embodiment, the heteroarylenyl is a bicyclic 9-membered heteroarylenyl. Exemplary non-limiting exemplary heteroarylenyl groups include:




embedded image


In the present disclosure, the term “alkylenyl” as used herein by itself or part of another group refers to a divalent form of an alkyl group, wherein the alkyl group is either unsubstituted or substituted with one or two groups independently selected from the group consisting of optionally substituted phenyl and optionally substituted 5- or 6-membered heteroaryl. In one embodiment, the alkylenyl is a divalent form of a C1-12 alkyl, i.e., a C1-C12 alkylenyl. In one embodiment, the alkylenyl is a divalent form of a C1-10 alkyl, i.e., a C1-C10 alkylenyl. In one embodiment, the alkylenyl is a divalent form of a C1-8 alkyl, i.e., a C1-C8 alkylenyl. In one embodiment, the alkylenyl is a divalent form of an unsubstituted C1-6 alkyl, i.e., a C1-C6 alkylenyl. In another embodiment, the alkylenyl is a divalent form of an unsubstituted C1-4 alkyl, i.e., a C1-C8 alkylenyl. In another embodiment, the alkylenyl is a divalent form of a C1-4 alkyl substituted with one or two optionally substituted phenyl groups. Non-limiting exemplary alkylenyl groups include —CH2—, —CH2CH2—, —CH(Ph)-, —CH(Ph)CH2—, —CH2CH2CH2—, —CH(Ph)CH2CH2—, —CH2(CH2)2CH2—, —CH(CH2)3CH2—, and —CH2(CH2)4CH2—.


The term “heteroalkylenyl” as used herein by itself or part of another group refers to a divalent form of a heteroalkyl group. In one embodiment, the heteroalkylenyl is a divalent form of a 3- to 20-membered heteroalkyl, i.e., a 3- to 20-membered heteroalkylenyl. In another embodiment, the heteroalkylenyl is a divalent form of a 3- to 10-membered heteroalkyl, i.e., a 3- to 10-membered heteroalkylenyl. In another embodiment, the heteroalkylenyl is a divalent form of a 3- to 8-membered heteroalkyl, i.e., a 3- to 8-membered heteroalkylenyl. In another embodiment, the heteroalkylenyl is a divalent form of a 3- to 6-membered heteroalkyl, i.e., a 3- to 6-membered heteroalkylenyl. In another embodiment, the heteroalkylenyl is a divalent form of a 3- or 4-membered heteroalkyl, i.e., a 3- or 4-membered heteroalkylenyl. In another embodiment, the heteroalkylenyl is a radical of the formula —(CH2CH2O)u1— wherein u1 is 1, 2, 3, 4, 5, or 6. Non-limiting exemplary heteroalkylenyl groups include —CH2OCH2—, —CH2CH2OCH2CH2O—, —CH2OCH2CH2CH2—, and —CH2CH2OCH2CH2OCH2CH2O—.


The term “heterocyclenyl” as used herein by itself or part of another group refers to a divalent form of an optionally substituted heterocyclo group. In another embodiment, the heterocyclenyl is a divalent form of a 4- to 14-membered heterocyclo group, i.e., a 4- to 14-membered heterocyclenyl. In another embodiment, the heterocyclenyl is a divalent form of a 4- to 10-membered heterocyclo group, i.e., a 4- to 10-membered heterocyclenyl. In another embodiment, the heterocyclenyl is a divalent form of a 4- to 8-membered heterocyclo group, i.e., a 4- to 8-membered heterocyclenyl. In one embodiment, the heterocyclenyl is a divalent form of an optionally substituted azetidine. In another embodiment, the heterocyclenyl is a divalent form of an optionally substituted piperidinyl. In another embodiment, the heterocyclenyl is a divalent form of an optionally substituted piperazinyl. Non-limiting exemplary heterocyclenyl groups include:




embedded image


In another embodiment, the heterocyclenyl is a spiroheterocyclenyl.


The term “spiroheterocyclenyl” as used herein by itself or part of another group refers to a divalent form of a spiroheterocyclo. Non-limiting exemplary spiroheterocyclenyl groups include:




embedded image


The term “cycloalkylenyl” as used herein by itself or part of another group refers to a divalent form of an optionally substituted C4-C6 cycloalkyl group. In one embodiment, the cycloalkylenyl is a 4-membered cycloalkylenyl. In another embodiment, the cycloalkylenyl is a 5-membered cycloalkylenyl. In another embodiment, the cycloalkylenyl is a 6-membered cycloalkylenyl. Non-limiting exemplary groups include:




embedded image


The term “phenylenyl” as used herein by itself or part of another group refers to a divalent form of an optionally substituted phenyl group. Non-limiting examples include:




embedded image


The present disclosure encompasses any of the Compounds of the Disclosure being isotopically-labelled (i.e., radiolabeled) by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2H (or deuterium (D)), 3H, 11C, 13C, 14C, 15N, 18O, 17O, 31P 32P, 35S, 18F, and 36Cl, respectively, e.g., 3H, 11C, and 14C. In one embodiment, provided is a composition wherein substantially all of the atoms at a position within the Compound of the Disclosure are replaced by an atom having a different atomic mass or mass number. In another embodiment, provided is a composition wherein a portion of the atoms at a position within the Compound of the disclosure are replaced, i.e., the Compound of the Disclosure is enriched at a position with an atom having a different atomic mass or mass number.” Isotopically-labelled Compounds of the Disclosure can be prepared by methods known in the art.


As noted above, Compounds of the Disclosure contain one or more asymmetric carbon atoms and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms. The present disclosure encompasses the use of all such possible forms, as well as their racemic and resolved forms and mixtures thereof. The individual enantiomers can be separated according to methods known in the art in view of the present disclosure. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that they include both E and Z geometric isomers. All tautomers are also encompassed by the present disclosure.


As used herein, the term “stereoisomers” is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers).


The term “chiral center” or “asymmetric carbon atom” refers to a carbon atom to which four different groups are attached.


The terms “enantiomer” and “enantiomeric” refer to a molecule that cannot be superimposed on its mirror image and hence is optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image compound rotates the plane of polarized light in the opposite direction.


The term “racemic” refers to a mixture of equal parts of enantiomers and which mixture is optically inactive. In one embodiment, Compounds of the Disclosure are racemic.


The term “absolute configuration” refers to the spatial arrangement of the atoms of a chiral molecular entity (or group) and its stereochemical description, e.g., R or S.


The stereochemical terms and conventions used in the specification are meant to be consistent with those described in Pure & Appl. Chem 68:2193 (1996), unless otherwise indicated.


The term “enantiomeric excess” or “ee” refers to a measure for how much of one enantiomer is present compared to the other. For a mixture of R and S enantiomers, the percent enantiomeric excess is defined as |R−S|*100, where R and S are the respective mole or weight fractions of enantiomers in a mixture such that R+S=1. With knowledge of the optical rotation of a chiral substance, the percent enantiomeric excess is defined as ([α]obs/[α]max)*100, where [α]obs is the optical rotation of the mixture of enantiomers and [α]max is the optical rotation of the pure enantiomer. Determination of enantiomeric excess is possible using a variety of analytical techniques, including NMR spectroscopy, chiral column chromatography or optical polarimetry.


The term “about,” as used herein, includes the recited number ±10%. Thus, “about 10” means 9 to 11.


EXAMPLES
Example 1
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 1)



embedded image


Step 1: Synthesis of 2-chloro-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-fluorobenzonitrile and tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in DMSO. To this solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected intermediate was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 275.12.


Step 2: Synthesis of 2-chloro-4-(8-(4-(piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 4-(4-(tert-butoxycarbonyl)piperazin-1-yl)benzoic acid were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(8-(4-(piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 463.21.


Step 3: Synthesis of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 3-(bromomethyl)azetidine-1-carboxylate in CH3CN was added K2CO3 (2 eq.) and KI (20%). The reaction mixture was refluxed for 4 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the intermediate Boc protected compound. 4-(8-(4-(4-(Azetidin-3-ylmethyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 532.27.


Step 4: Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 1)

To a solution of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1H NMR (400 MHz, DMSO-d6) δ 11.08 (d, J=4.8 Hz, 1H), 7.68 (d, J=8.2 Hz, 1H), 7.60-7.58 (m, 1H), 7.34 (d, J=8.5 Hz, 2H), 7.08-7.04 (m, 2H), 6.81 (d, J=2.1 Hz, 1H), 6.72 (d, J=2.4 Hz, 1H), 6.67 (dd, J=8.4, 2.1 Hz, 1H), 6.57 (dd, J=8.9, 2.3 Hz, 1H), 5.09-5.03 (m, 1H), 4.26 (t, J=8.2 Hz, 2H), 3.87 (dd, J=8.6, 5.6 Hz, 4H), 3.60 (t, J=10.5 Hz, 6H), 3.43 (dt, J=24.9, 7.9 Hz, 8H), 2.92-2.84 (m, 1H), 2.63-2.53 (m, 3H), 2.05-1.99 (m, 1H), 1.92 (t, J=7.0 Hz, 2H), 1.55 (t, J=5.8 Hz, 5H), 1.26 (q, J=6.3 Hz, 1H). LC-MS (ESI) m/z (M+H)+: 789.48; calcd: 789.33; >95% purity.


Example 2
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 3)



embedded image


Step 1: Synthesis of 2-chloro-4-(8-(4-(4-(piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-oxopiperidine-1-carboxylate in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the Boc protected compound. 2-Chloro-4-(8-(4-(4-(piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 546.29.


Step 2: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 3)

To a solution of 2-chloro-4-(8-(4-(4-(piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 7.72 (d, J=8.5 Hz, 1H), 7.61 (d, J=8.8 Hz, 1H), 7.43 (s, 1H), 7.33 (d, J=8.4 Hz, 3H), 7.04 (d, J=8.5 Hz, 2H), 6.73 (s, 1H), 6.58 (dd, J=8.9, 2.3 Hz, 1H), 5.09 (dd, J=12.9, 5.4 Hz, 1H), 4.27 (d, J=13.2 Hz, 2H), 3.96 (d, J=12.6 Hz, 2H), 3.76-3.48 (m, 8H), 3.28 (s, 2H), 3.24-2.86 (m, 7H), 2.64-2.54 (m, 2H), 2.26-2.16 (m, 2H), 2.08-2.00 (m, 1H), 1.93 (t, J=7.0 Hz, 2H), 1.73 (qd, J=12.3, 4.0 Hz, 2H), 1.55 (t, J=5.9 Hz, 4H), 1.30-1.19 (m, 1H). LC-MS (ESI) m/z (M+H)+: 803.41; calcd: 803.34; >95% purity.


Example 3
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 44)



embedded image


Step 1: Synthesis of 4-(2-(3-chloro-4-cyanophenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid

2-Chloro-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane at rt. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The t-Bu ester compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 4-(2-(3-Chloro-4-cyanophenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-Bu group using TFA in DCM. ESI-MS: 395.14.


Step 2: Synthesis of 2-chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

4-(2-(3-Chloro-4-cyanophenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and tert-butyl piperazine-1-carboxylate were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 463.21.


Step 3: Synthesis of 2-chloro-4-(8-(4-(4-(piperidin-4-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-(bromomethyl)piperidine-1-carboxylate in CH3CN was added K2CO3 (2 eq.) and KI (20%). The reaction mixture was refluxed for 4 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the intermediate Boc protected compound. 2-Chloro-4-(8-(4-(4-(piperidin-4-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 560.30.


Step 4: Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 44)

To a solution of 2-chloro-4-(8-(4-(4-(piperidin-4-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H), 7.68 (d, J=8.5 Hz, 1H), 7.60 (d, J=8.7 Hz, 1H), 7.37 (d, J=8.6 Hz, 3H), 7.27 (dd, J=8.7, 2.2 Hz, 1H), 7.03 (d, J=8.4 Hz, 2H), 6.74 (d, J=2.1 Hz, 1H), 6.59 (dd, J=8.9, 2.2 Hz, 1H), 5.08 (dd, J=12.8, 5.4 Hz, 2H), 4.09 (d, J=13.1 Hz, 2H), 3.42 (q, J=6.0, 5.3 Hz, 4H), 3.28 (d, J=6.8 Hz, 4H), 3.11-2.84 (m, 8H), 2.58 (ddd, J=18.6, 13.4, 5.9 Hz, 2H), 2.19-1.99 (m, 2H), 1.94-1.81 (m, 4H), 1.65 (dt, J=15.7, 8.0 Hz, 6H), 1.36-1.15 (m, 4H). LC-MS (ESI) m/z (M+H)+: 817.42; calcd: 817.36; >95% purity.


Example 4
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 51)



text missing or illegible when filed


Step 1: Synthesis of 2-chloro-4-(8-(4-(4-(piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-oxopiperidine-1-carboxylate in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH and TEA. The reaction mixture was stirred at r.t. for 6 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the intermediate Boc protected compound. 2-Chloro-4-(8-(4-(4-(piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 546.29.


Step 2: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 51)

To a solution of 2-chloro-4-(8-(4-(4-(piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 7.70 (d, J=8.5 Hz, 1H), 7.59 (d, J=8.8 Hz, 1H), 7.43-7.37 (m, 3H), 7.32 (dd, J=8.7, 2.3 Hz, 1H), 7.02 (d, J=8.3 Hz, 2H), 6.73 (d, J=2.3 Hz, 1H), 6.58 (dd, J=8.9, 2.3 Hz, 1H), 5.12-5.07 (m, 2H), 4.25 (d, J=13.1 Hz, 2H), 3.63-3.38 (m, 8H), 3.29 (d, J=8.6 Hz, 6H), 3.03-2.84 (m, 4H), 2.61 (dt, J=13.8, 4.0 Hz, 2H), 2.20-2.13 (m, 2H), 2.05-1.99 (m, 1H), 1.92 (t, J=7.0 Hz, 2H), 1.68 (h, J=10.7, 7.2 Hz, 7H). LC-MS (ESI) m/z (M+H)+: 803.43; calcd: 803.34; >95% purity.


Example 5
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 63)



embedded image


Step 1: Synthesis of 4-(8-(4-(4-(azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 3-oxoazetidine-1-carboxylate in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH and TEA. The reaction mixture was stirred at r.t. for 6 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the Boc protected compound. 4-(8-(4-(4-(Azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 518.26.


Step 2: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 63)

To a solution of 4-(8-(4-(4-(azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. LC-MS (ESI) m/z (M+H)+: 815.45; calcd: 815.34; >95% purity.


Example 6
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 72)



embedded image


Step 1: Synthesis of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile

To a solution of 2-chloro-4-(8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 3-(bromomethyl)azetidine-1-carboxylate in CH3CN was added K2CO3 (2 eq.) and KI (20%). The reaction mixture was refluxed for 4 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the Boc protected compound. 4-(8-(4-(4-(Azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 532.27.


Step 2: Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 72)

To a solution of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. LC-MS (ESI) m/z (M+H)+: 789.41; calcd: 789.33; >95% purity.


Example 7
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 112)



embedded image


Step 1: Synthesis of 2-chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-fluorobenzonitrile and tert-butyl 3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in DMSO. To this solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 289.13.


Step 2: Synthesis of 4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid

2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The t-Bu ester compound was obtained by removing the solvent under vacuum and purified by flash column. 4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-Bu group using TFA in DCM. ESI-MS: 409.16.


Step 3: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione

tert-Butyl 4-(azetidin-3-yl)piperazine-1-carboxylate and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione were dissolved in DMSO. To this solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-(2,6-Dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione was obtained by removing the Boc group using TFA in DCM. ESI-MS: 397.18.


Step 4: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 112)

4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel to give 2-Chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 7.73 (d, J=8.2 Hz, 1H), 7.60 (d, J=8.8 Hz, 1H), 7.37 (d, J=8.3 Hz, 2H), 7.00 (d, J=8.4 Hz, 2H), 6.92 (s, 1H), 6.84-6.74 (m, 2H), 6.66 (d, J=8.9 Hz, 1H), 5.08 (dd, J=12.8, 5.4 Hz, 1H), 4.39-4.17 (m, 8H), 4.04 (h, J=6.7 Hz, 2H), 3.86-3.64 (m, 4H), 3.44-3.28 (m, 5H), 2.89 (ddd, J=19.0, 14.1, 5.7 Hz, 2H), 2.59 (dd, J=19.8, 5.9 Hz, 2H), 2.25 (dd, J=12.9, 7.7 Hz, 1H), 2.02 (dd, J=12.8, 6.5 Hz, 1H), 1.72 (h, J=5.7 Hz, 2H), 1.58-1.45 (m, 3H), 1.21 (d, J=6.0 Hz, 3H). LC-MS (ESI) m/z (M+H)+: 789.40; calcd: 789.33; >95% purity.


Example 8
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 114)



embedded image


Step 1: Synthesis of 2-chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and tert-butyl piperazine-1-carboxylate were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 477.23.


Step 2: Synthesis of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile

To a solution of 2-chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 3-(bromomethyl)azetidine-1-carboxylate in CH3CN was added K2CO3 (2 eq.) and KI (20%). The reaction mixture was refluxed for 4 h. All volatiles were removed and the residue was chromatographed on silica gel to afford the Boc protected compound. 4-(8-(4-(4-(Azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 546.29.


Step 3: Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 114)

To a solution of 4-(8-(4-(4-(azetidin-3-ylmethyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-chlorobenzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. 2-Chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1H NMR (400 MHz, DMSO-d6) δ 11.08 (s, 1H), 7.68 (d, J=8.2 Hz, 1H), 7.59 (d, J=8.8 Hz, 1H), 7.39 (d, J=8.5 Hz, 2H), 7.04 (d, J=8.4 Hz, 2H), 6.80 (d, J=2.1 Hz, 2H), 6.72-6.60 (m, 2H), 5.07 (dd, J=12.9, 5.4 Hz, 1H), 4.24 (t, J=8.2 Hz, 2H), 4.04 (q, J=6.6 Hz, 1H), 3.85 (dd, J=8.6, 5.6 Hz, 2H), 3.68-3.05 (m, 16H), 2.89 (ddd, J=17.4, 13.9, 5.5 Hz, 1H), 2.64-2.54 (m, 2H), 2.25 (dd, J=12.8, 7.7 Hz, 1H), 2.10-1.98 (m, 1H), 1.76 (td, J=8.2, 7.0, 3.9 Hz, 2H), 1.63-1.46 (m, 3H), 1.37-1.24 (m, 1H), 1.21 (d, J=6.0 Hz, 3H). LC-MS (ESI) m/z (M+H)+: 803.42; calcd: 803.34; >95% purity.


Example 9
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 120)



embedded image


Step 1: Synthesis of 2-chloro-4-(1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-fluorobenzonitrile and tert-butyl 1-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in DMSO. To this solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-4-(1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 289.13.


Step 2: Synthesis of 4-(2-(3-chloro-4-cyanophenyl)-1-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid

2-Chloro-4-(1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The t-Bu ester compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 4-(2-(3-Chloro-4-cyanophenyl)-1-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-Bu group using TFA in DCM. ESI-MS: 409.16.


Step 3: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 120)

4-(2-(3-Chloro-4-cyanophenyl)-1-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4, and purified by flash column chromatography on silica gel to give 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 7.72 (d, J=8.2 Hz, 1H), 7.57 (d, J=8.9 Hz, 1H), 7.43 (d, J=8.4 Hz, 2H), 7.12 (d, J=8.5 Hz, 2H), 6.92 (d, J=2.0 Hz, 1H), 6.79-6.73 (m, 2H), 6.60 (dd, J=9.0, 2.2 Hz, 1H), 5.09 (dd, J=12.8, 5.5 Hz, 1H), 4.35 (dt, J=21.2, 7.6 Hz, 5H), 4.00-3.69 (m, 4H), 3.42 (dt, J=20.2, 10.0 Hz, 10H), 2.95-2.85 (m, 1H), 2.58 (ddd, J=22.8, 13.1, 4.2 Hz, 2H), 2.08-1.91 (m, 3H), 1.79-1.64 (m, 2H), 1.54 (q, J=8.5, 7.2 Hz, 2H), 1.26 (td, J=7.5, 7.0, 4.4 Hz, 1H), 1.04 (d, J=6.2 Hz, 3H). LC-MS (ESI) m/z (M+H)+: 789.43; calcd: 789.33; >95% purity.


Example 10
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 293)



embedded image


Step 1: Synthesis of (S)-2-chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-fluorobenzonitrile and tert-butyl (S)-3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in DMSO. To this solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, and dried over Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 289.13.


Step 2: Synthesis of (S)-4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid

(S)-2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The t-Bu ester compound was obtained by removing the solvent under vacuum and purified by flash column. (S)-4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-Bu group using TFA in DCM. ESI-MS: 409.16.


Step 3: Synthesis of 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 293)

(S)-4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel to give 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.42; calcd: 789.33; >95% purity.


Example 11
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile (Cpd. No. 109)



embedded image


Step 1: Synthesis of 2-chloro-3-methyl-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile

2-Chloro-4-iodo-3-methylbenzonitrile and tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in dioxane. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-Chloro-3-methyl-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 289.13.


Step 2: Synthesis of 4-(2-(3-chloro-4-cyano-2-methylphenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid

2-Chloro-3-methyl-4-(2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane. To this solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 h. The t-Bu ester compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 4-(2-(3-Chloro-4-cyano-2-methylphenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-Bu group using TFA in DCM. ESI-MS: 409.16.


Step 3: Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile (Cpd. No. 109)

4-(2-(3-Chloro-4-cyano-2-methylphenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To this solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 h. The reaction mixture was partitioned between water and ethyl acetate. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel to give 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. 1H NMR (400 MHz, DMSO-d6) δ 11.09 (s, 1H), 7.72 (d, J=8.3 Hz, 1H), 7.54 (d, J=8.7 Hz, 1H), 7.42 (d, J=8.4 Hz, 2H), 7.11 (d, J=8.3 Hz, 2H), 6.95-6.72 (m, 4H), 5.09 (dd, J=12.8, 5.4 Hz, 1H), 4.34 (dd, J=21.4, 8.7 Hz, 6H), 3.80 (s, 3H), 3.52-3.32 (m, 10H), 3.00-2.81 (m, 2H), 2.56 (dd, J=31.2, 14.5 Hz, 3H), 2.06-1.60 (m, 9H). LC-MS (ESI) m/z (M+H)+: 789.43; calcd: 789.33; >95% purity.


Example 12
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 307)



embedded image


embedded image


Steps 1 and 2

Compound 1 (1.5 eq) and 2 (1 eq) were dissolved in DMF, and Cs2CO3 (3.0 eq) was added. The reaction mixture was stirred at 90° C. overnight. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash, hexane and EtOAc). UPLC-MS:, 6.3 min, 390.31. The product was dissolved in 10×DCM, and TFA (2×) was added and stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 4.


Steps 3 and 4

Compound 4 (1.0 eq) and compound 5 (2.0 eq) were dissolved in DMF, and K2CO3 (3.0 eq) was added. The reaction mixture was stirred at 120° C. overnight. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash, Hexane and EtOAc). The product was dissolved in 10×DCM, and TFA (3×) was added and stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 7.


Step 5

Compound 7 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 8 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The basified compound 8 solution was poured into the compound 7 solution, and the reaction mixture was allowed to stir for 0.5 h. The reaction mixture was partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash, hexane and EtOAc).


Step 6

Compound 9 (2.0 eq) was dissolved in DCE (10×), and compound 10 (1.0 eq), and AcOH (3 eq.) were added. The mixture was stirred at rt for 2 h. Molecular sieves (4 angstrom) (3×) were added, and the mixture was stirred for 12 h. NaB(OAc)3H (3.0 eq) was added, and the mixture was stirred at rt overnight to give Cpd. No. 307. UPLC-MS: 3.6 min, 774.23. HPLC 35%.


Example 13
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 311)



embedded image


embedded image


The synthesis of Cpd. No. 311 was similar to the synthesis of Cpd. No. 307 as shown in EXAMPLE 12, except compound 13 was converted to compound 14 as follows. Compound 13 (1.0 eq) was dissolved in DCE (10×), Dess Martin reagent (1.4 eq.) was added. The above mixture was stirred at rt for 2 h. The reaction mixture was placed on a Combiflash and eluted with DCM/MeOH to give Cpd. No. 311. UPLC-MS: 3.7 min, 788.32. HPLC 36%.


Example 14
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 201)



embedded image


embedded image


Steps 1 and 2

Compound 1 (1.5 eq) and 2 (1 eq) were dissolved in DMF, and Cs2CO3 (3.0 eq) was added. The reaction mixture was stirred at 90° C. overnight. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic layer was dried and concentrated and purified by flash column chromatography on silica gel (Combiflash, Hexanes and EtOAc). The product was dissolved in 10×DCM, and TFA (2×) was added with stirring at rt for 2 h. The solvent was removed and dried on the lyophilizer overnight to give compound 4. Compound 2 was synthesized following the procedure described in Journal of Organic Chemistry, 81(9):3509-3519 (2016).


Steps 3 and 4

Compound 4 (1.0 eq.), K2CO3 (4.0 eq), and compound 5 (1.0 eq) were dissolved in DMF (5×). The mixture was stirred at 120˜130° C. overnight. The reaction was partitioned between EtOAc and H2O. The organic layer was dried and concentrated and purified by flash column chromatography on silica gel (Combiflash, Hexanes and EtOAc). The product was dissolved in 10×DCM, and TFA (10×) was added with stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 7.


Steps 5 and 6

Compound 7 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). After 10 minutes, compound 8a (1.3 eq) was added, and the reaction was allowed to stir for 0.5 h. The reaction was partitioned between EtOAc and H2O. The organic layer was dried and concentrated and purified by flash column chromatography on silica gel (Combiflash, Hexanes and EtOAc). The product was dissolved in 10×DCM, and TFA (2×) was added with stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 9.


Steps 7 and 8

Compound 9 (1.0 eq) was dissolved in DCE (10×), and compound 10 (1.8 eq), and AcOH (3 eq.) were added. The above mixture was stirred at rt for 2 h. NaB(OAc)3H (3.0 eq) was added, and the mixture was stirred at rt overnight. After UPLC-MS validating full conversion of compound 9, the reaction mixture was directly placed on cartridge with Celite® at the bottom, and was eluted with DCM and MeOH using Combiflash. The product was dissolved in 10×DCM, and TFA (2×) was added with stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 13.


Step 7

Compound 13 (1.0 eq)) was dissolved in DMF (4×), and compound 14 (2.0 eq) and DIPEA (4 eq.) were added. The above mixture was stirred at 90° C. overnight. LC-MS indicated compound 10 was fully consumed. Cpd. No. 201 was purified by preparative HPLC. UPLC-MS: LC-MS, 4.3 min, 831.42; HPLC 41% ACN in water. Compound 14 was synthesized in one step reaction following the procedure described in J Med Chem 61(2):462-481 (2018).


Example 15
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 200)



embedded image


The synthesis of Cpd. No. 200 was similar to the synthesis of Cpd. No. 201 as shown in EXAMPLE 14, except the reaction of compound 9 with compound 16 as shown in the scheme above.


Step 9 to 17

Compound 9 (1.0 eq) was dissolved in DCE (10×), and compound 10 (1.8 eq), and AcOH (3 eq.) were added. The above mixture was stirred at rt for 2 h. NaB(OAc)3H (3.0 eq) was added and the mixture was stirred at rt for 4 h. After UPLC-MS validating full conversion of compound 9, the reaction was purified by combiflash to give Cpd. No. 200: UPLC-MS: 4.2 min, 817.30; HPLC 42% ACN in water.


Example 16
Synthesis of 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 203)



embedded image


The synthesis of Cpd. No. 203 was similar to the synthesis of Cpd. No. 201 as shown in EXAMPLE 14.


Example 17
Synthesis of 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 202)



embedded image


The synthesis of Cpd. No. 202 is similar to the synthesis of Cpd. No. 200 as shown in EXAMPLE 16.


Example 18
Synthesis of tert-butyl (S)-1-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate (S-2) and tert-butyl (R)-1-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate (R-2)



embedded image


Method 1—Synthesis of Racemic (Rac)-2




embedded image


Step 1

Compound a (1.0 eq) was dissolved in anhydrous THF in a well dried flask at ° C. NaH (1.2 eq) was added. After 0.5 h, the reaction was warmed to rt and stirred for 2˜3 h. The reaction was cooled to 0° C., and TMSCl (1.15 eq) was dropped slowly. After 0.5 h, the reaction was warmed to rt and stirred for 3˜4 h. The reaction was cooled to −78° C., and MeLi.LiBr solution (1.1 eq) was added dropwise. After 5 h, the reaction was quenched with H2O. DCM was added and organic layer was washed and dried. Combiflash: DCM and MeOH. MS: a: 255.23; b: 253.31.


Step 2

Compound b (1.0 eq) was dissolved in THF (10×), and NaBH4 (1.5 eq) was added. After 2 h, the reaction was quenched by water. DCM (20×) was added and the organic layer was washed with NH4OH (conc) and water, and purified by Combiflash: 100% EtOAc to DCM and MeOH.


Method 2—Synthesis of Racemic (Rac)-2




embedded image


The procedure was reported in J. Org. Chem 81:3509-3519 (2016)


Chiral separation of rac-2 (R, S will be determined by further analysis):


Resolution:




embedded image


Step a

Rac-2 (1.0 eq) was dissolved in EtOH (5×) and (L-DTTA (1.0 eq) was dissolved in EtOH (5×). The solutions were combined in an ice-bath slowly with stirring. The mixture was stirred at rt overnight. The precipitate was filtered and dried. The filtrate was collected to be used in step d.


Step b

The solid from step a was dissolved in EtOH (2% water was added) at reflux. The solvent was distilled to a point, at which suspension start to precipitate. The distillation was stopped, and the solution was heated to reflux. EtOH was added slowly until the suspension disappeared. The temperature was slowly decreased, and the solution was stirred at rt for 1 day and 0° C. for 1 h. The suspension was filtered and dried. The filtrate was collected for step d.


Step c

The solid for step b was basified by NaOH and extracted with DCM. The organic layer was washed with water and dried to give S-2.


Step d

The filtrates from step a and step b were combined, and distilled to get a semisolid, which was then basified by NaOH and extracted with DCM and concentrated and dried. The resulting solid was resolved with D-DTTA following the step a, step b and step c to provide R-2.


The yield for S-2 was about 29% and R-2 was 21%.


Example 19
Synthesis of tert-butyl (S)-3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate (S-3) and tert-butyl (R)-3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate (R-3)



embedded image


The same resolution method described above for S-2 and R-2 can be used to prepare S-3 and R-3. S-3 and R-3 can also be prepared using the following chiral synthesis.




embedded image


Example 20
Synthesis of 2-chloro-4-(8-(4-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 151)



embedded image


embedded image


Step 1

To a suspension of 2-chloro-4-fluorobenzonitrile (1.0 g, 6.5 mmol) in DMF (3 mL) was added tert-butyl 3-methyl-313-2,8-diazaspiro[4.5]decane-8-carboxylate (1.65 g, 6.5 mmol, 1 eq), and the reaction mixture was heated to 900 for 10 h. The reaction mixture was cooled and poured into the mixture of ice-water. The reaction mixture was extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine, dried over Na2SO4, and concentrated. The crude was purified on silica gel (Hexane/EtOAc 2:1) to give tert-butyl 2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate as yellow oil. Then 4.0 M Hydrogen chloride solution in dioxane (4 mL) was added and the mixture was stirred for 2 h. The volatiles were evaporated under vacuum to afford Compound 1 as a white solid (1.5 g, 83%).


Step 2

To a Schlenk tube was charged with Compound 1 (1.5 g, 5.4 mmol), tert-butyl 4-iodobenzoate (2.1 g, 7.0 mmol), Pd2(dba)3 (64 mg, 0.07 mmol), Xantphos (81 mg, 0.14 mmol), Cs2CO3 (3.9 g, 12 mmol), toluene (5 mL) under N2. The tube was sealed and heated at 100° C. oil bath for 12 h. The reaction mixture was extracted with EtOAc and the organic layer was washed with brine, dried and concentrated. The residue was purified on silica gel to afford Compound 2 as a dark oil (1.4 g, 56%). ESI: M+H 466.40.


Step 3

Compound 2 was added to 4 ml of TFA. The Mixture was stirred at room temperature overnight. The volatiles were evaporated under vacuum to afford Compound 3 as a yellow oil (1.1 g, 92%). ESI: M+H 410.32.


Step 4

To a solution of Compound 4 (1000 mg, 3.1 mmol)) in DCE (10 mL) was added NaBH(OAc)3 (1.7 g, 8 mmol) and tert-butyl 4-formylpiperidine-1-carboxylate (1.3 g, 6 mmol). The reaction mixture was stirred for 4 h prior to being quenched with Na2CO3 solution (2 M). The reaction mixture was extracted with EtOAc, washed with saturated NaHCO3 solution. The residue was purified by chromatography on silica gel (DCM and methanol) to give tert-butyl 4-((4-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carboxylate. Then 4.0 M hydrogen chloride solution in dioxane (4 mL) was added and the mixture was stirred for 2 h. The volatiles were evaporated under vacuum to afford Compound 5 as a dark solid (935 mg, 71%). ESI: M+H 425.44.


Step 5

To a solution of Compound 3 (100 mg, 0.24 mmol)) in DMF (4 mL) was added Compound 5 (127 mg, 0.30 mmol), DIPEA (78 mg, 0.6 mmol) and HATU (152 mg, 0.4 mmol). The reaction mixture was stirred for 12 h. The reaction mixture was extracted with EtOAc and the organic layer was washed with brine, dried and concentrated. The residue was purified on HPLC to afford the title compound (113 mg, 58%). 1H NMR (400 MHz, MeOD) δ 7.75 (d, J=9.6 Hz, 2H), 7.53 (d, J=8.8 Hz, 2H), 7.48-7.42 (m, 2H), 7.28-7.18 (m, 5H), 6.83-6.80 (m, 2H), 6.72-6.68 (m, 2H), 5.19-5.10 (m, 3H), 4.49-4.44 (m, 4H), 3.60-3.34 (m, 12H), 3.24-2.80 (m, 7H), 2.80-2.14 (m, 6H), 1.98-1.60 (m, 5H), ESI-MS: 817.42.


Example 21
Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5,6,7,8-tetrahydro-1H-pyrrolo[3,4-g]isoquinoline-1,3(2H)-dione (Compound 9)



embedded image


Step 1: Synthesis of dimethyl isoquinoline-6,7-dicarboxylate (Compound 3)

A mixture of 3-bromopyridine-4-carbaldehyde (1, 0.093 g, 0.5 mmol), dimethyl itaconate (2, 0.079 g, 0.5 mmol), Pd(OAc)2 (0.0056 g, 0.025 mmol), PPh3 (0.013 g, 0.05 mmol) and NaOAc (0.123 g, 1.5 mmol) in dioxane (10 mL) was placed in a 50 mL pressure vessel. After the system was flushed with argon, the reaction mixture was allowed to react at 150° C. for 24 h, and then the reaction mixture was cooled to room temperature. The reaction mixture was filtered through Celite® to eliminate inorganic salts and washed by ethyl acetate. Removal of the solvent left a crude mixture which was purified by flash chromatography on silica gel (ethyl acetate-hexane) to give dimethyl isoquinoline-6,7-dicarboxylate (3, 0.082 g, 67%).


Step 2: Synthesis of 2-(tert-butyl) 6,7-dimethyl 3,4-dihydroisoquinoline-2,6,7(1H)-tricarboxylate (Compound 4)

Compound 3 (279.6 mg, 1.14 mmol) was dissolved in mixture solvent of methanol (4 mL) and acetic acid (0.2 mL). PtO2 (30 mg) was added, and the reaction mixture was stirred under hydrogen at room temperature for 4 h. The reaction mixture was filtered through Celite®. The filtrate was collected and concentrated under reduced pressure to give the crude product.


The crude product was dissolved in mixture of THF (4 mL) and water (1 mL), and Na2CO3 (500 mg) and Boc2O (500 mg, 2.28 mmol) were added to the mixture. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated under reduced pressure to remove the THF, and the crude mixture dissolved in water (5 mL) and ethyl acetate (10 mL). The organic layer was separated, washed with water and brine, dried (MgSO4), concentrated under reduced pressure, and purified by flash chromatography on silica gel (ethyl acetate-hexane) to give compound 4 (130 mg).


Step 3: Synthesis of 2-(tert-butoxycarbonyl)-1,2,3,4-tetrahydroisoquinoline-6,7-dicarboxylic acid (Compound 5)

3N NaOH (0.37 mL, 1.12 mmol) was added to a solution of compound 4 (130 mg, 0.37 mmol) in EtOH (3.7 mL) and the resulting mixture heated at 80° C. for 2 h. The reaction was concentrated under reduced pressure and the crude mixture dissolved in water (5 mL) and ethyl acetate (10 mL) and then acidified using 1N HCl to pH ˜4 in an ice bath. The organic layer was separated and the aqueous layer was extracted with ethyl acetate two more times. The combined the organic layers were washed with brine (10 mL), dried (MgSO4), and concentrated under reduced pressure. The crude product was used in the next step without further purification.


Step 4: Synthesis of tert-butyl 1,3-dioxo-1,5,7,8-tetrahydrofuro[3,4-g]isoquinoline-6(3H)-carboxylate (Compound 6)

Compound 5 (the crude product from step 3) was dissolved in acetic anhydride (2 mL) and the reaction mixture was stirred at 100° C. for 3 h. The reaction mixture was cooled to room temperature, and 10 mL ethyl acetate was added. The reaction mixture was washed with water and brine, dried (MgSO4), concentrated under reduced pressure, and purified by flash chromatography on silica gel (ethyl acetate-hexane) to give compound 6 (123.1 mg).


Step 5: Synthesis of tert-butyl 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,7,8-hexahydro-6H-pyrrolo[3,4-g]isoquinoline-6-carboxylate (Cpd. No. 249)

Compound 6 (123.1 mg, 0.41 mmol), compound 7 (73.5 mg, 0.45 mmol) and Et3N (0.17 mL, 1.23 mmol) were added to toluene (5 mL). The reaction mixture was stirred at 80° C. for 3 h and then cooled to room temperature. The reaction was concentrated under reduced pressure and the crude mixture dissolved in water (5 mL) and ethyl acetate (10 mL). The organic layer was separated, washed with water and brine, dried (MgSO4), concentrated under reduced pressure, and purified by flash chromatography (ethyl acetate-hexane) to give Compound 8.


Step 6: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5,6,7,8-tetrahydro-1H-pyrrolo[3,4-g]isoquinoline-1,3(2H)-dione (Compound 9)

Compound 8 (102.1 mg, 0.24 mmol) was added to 1 mL HCl (4M in 1,4-dioxane), and the mixture reaction mixture was stirred at room temperature for 2 h. The 1,4-dioxane was removed under reduced pressure to give compound 9 as the HCl salt.


Example 22
Synthesis of 2-(2,6-dioxopiperidin-3-yl)-6,7-dihydropyrrolo[3,4-f]isoindole-1,3(2H,5H)-dione (Compound 18)



embedded image


Step 1: Synthesis of tert-butyl di(prop-2-yn-1-yl)carbamate (Compound 12)

A solution of N-(tert-butyloxy)carbonyl propargylamine (compound 10; 33.36 g, 215 mmol) in 50 mL of DMF was treated portionwise (4 times) with 60% NaH (10.4 g) at 0° C. After stirring for 30 min at 25° C., 39 mL of an 80% solution of propargyl bromide (compound 11) in toluene was added. The reaction mixture was stirred for an additional 5 h at 25° C., and then quenched with the addition of ice-water. The mixture was extracted with Et2O (3×200 mL), and the combined extracts were washed with saturated aqueous NaCl, dried (Na2SO4), concentrated in vacuo, and purified by flash chromatography on silica gel (ethyl acetate-hexane) to give compound 12.


Step 2: Synthesis of 2-(tert-butyl) 5,6-dimethyl isoindoline-2,5,6-tricarboxylate (Compound 14)

A solution of compound 12 (10.4 g, 53.9 mmol) and dimethyl acetylenedicarboxylate (compound 13, 30.7 g, 216 mmol) in 110 mL of absolute EtOH was degassed by bubbling N2 through the solution for 10 min. To this solution was added 1.0 g (0.02 equiv) of Wilkinson's catalyst [(Ph3P)3RhCl] at 25° C. After being warmed at reflux for 18 h, the reaction mixture was cooled to 25° C. and concentrated in vacuo. The resulting brown residue was diluted in 200 mL of Et2O, and the precipitate was removed by filtration over Celite®. The filtrate was concentrated and the crude product purified by column chromatography on silica gel (20% EtOAc/hexane) to give 4.60 g (26%) of compound 14.


The remaining steps for synthesizing Compound 18 (as the HCl salt) are essentially the same as Steps 3-6 described above in EXAMPLE 21.


Example 23
Synthesis of 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile (Cpd. No. 355)



embedded image


Compound 1 (1.0 eq) was dissolved in DCE (10×), and compound 2 (2.0 eq) and AcOH (3 eq.) were added. The mixture was stirred at rt for 2 h. Molecular sieves (4 angstrom) (3×) were added, and the mixture was stirred for 12 h. NaB(OAc)3H (3.0 eq) was added, and the mixture was stirred at rt overnight. The reaction was concentrated and purified on a Combiflash chromatography system using MeOH/DCM as the eluent to give compound 3 in 70% yield. The product was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 4.


Compound 5 (1.5 eq) and compound 6 (1 eq) were dissolved in DMF, and Cs2CO3 (3.0 eq), Pd2(dba)3 (0.05×), xphose (0.05×) were added. The reaction mixture was stirred overnight at 90° C. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash using hexane and EtOAc at the eluent). The product was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 8.


Compound 8 (1.0 eq) and compound 9 (2.0 eq) were dissolved in DMF, and KHCO3 (3.0 eq) was added. The reaction mixture was stirred at 120° C. for 2 h. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash using Hexane and EtOAc as the eluent). The product was dissolved in 10×DCM, and TFA (5×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 11.


Compound 11 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 4 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The compound 4 solution was poured into the compound 11 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 355 in 39% yield. UPLC-MS: 3.9 min, 788.43.


Example 24
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5-oxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 364)



embedded image


Compound 1 (1.0 eq) was dissolved in DCE (10×), and compound 2 (2.0 eq) and AcOH (3 eq.) were added. The mixture was stirred at rt for 2 h. Molecular sieves (4 angstrom) (3×) were added, and the mixture was stirred for 12 h. NaB(OAc)3H (3.0 eq) was added, and the mixture was stirred at rt overnight. The reaction was concentrated and purified on a Combiflash chromatography system using MeOH/DCM as the eluent to give compound 3 in 90% yield. Compound 3 was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and the product was dried on a lyophilizer overnight to give compound 4.


Compound 5 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 4 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The compound 4 solution was poured into the compound 5 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 364 in 37% yield. UPLC-MS: 3.6 min, 788.36.


Example 25
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-4-fluoropiperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 365)



embedded image


Cpd. No. 365 was synthesized following the procedure of EXAMPLE 24 with the starting chemicals showed in the above scheme.


Example 26
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-4-methoxypiperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 366)



embedded image


Cpd. No. 366 was synthesized following the procedure of EXAMPLE 24 with the starting chemicals showed in the above scheme.


Example 27
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-ethyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 367)



embedded image


embedded image


Compound 4 was prepared using methods described in EXAMPLE 19.


Compound 5 (1.5 eq) and 4 (1 eq) were dissolved in DMF, and Cs2CO3 (3.0 eq) was added. The reaction mixture was stirred overnight at 90° C. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash using hexane and EtOAc as the eluent). The product was dissolved in 10×DCM, and TFA (2×) was added and stirring at rt for 2 h. The product was dissolved in 10×DCM, and TFA (5×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 7.


Compound 8 (1.5 eq) and compound 7 (1 eq) were dissolved in DMF, and Cs2CO3 (3.0 eq), Pd2(dba)3 (0.05×), and xphose (0.05×) were added. The reaction mixture was stirred overnight at 90° C. The reaction mixture was cooled to rt and partitioned between EtOAc and H2O. The organic phase was separated, washed with water, dried over Na2SO4, and purified by flash column chromatography on silica gel (Combiflash using hexane and EtOAc as the eluent). The product was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 10.


Compound 10 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 11 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The compound 11 solution was poured into the compound 10 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 367 in 38% yield. UPLC-MS: 3.7 min, 788.42.


Example 28
Synthesis of 2-chloro-4-((1S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 370)



embedded image


Cpd. No. 370 was prepared following the procedure of EXAMPLE 27 with the starting chemicals showed in the above scheme.


Example 29
Synthesis of 2-chloro-4-((3S)-8-(4-(3-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)azetidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 371)



embedded image


Cpd. No. 371 was prepared following the procedure of EXAMPLE 24 with the starting chemicals showed in the above scheme.


Example 30
Synthesis of 2-chloro-4-((3S)-8-(4-(3-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)azetidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 372)



embedded image


Cpd. No. 372 was prepared following the procedure of EXAMPLE 24 with the starting chemicals showed in the above scheme.


Example 31
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(2-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)-2-oxoethyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 402)



embedded image


embedded image


Compound 2 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq) and compound 3 (1.0 eq). The reaction mixture was allowed to stir for 0.5 h, and was concentrated to give syrup. The crude product was purified on a Combiflash chromatography system using hexane/EtOAc as the eluent to give compound 4. Compound 4 was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 5.


Compound 5 (1.5 eq) and compound 6 (1 eq) were dissolved in ACN, and Cs2CO3 (3.0 eq) was added. The reaction mixture was stirred at rt overnight. The reaction mixture was concentrated and purified by flash column chromatography on silica gel (Combiflash using MeOH and DCM as the eluent). The product was dissolved in 10×DCM, and TFA (2×) was added. The reaction mixture was stirred at rt for 2 h. The solvent was distilled and dried on a lyophilizer overnight to give compound 8.


Compound 8 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 9 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The compound 9 solution was poured into the compound 8 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 402 in 41% yield. UPLC-MS: 4.3 min, 817.424.


Example 33
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-1,2,3,5,6,7-hexahydropyrrolo[3,4-f]isoindole-2-carbonyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 403)



embedded image


embedded image


Compound 2 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq) and compound 3 (1.0 eq). The above reaction mixture was allowed to stir for 0.5 h, and was concentrated to give syrup. The crude product was purified by Combiflash with hexane and EtOAc to give compound 4. The product was dissolved in 10×DCM, and TFA (4×) was added and stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 5.


Compound 5 (1.0 eq.) was dissolved in DCM (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 6 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The basified compound 6 solution was poured into the compound 5 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 403 in 44% yield. UPLC-MS: 4.6 min, 802.40.


Example 34
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(2-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)acetyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 404)



embedded image


Compound 1 (1.0 eq) and 2 (1.0 eq) were dissolved in DMF, and DIPEA (3.0 eq) was added. The reaction mixture was stirred at rt for 0.5 h. The reaction mixture was purified by prep HPLC with 28% of ACN in water. The compound was lyophilized to give 3. The product was dissolved in 10×DCM, and TFA (2×) was added and stirring at rt for 2 h. The solvent was distilled and dried on the lyophilizer overnight to give compound 4.


Compound 4 (1.0 eq.) was dissolved in DMF (5×). DIPEA (2.0 eq) was added followed by HATU (1.3 eq). In a separate flask, compound 5 (1.0 eq) was dissolved in DMF (5×) and DIPEA (2.0 eq) was added slowly. The basified compound 5 solution was poured into the compound 4 solution, and the reaction mixture was allowed to stir for 0.5 h to give Cpd. No. 404 in 40% yield. UPLC-MS: 4.1 min, 817.40.


Example 35
Synthesis of 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 417)



embedded image


Step 1: Synthesis of (S)-5-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)picolinic acid



embedded image


(S)-2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 5-bromopicolinate were dissolved in dioxane. To the solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 hours. The t-butyl ester was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-5-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)picolinic acid was obtained by removing the t-butyl group using TFA in DCM. ESI-MS: 410.15.


Step 2: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione



embedded image


tert-Butyl 4-(azetidin-3-yl)piperazine-1-carboxylate and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione were dissolved in DMSO. To the solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. Water was added. The reaction mixture and extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 2-(2,6-Dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione was obtained by removing the Boc group using TFA in DCM. ESI-MS: 397.18.


Step 3: Synthesis of 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 417)



embedded image


(S)-5-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)picolinic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. 2-Chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 789.32.


Example 36
Synthesis of 2-chloro-4-((3S)-8-(4-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 407)



embedded image


Step 1: Synthesis of (S)-2-chloro-4-(3-methyl-8-(4-(3-(piperazin-1-yl)azetidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile



embedded image


4-(2-(3-Chloro-4-cyanophenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and tert-butyl 4-(azetidin-3-yl)piperazine-1-carboxylate were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-2-Chloro-4-(3-methyl-8-(4-(3-(piperazin-1-yl)azetidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 532.27.


Step 2: Synthesis of 2-chloro-4-((3S)-8-(4-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 407)



embedded image


To a solution of (S)-2-chloro-4-(3-methyl-8-(4-(3-(piperazin-1-yl)azetidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione in DMSO was added DIPEA (5 eq.). The reaction mixture was stirred at 100° C. for 12 hours. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. 2-Chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 788.32.


Example 37
Synthesis of 2-chloro-4-((3S)-8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 410)



embedded image


Step 1: Synthesis of 5-(3-((1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione



embedded image


tert-Butyl (1S,4S)-5-(azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carboxylate and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione were dissolved in DMSO. To the solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 5-(3-((1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione was obtained by removing the Boc group using TFA in DCM. ESI-MS: 409.18.


Step 2: Synthesis of 2-chloro-4-((3S)-8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 410)



embedded image


4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 5-(3-((1S,4S)-2,5-diazabicyclo[2.2.1]heptan-2-yl)azetidin-1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. 2-Chloro-4-((3S)-8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 800.32.


Example 38
Synthesis of 3-(4-(4-((S)-2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoyl)piperazin-1-yl)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidine-3-carbonitrile (Cpd. No. 431)



embedded image


Step 1: Synthesis of 1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-(piperazin-1-yl)azetidine-3-carbonitrile



embedded image


tert-Butyl 4-(3-cyanoazetidin-3-yl)piperazine-1-carboxylate and 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-1,3-dione were dissolved in DMSO. To the solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. 1-(2-(2,6-Dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-(piperazin-1-yl)azetidine-3-carbonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 422.17.


Step 2: Synthesis of 3-(4-(4-((S)-2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoyl)piperazin-1-yl)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidine-3-carbonitrile (Cpd. No. 431)



embedded image


4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-(piperazin-1-yl)azetidine-3-carbonitrile were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. 3-(4-(4-((S)-2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoyl)piperazin-1-yl)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidine-3-carbonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 813.32.


Example 39
Synthesis of 2-chloro-4-((3S)-8-(4-(7-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 408)



embedded image


Step 1: Synthesis of (S)-2-chloro-4-(3-methyl-8-(4-(7-oxo-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile



embedded image


(S)-4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-azaspiro[3.5]nonan-7-one were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4 to give (S)-2-chloro-4-(3-methyl-8-(4-(7-oxo-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. ESI-MS: 530.24.


Step 2: Synthesis of (S)-2-chloro-4-(3-methyl-8-(4-(7-oxo-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 408)



embedded image


To a solution of (S)-2-chloro-4-(3-methyl-8-(4-(7-oxo-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-6,7-dihydropyrrolo[3,4-f]isoindole-1,3(2H,5H)-dione in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford (S)-2-chloro-4-(3-methyl-8-(4-(7-oxo-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. ESI-MS: 813.34.


Example 40
Synthesis of 2-chloro-4-((3S)-8-(4-(6-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 409)



embedded image


Step 1: Synthesis of (S)-2-chloro-4-(8-(4-(6-formyl-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile



embedded image


(S)-4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and 2-azaspiro[3.5]nonan-7-one were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The (S)-2-chloro-4-(8-(4-(6-formyl-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purified by flash column. ESI-MS: 516.23.


Step 2: Synthesis of 2-chloro-4-((3S)-8-(4-(6-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 409)



embedded image


To a solution of (S)-2-chloro-4-(8-(4-(6-formyl-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-6,7-dihydropyrrolo[3,4-f]isoindole-1,3(2H,5H)-dione in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 2-chloro-4-((3S)-8-(4-(6-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. ESI-MS: 799.32.


Example 41
Synthesis of 4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile (Cpd. No. 420)



embedded image


Step 1: Synthesis of (S)-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile



embedded image


4-fluoro-2-(trifluoromethyl)benzonitrile and tert-butyl (S)-3-methyl-2,8-diazaspiro[4.5]decane-8-carboxylate were dissolved in DMSO. To the solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-4-(3-Methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 323.16.


Step 2: Synthesis of (S)-4-(2-(4-cyano-3-(trifluoromethyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-1 benzoic acid



embedded image


(S)-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile and tert-butyl 4-bromobenzoate were dissolved in dioxane. To the solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 hours. The t-butyl ester was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-4-(2-(4-cyano-3-(trifluoromethyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid was obtained by removing the t-butyl group using TFA in DCM. ESI-MS: 443.18.


Step 3: Synthesis of (S)-4-(3-methyl-8-(4-(4-oxopiperidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile



embedded image


(S)-4-(2-(4-cyano-3-(trifluoromethyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and piperidin-4-one were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. (S)-4-(3-methyl-8-(4-(4-oxopiperidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 524.24.


Step 4: Synthesis of 4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile (Cpd. No. 420)



embedded image


To a solution of (S)-4-(3-methyl-8-(4-(4-oxopiperidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-6,7-dihydropyrrolo[3,4-f]isoindole-1,3(2H,5H)-dione in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. ESI-MS: 807.34.


Example 42
Synthesis of 4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile (Cpd. No. 423)



embedded image


Step 1: Synthesis of (S)-4-(8-(4-(4-formylpiperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile



embedded image


(S)-4-(2-(4-cyano-3-(trifluoromethyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and piperidine-4-carbaldehyde were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. (S)-4-(8-(4-(4-formylpiperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 538.26.


Step 2: Synthesis of 4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile (Cpd. No. 423)



embedded image


To a solution of (S)-4-(8-(4-(4-formylpiperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-6,7-dihydropyrrolo[3,4-f]isoindole-1,3(2H,5H)-dione in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. ESI-MS: 821.35.


Example 43
Synthesis of 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-exahydrocyclopenta[f]isoindole-6-carbaldehyde



embedded image


embedded image


Step 1: Synthesis of diethyl 2,2-di(prop-2-yn-1-yl)malonate



embedded image


To a suspension of sodium hydride (60% wt in mineral oil, 4.22 g, 105.5 mmol) in dry THF (100 mL) stirring at −10° C., dimethyl malonate (6.0 mL, 52.5 mmol) was added dropwise over 10 min. The reaction mixture was stirred at −10° C. for 5 min, and then propargyl bromide (80% wt. in toluene, 12.0 mL, 107.7 mmol) was added dropwise. The reaction mixture was warmed to 25° C. and stirred for 20 h. The reaction mixture was then poured into H2O (50 mL) and Et2O (50 mL), and the layers were separated. The aq layer was extracted with Et2O (3×50 mL). The combined organic phases were washed with brine (50 mL), dried over MgSO4, filtered, and concentrated on a rotary evaporator leaving a white solid. The solid was recrystallized from ethyl acetate and hexanes resulting in 9.44 g of a crystalline white solid (84% yield).


Step 2: Synthesis of ethyl 2-(prop-2-yn-1-yl)pent-4-ynoate



embedded image


Dimethyl 2,2-di(2-propynyl)malonate (4.70 g, 22.6 mmol) and lithium chloride (2.95 g, 69.7 mmol) were dissolved in a solution of H2O (1.0 mL, 55.5 mmol) and DMSO (40 mL). This solution was then heated to reflux for 1 h. After cooling, the reaction mixture was poured into CHCl3 (40 mL) and H2O (40 mL). The layers were separated and the aq layer was extracted with CHCl3 (3×40 mL). The combined organic layers were washed with H2O (50 mL) and brine (50 mL), dried, filtered through silica gel, and concentrated, leaving a yellow oil. The crude oil was purified by flash chromatography on a silica gel column using 20% EtOAc in hexanes as S4 the eluent resulting in 3.06 g of a pale yellow oil (90% yield).


Step 3: Synthesis of ethyl 2-(prop-2-yn-1-yl)pent-4-yn-1-ol



embedded image


To a suspension of lithium aluminum hydride (1.25 g, 33.0 mmol) in dry THF (40 mL) stirring at −10° C. was added a solution of methyl 2-(2-propynyl)-4-pentynoate (3.06 g, 20.4 mmol) in dry THF (10 mL). The reaction mixture was allowed to warm to 25° C. and stirred for 12 h. The reaction mixture was then quenched through the dropwise addition of H2O (1.25 mL), an aq 10% NaOH solution (1.25 mL), and then additional H2O (3.75 mL). The reaction mixture was then stirred for 30 min until the suspended solids turned white. The mixture was then filtered, and the solids were washed with diethyl ether (100 mL). The resulting solution was concentrated on a rotary evaporator yielding a pale yellow oil. The crude oil was purified by flash chromatography on a silica gel column using 10% EtOAc in hexanes as the eluent, resulting in 1.95 g of a clear oil (78% yield).


Step 4: Synthesis of dimethyl 2-(hydroxymethyl)-2,3-dihydro-1H-indene-5,6-dicarboxylate



embedded image


A solution of 5 and dimethyl acetylenedicarboxylate (6, 30.7 g, 216 mmol) in 110 mL of absolute EtOH was degassed by bubbling N2 through the solution for 10 min. To this was added 1.0 g (0.02 equiv) of Wilkinson's catalyst [(Ph3P)3RhCl] at 25° C. After being heated at reflux for 18 h, the reaction mixture was cooled to 25° C. and then concentrated in vacuo. The resulting brown residue was diluted in 200 mL of Et2O, and the precipitate was removed by filtration over Celite. The filtrate was concentrated and the crude product purified by column chromatography (20% EtOAc/hexane) to give 4.60 g (26%) of compound 7.


Step 5: Synthesis of 2-(hydroxymethyl)-2,3-dihydro-1H-indene-5,6-dicarboxylic acid



embedded image


NaOH (3N) was added to a solution of 7 in EtOH and stirred at 80° C. for 4 h. The EtOH was removed under reduced pressure, the pH was adjusted to acidity with 2M HCl, and the mixture was extracted with EtOAc. The solvent was removed to afford the product 8 which was used without further purification.


Step 6: Synthesis of 6-(hydroxymethyl)-6,7-dihydro-1H-indeno[5,6-c]furan-1,3(5H)-dione



embedded image


The mixture of 8 in Ac2O was stirred at 120° C. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 9.


Step 7: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-6-(hydroxymethyl)-6,7-dihydrocyclopenta[f]isoindole-1,3(2H,5H)-dione



embedded image


To a solution of 9 and 10 in toluene was added TEA (3 eq.). The mixture was stirred at reflux for 8 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 11.


Step 8: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindole-6-carbaldehyde



embedded image


To a solution of 11 in DCM was added DMP (1.2 eq.). The reaction mixture was stirred at reflux for 4 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindole-6-carbaldehyde. ESI-MS: 326.09.


Example 44
Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5,7-dihydrocyclopenta[f]isoindole-1,3,6(2H)-trione



embedded image


Step 1: Synthesis of hepta-1,6-diyn-4-ol



embedded image


To a solution of n-BuLi in hexane (6.2 eq., 75 mL) in Et2O/hexane (100 mL) was added TMEDA (7.5 mL) and 2 (3.1 eq.) by dropwise at −78° C. The reaction mixture was stirred at −78° C. for 40 min, and then 12 in THF (20 mL) was added dropwise with 10 min. The reaction mixture was warmed to 25° C. and stirred for 2 h. The reaction mixture was then cooled to −78° C. and added 20 mL THF and Paraformaldehyde (13.5 g) in one portion. Then, the mixture was stirred at r.t. overnight. The mixture was added ice-cold NH4Cl solution and extracted with Et2O (3×50 mL). The combined organic phases were washed with brine (50 mL), dried over MgSO4, filtered, and concentrated on a rotary evaporator leaving a white solid. The solid was recrystallized from ethyl acetate and hexanes resulting in 13.


Step 2: Synthesis of dimethyl 2-hydroxy-2,3-dihydro-1H-indene-5,6-dicarboxylate



embedded image


A solution of 13 and dimethyl acetylenedicarboxylate (6, 30.7 g, 216 mmol) in 110 mL of absolute EtOH was degassed by bubbling N2 through the solution for 10 min. To this was added 1.0 g (0.02 equiv) of Wilkinson's catalyst [(Ph3P)3RhCl] at 25° C. After being warmed at reflux for 18 h, the reaction mixture was cooled to 25° C. and then concentrated in vacuo. The resulting brown residue was diluted in 200 mL of Et2O, and the precipitate was removed by filtration over Celite. The filtrate was concentrated and the crude product purified by column chromatography (20% EtOAc/hexane) to give 4.60 g (26%) of compound 14.


Step 3: Synthesis of 2-hydroxy-2,3-dihydro-1H-indene-5,6-dicarboxylic acid



embedded image


NaOH (3N) was added to a solution of 14 in EtOH and stirred at 80° C. for 4 h. Then the EtOH was removed under reduced pressure, the pH was adjusted to acidity with 2M HCl and the mixture was extracted with EtOAc. The solvent was removed to afford the product 15 which was used without further purification.


Step 4: Synthesis of 6-hydroxy-6,7-dihydro-1H-indeno[5,6-c]furan-1,3(5H)-dione



embedded image


The mixture of 15 in Ac2O was stirred at 120° C. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 16.


Step 5: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-6-hydroxy-6,7-dihydrocyclopenta[f]isoindole-1,3(2H,5H)-dione



embedded image


To a solution of 16 and 10 in toluene was added TEA (3 eq.). The mixture was stirred at reflux for 8 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 17.


Step 6: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5,7-dihydrocyclopenta[f]isoindole-1,3,6(2H)-trione



embedded image


To a solution of 17 in DCM was added DMP (1.2 eq.). The reaction mixture was stirred at reflux for 4 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford intermediate 2-(2,6-dioxopiperidin-3-yl)-5,7-dihydrocyclopenta[f]isoindole-1,3,6(2H)-trione. ESI-MS: 312.07.


Example 45
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 428)



embedded image


Step 1: Synthesis of (S)-2-chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile



embedded image


4-(2-(3-chloro-4-cyanophenyl)-2,8-diazaspiro[4.5]decan-8-yl)benzoic acid and tert-butyl piperazine-1-carboxylate were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added. The reaction mixture was extracted by EA, and the organic phase was washed by water and dried by Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-2-Chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the Boc group using TFA in DCM. ESI-MS: 477.23.


Step 2: Synthesis of 2-chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 428)



embedded image


To a solution of (S)-2-chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindole-6-carbaldehyde in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 2-chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. ESI-MS: 787.32.


Example 46
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 429)



embedded image


To a solution of (S)-2-chloro-4-(3-methyl-8-(4-(piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and 2-(2,6-dioxopiperidin-3-yl)-5,7-dihydrocyclopenta[f]isoindole-1,3,6(2H)-trione in DCE was added NaBH(OAc)3 (1.5 eq.), AcOH, and TEA. The reaction mixture was stirred at r.t. for 6 hours. All volatiles were removed and the residue was chromatographed on silica gel to afford 2-chloro-4-((3S)-8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. ESI-MS: 773.31.


Example 47
Synthesis of 2-chloro-4-((3S)-8-(4-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 442) and 2-chloro-4-((3S)-8-(4-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 444)



embedded image


embedded image


Example 48
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 443) and 2-chloro-4-((3S)-8-(4-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 445)



embedded image


embedded image


Example 49
Synthesis of 2-chloro-4-((3S)-8-(2-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyrimidin-5-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 446)



embedded image


Example 50
Synthesis of 2-chloro-4-((3S)-8-(6-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 447)



embedded image


Example 51
Synthesis of 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 505)



embedded image


Step 1: Synthesis of (S)-6-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)nicotinic acid



embedded image


(S)-2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 6-fluoronicotinate were dissolved in DMSO. To the solution was added DIPEA (3 eq.), and the reaction mixture was stirred at 100° C. for 6 hours. The tert-butyl ester compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. (S)-6-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)nicotinic acid was obtained by removing the tert-butyl group using TFA in DCM. ESI-MS: 410.15.


Step 2: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-1,3-dione



embedded image


5,6-Difluoroisobenzofuran-1,3-dione and 3-aminopiperidine-2,6-dione hydrogen chloride were dissolved in toluene. To the solution was added TEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. The final compound was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 294.05.


Step 3: Synthesis of 2-(2,6-dioxopiperidin-3-yl)-5-fluoro-6-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione



embedded image


tert-Butyl 4-(azetidin-3-yl)piperazine-1-carboxylate and 2-(2,6-dioxopiperidin-3-yl)-5,6-difluoroisoindoline-1,3-dione were dissolved in DMSO. To the solution was added DIPEA (5 eq.), and the reaction mixture was stirred at 100° C. for 4 hours. Water was added to the reaction mixture. The reaction mixture was extracted with EA, and the collected organic phase was washed with water and dried with Na2SO4. The Boc protected compound was obtained by removing the solvent under vacuum and purified by flash column chromatography. 2-(2,6-Dioxopiperidin-3-yl)-5-fluoro-6-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione was obtained by removing the Boc group using TFA in DCM. ESI-MS: 415.17.


Step 4: Synthesis of 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 505)

(S)-6-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)nicotinic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added to the reaction mixture. The reaction mixture was extracted with EA, and the collected organic phase was washed by water and dried with Na2SO4. 2-Chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatograph on silica gel. ESI-MS: 807.31.


Example 52
Synthesis of 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 510)



embedded image


Step 1: Synthesis of (S)-4-(2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)-3-fluorobenzoic acid



embedded image


(S)-2-Chloro-4-(3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile and tert-butyl 4-bromo-3-fluorobenzoate were dissolved in dioxane. To the solution was added Pd2(dba)3 (10%), xantphos (10%), and Cs2CO3 (3 eq.), and the reaction mixture was stirred at 100° C. for 6 hours. The tert-butyl ester compound was obtained by removing the solvent under vacuum and purified by flash column chromatography on silica gel. (S)-4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)-3-fluorobenzoic acid was obtained by removing the Boc group using TFA in DCM. ESI-MS: 427.15.


Step 2: Synthesis of 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 510)

(S)-4-(2-(3-Chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)-3-fluorobenzoic acid and 2-(2,6-dioxopiperidin-3-yl)-5-(3-(piperazin-1-yl)azetidin-1-yl)isoindoline-1,3-dione were dissolved in DMF. To the solution was added DIPEA (5 eq.) and HATU (1.2 eq.), and the reaction mixture was stirred at r.t. for 1 hour. Water was added to the reaction mixture. The reaction mixture was extracted with EA, and the collected organic phase was washed with water and dried with Na2SO4. The 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile was obtained by removing the solvent under vacuum and purifying by flash column chromatography on silica gel. ESI-MS: 806.31.


Example 53
Synthesis of 2-Chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3,5,6,8,9-hexahydroazepino[4,5-f]isoindol-7(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 558)



embedded image


Compounds 1 (1.0 eq) and 2 (2.0 eq) were dissolved in DCE (20×), and AcOH (2.0 eq) was added. After 12 h at room temperature, NaB(OAc)3H (4.0 eq) was added. After 2 h, the volatiles were removed and the residue was purified using a Combiflash chromatography system (DCM and MeOH) to give compound 3 in 65% yield.


Compound 3 was dissolved in DCM (10×) and TFA (5×) was added at ambient temperature. The volatiles were removed to give compound 4 in 100% yield.


Compound 5 (1.0 eq), DIPEA (3.0 eq), and HATU (1.4 eq) were dissolved in DMF. After 10 mins, compound 4 (1.0 eq) was added. After 2 h, the reaction mixture was acidified by TFA and purified by preparative HPLC using 45% acetonitrile in water as the eluent to give Cpd. No. 558.


Example 54
Synthesis of 2-chloro-4-((3S)-8-(6-(1′-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-[4,4′-bipiperidine]-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile (Cpd. No. 568)



embedded image


Compound 1 (1.0 eq), compound 2 (1.3 eq), and Cs2CO3 (3.0 eq) were dissolved in DMF and stirred for 4 h at 60° C. The reaction mixture was partitioned between EtOAc and H2O. The organic layer was separated, concentrated, purified using Combiflash chromatography system (hexane and EtOAc) to give ester of compound 3. The ester was dissolved in water, MeOH, and THF, and NaOH (3 N) was added. After 4 h, the reaction mixture was acidified using HCl to pH 1. The volatiles were removed and residue was purified by column column chromatography on silica gel (DCM and MeOH) to give compound 3 in 80% yield.


Compound 3 (1.0 eq), DIPEA (3.0 eq), and HATU (1.4 eq) were dissolved in DMF. After 10 mins, compound 4 (1.0 eq) was added. The reaction was complete in 0.5 h, and the volatiles were removed. The residue was purified using a Combiflash chromatography system (DCM and MeOH) to give compound 5 in 70%.


Compound 5 was dissolved in DCM (10×) and TFA (5X×) was added at ambient temperature. The volatiles were removed to give compound 6 in 100% yield.


Compound 6 (1.0 eq) and DIPEA (4.0 eq) were dissolved in DMF, and compound 7 (1.5 eq) was added. The mixture was stirred at 90° C. for 12 h. UPLC-MS indicated complete conversion of compound 6. The reaction was cooled to rt, acidified with TFA, and purified by preparative HPLC (46% acetonitrile) to give Cpd. No. 568 in 65% yield.


Example 55
Analytical Characterization of Representative Compounds of the Disclosure

The following Compounds of the Disclosure were prepared using the methods described in the EXAMPLEs above and/or synthetic reagents and techniques known in the art.


Cpd. No. 2: 2-chloro-4-(8-(4-(1-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperidin-4-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 816.39; calcd: 816.36; >95% purity.


Cpd. No. 4: 2-chloro-4-(8-(4-(5-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 843.41; calcd: 843.37; >95% purity.


Cpd. No. 5: 2-chloro-4-(8-(4-(5-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 815.37; calcd: 815.34; >95% purity.


Cpd. No. 6: 2-chloro-4-(8-(3-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.33; calcd: 817.36; >95% purity.


Cpd. No. 7: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-fluoropiperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.39; calcd: 835.35; >95% purity.


Cpd. No. 8: 5-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-fluoropiperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 870.33; calcd: 870.37; >95% purity.


Cpd. No. 9: 5-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 824.39; calcd: 824.35; >95% purity.


Cpd. No. 10: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-fluoroazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.35; calcd: 807.32; >95% purity.


Cpd. No. 11: 5-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-fluoroazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 842.31; calcd: 842.34; >95% purity.


Cpd. No. 12: 5-(2-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-fluoroazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-8-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 842.38; calcd: 842.34; >95% purity.


Cpd. No. 13: 5-(2-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-fluoropiperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-8-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 870.41; calcd: 870.37; >95% purity.


Cpd. No. 14: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-methylazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.37; calcd: 803.34; >95% purity.


Cpd. No. 15: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-methoxypiperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 847.32; calcd: 847.37; >95% purity.


Cpd. No. 16: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-methylpiperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.33; calcd: 831.37; >95% purity.


Cpd. No. 17: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-methoxyazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.38; calcd: 819.34; >95% purity.


Cpd. No. 18: 2-chloro-4-(8-(4-(5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 801.37; calcd: 801.33; >95% purity.


Cpd. No. 19: 2-chloro-4-(8-(4-(4-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)azetidin-3-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 858.42; calcd: 858.39; >95% purity.


Cpd. No. 20: 2-chloro-4-(8-(4-(4-(1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 858.41; calcd: 858.39; >95% purity.


Cpd. No. 21: 2-chloro-4-(8-(4-(7-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-2,7-diazaspiro[3.5]nonan-2-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 760.34; calcd: 760.30; >95% purity.


Cpd. No. 22: 2-chloro-4-(8-(4-(5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)hexahydropyrrolo[3,4-c]pyrrol-2(1H)-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.39; calcd: 829.36; >95% purity.


Cpd. No. 23: 2-chloro-4-(8-(4-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-2,7-diazaspiro[3.5]nonan-7-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 760.33; calcd: 760.30; >95% purity.


Cpd. No. 24: 2-chloro-4-(8-(4-(4-(((3R)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)pyrrolidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.37; calcd: 803.34; >95% purity.


Cpd. No. 25: 2-chloro-4-(8-(4-(4-(((3S)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)pyrrolidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.38; calcd: 803.34; >95% purity.


Cpd. No. 26: 2-chloro-4-(8-(4-(4-(2-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)-2-oxoethyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.33; calcd: 803.31; >95% purity.


Cpd. No. 27: 2-chloro-4-(8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)nicotinoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 818.37; calcd: 818.35; >95% purity.


Cpd. No. 28: 2-chloro-4-(8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)nicotinoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.34; calcd: 790.32; >95% purity.


Cpd. No. 29: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)-3-fluorobenzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.37; calcd: 835.35; >95% purity.


Cpd. No. 30: 5-(8-(6-(4-((1-(2-(2,4-dioxocyclohexyl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)nicotinoyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 852.41; calcd: 852.38; >95% purity.


Cpd. No. 31: 4-(8-(4-(4-((1-(2-(2,4-dioxocyclohexyl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 850.37; calcd: 850.39; >95% purity.


Cpd. No. 32: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 775.33; calcd: 775.31; >95% purity.


Cpd. No. 33: 2-chloro-4-(8-(4-(4-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,7,8-hexahydro-6H-pyrrolo[3,4-g]isoquinolin-6-yl)-2-oxoethyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.34; calcd: 817.32; >95% purity.


Cpd. No. 34: 4-(8-(6-(4-((1-(2-(2,4-dioxocyclohexyl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)nicotinoyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 851.42; calcd: 851.39; >95% purity.


Cpd. No. 35: 2-chloro-4-(8-(2-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)pyrimidine-5-carbonyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.37; calcd: 819.35; >95% purity.


Cpd. No. 36: 2-chloro-4-(8-(4-((4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)bicyclo[2.2.2]octan-1-yl)ethynyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.32; calcd: 823.34; >95% purity.


Cpd. No. 37: 2-chloro-4-(8-(4-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.39; calcd: 817.36; >95% purity.


Cpd. No. 38: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.34; calcd: 807.32; >95% purity.


Cpd. No. 39: 2-chloro-4-(8-(4-((1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperidin-4-yl)ethynyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 798.34; calcd: 798.32; >95% purity.


Cpd. No. 40: 2-chloro-4-(8-(4-((1-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)azetidin-3-yl)ethynyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 798.35; calcd: 798.32; >95% purity.


Cpd. No. 41: 2-chloro-4-(8-(4-((1′-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)-[1,4′-bipiperidin]-4-yl)ethynyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 826.37; calcd: 826.35; >95% purity.


Cpd. No. 42: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.39; calcd: 817.36; >95% purity.


Cpd. No. 43: 2-chloro-4-(8-(4-(((1r,4r)-4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)cyclohexyl)ethynyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 797.35; calcd: 797.32; >95% purity.


Cpd. No. 45: 2-chloro-4-(8-(3-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.38; calcd: 817.36; >95% purity.


Cpd. No. 46: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-1-azaspiro[3.3]heptan-6-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.35; calcd: 829.36; >95% purity.


Cpd. No. 47: 2-chloro-4-(8-(4-(5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-1,5-diazocane-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.34; calcd: 831.37; >95% purity.


Cpd. No. 48: 2-chloro-4-(8-(4-(5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)octahydropyrrolo[3,4-c]pyrrole-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.39; calcd: 829.36; >95% purity.


Cpd. No. 49: 2-chloro-4-(8-(4-(2-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,6-diazaspiro[3.4]octane-6-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.38; calcd: 829.36; >95% purity.


Cpd. No. 50: 2-chloro-4-(8-(4-(7-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-3,7-diazabicyclo[3.3.1]nonane-3-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 843.40; calcd: 843.37; >95% purity.


Cpd. No. 51: 2-chloro-4-(8-(4-(8-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,8-diazaspiro[4.5]decan-2-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 857.41; calcd: 857.39; >95% purity.


Cpd. No. 53: 5-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 838.35; calcd: 838.37; >95% purity.


Cpd. No. 54: 2-(difluoromethyl)-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.38; calcd: 833.40; >95% purity.


Cpd. No. 55: 5-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 852.36; calcd: 852.38; >95% purity.


Cpd. No. 56: 2-chloro-4-(8-(4-((3aR,6aS)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)octahydropyrrolo[3,4-c]pyrrole-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.38; calcd: 829.36; >95% purity.


Cpd. No. 57: 2-chloro-4-(8-(4-(8-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,8-diazaspiro[4.5]decan-2-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.39; calcd: 829.36; >95% purity.


Cpd. No. 58: 2-(difluoromethyl)-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.36; calcd: 819.38; >95% purity.


Cpd. No. 59: 2-(difluoromethyl)-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.42; calcd: 833.40; >95% purity.


Cpd. No. 60: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-1,4-diazepane-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.39; calcd: 817.36; >95% purity.


Cpd. No. 61: 2-chloro-4-(8-(4-(6-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,6-diazaspiro[3.3]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 815.37; calcd: 815.34; >95% purity.


Cpd. No. 62: 5-(2-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-8-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 838.39; calcd: 838.37; >95% purity.


Cpd. No. 64: 2-chloro-4-(8-(4-(4-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-2-azaspiro[3.3]heptan-6-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 815.37; calcd: 815.34; >95% purity.


Cpd. No. 65: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.36; calcd: 821.33; >95% purity.


Cpd. No. 66: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-3-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.35; calcd: 821.33; >95% purity.


Cpd. No. 67: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)-2-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.34; calcd: 835.35; >95% purity.


Cpd. No. 68: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)-3-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.37; calcd: 835.35; >95% purity.


Cpd. No. 69: 2-chloro-4-(8-(4-(((1r,4r)-4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,7,8-hexahydro-6H-pyrrolo[3,4-g]isoquinolin-6-yl)methyl)cyclohexyl)ethynyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 783.36; calcd: 783.34; >95% purity.


Cpd. No. 70: 5-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 838.39; calcd: 838.37; >95% purity.


Cpd. No. 71: 5-(2-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-8-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 838.36; calcd: 838.37; >95% purity.


Cpd. No. 73: 4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 851.37; calcd: 851.39; >95% purity.


Cpd. No. 74: 4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 837.35; calcd: 837.37; >95% purity.


Cpd. No. 75: 2-chloro-4-(8-(4-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.37; calcd: 803.34; >95% purity.


Cpd. No. 76: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.36; calcd: 831.34; >95% purity.


Cpd. No. 77: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.35; calcd: 821.33; >95% purity.


Cpd. No. 78: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 845.37; calcd: 845.35; >95% purity.


Cpd. No. 79: 5-(2-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-8-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 838.40; calcd: 838.37; >95% purity.


Cpd. No. 80: 2-chloro-4-(8-(4-((4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)sulfonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 853.33; calcd: 853.33; >95% purity.


Cpd. No. 81: 2-chloro-4-(8-(3-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.32; calcd: 831.34; >95% purity.


Cpd. No. 82: 2-chloro-4-(8-(3-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 845.36; calcd: 845.35; >95% purity.


Cpd. No. 83: 2-chloro-4-(8-(3-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.35; calcd: 817.32; >95% purity.


Cpd. No. 84: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-fluoroazetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.34; calcd: 807.32; >95% purity.


Cpd. No. 85: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-methylazetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.31; calcd: 803.34; >95% purity.


Cpd. No. 86: 2-chloro-4-(8-(4-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.39; calcd: 817.36; >95% purity.


Cpd. No. 87: 2-chloro-4-(8-(4-(6-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,6-diazaspiro[3.3]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 787.33; calcd: 787.31; >95% purity.


Cpd. No. 88: 2-chloro-4-(8-(4-((3aR,6aR)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)octahydropyrrolo[3,4-c]pyrrole-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.37; calcd: 829.36; >95% purity.


Cpd. No. 89: 2-chloro-4-(8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 748.29; calcd: 748.27; >95% purity.


Cpd. No. 90: 2-chloro-4-(8-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazine-1-carbonyl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 748.28; calcd: 748.27; >95% purity.


Cpd. No. 91: 2-chloro-4-(8-(4-((3aR,6aS)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-3a,6a-dimethyloctahydropyrrolo[3,4-c]pyrrole-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 857.37; calcd: 857.39; >95% purity.


Cpd. No. 92: 2-chloro-4-(8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 720.25; calcd: 720.27; >95% purity.


Cpd. No. 93: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-3-methoxyazetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.35; calcd: 819.34; >95% purity.


Cpd. No. 94: 2-chloro-4-(8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,7,8-hexahydro-6H-pyrrolo[3,4-g]isoquinolin-6-yl)methyl)piperidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 788.36; calcd: 788.33; >95% purity.


Cpd. No. 95: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-3-oxo-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.34; calcd: 817.32; >95% purity.


Cpd. No. 96: 2-chloro-4-(8-(4-(3-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)azetidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 732.26; calcd: 732.27; >95% purity.


Cpd. No. 97: 2-chloro-4-(8-(4-(6-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 772.32; calcd: 772.30; >95% purity.


Cpd. No. 98: 2-chloro-4-(8-(4-(6-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 786.34; calcd: 786.32; >95% purity.


Cpd. No. 99: 2-chloro-4-(8-(4-(6-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,7,8-hexahydro-6H-pyrrolo[3,4-g]isoquinolin-6-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 800.32; calcd: 800.33; >95% purity.


Cpd. No. 100: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-3-oxo-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.37; calcd: 831.34; >95% purity.


Cpd. No. 101: 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidin-1-yl)benzoyl)-3-oxo-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 774.31 calcd: 774.28; >95% purity.


Cpd. No. 102: 2-chloro-4-(8-(4-(3-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)azetidine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 746.27; calcd: 746.29; >95% purity.


Cpd. No. 103: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-1,4-diazepane-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.35; calcd: 789.33; >95% purity.


Cpd. No. 104: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-methoxypiperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 847.35; calcd: 847.37; >95% purity.


Cpd. No. 105: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-4-fluoropiperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.33; calcd: 835.35; >95% purity.


Cpd. No. 106: 2-chloro-4-(8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)-3-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 792.33; calcd: 792.31; >95% purity.


Cpd. No. 107: 2-chloro-4-(8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)-2-fluorophenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 792.32; calcd: 792.31; >95% purity.


Cpd. No. 108: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 803.36; calcd: 803.34; >95% purity.


Cpd. No. 110: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 835.38; calcd: 835.35; >95% purity.


Cpd. No. 111: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 849.39; calcd: 849.37; >95% purity.


Cpd. No. 113: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.39; calcd: 817.36; >95% purity.


Cpd. No. 115: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.35; calcd: 831.37; >95% purity.


Cpd. No. 116: 2-chloro-4-(8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 815.36; calcd: 815.34; >95% purity.


Cpd. No. 117: 2-chloro-4-(8-(4-(1′-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)-[3,3′-biazetidine]-1-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 746.31; calcd: 746.29; >95% purity.


Cpd. No. 118: 2-chloro-4-(8-(4-((1R,4R)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 815.36; calcd: 815.34; >95% purity.


Cpd. No. 119: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.36; calcd: 835.35; >95% purity.


Cpd. No. 121: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazin-1-yl)benzoyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 817.37; calcd: 817.35; >95% purity.


Cpd. No. 122: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.35; calcd: 789.33; >95% purity.


Cpd. No. 123: 2-chloro-4-(8-((1r,4r)-4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)cyclohexyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 795.35; calcd: 795.37; >95% purity.


Cpd. No. 124: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazin-1-yl)benzoyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.34; calcd: 789.33; >95% purity.


Cpd. No. 125: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.35; calcd: 803.34; >95% purity.


Cpd. No. 126: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.36; calcd: 803.34; >95% purity.


Cpd. No. 127: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazin-1-yl)benzoyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 831.40; calcd: 831.37; >95% purity.


Cpd. No. 128: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazin-1-yl)benzoyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.36; calcd: 803.34; >95% purity.


Cpd. No. 129: 2-chloro-4-(8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.38; calcd: 829.36; >95% purity.


Cpd. No. 130: 2-chloro-4-(8-(4-((1R,4R)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 829.35; calcd: 829.36; >95% purity.


Cpd. No. 289: 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 774.34; calcd: 774.32; >95% purity.


Cpd. No. 290: 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidin-1-yl)benzoyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 774.36; calcd: 774.32; >95% purity.


Cpd. No. 291: 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)-3-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 792.35; calcd: 792.31; >95% purity.


Cpd. No. 292: 2-chloro-4-(8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 792.36; calcd: 792.31; >95% purity.


Cpd. No. 294: 2-chloro-4-((3R)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.36; calcd: 789.33; >95% purity.


Cpd. No. 295: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 803.39; calcd: 803.34; >95% purity.


Cpd. No. 296: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.36; calcd: 807.32; >95% purity.


Cpd. No. 345: 2-chloro-4-((3R)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.35; calcd: 807.32; >95% purity.


Cpd. No. 344: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.34; calcd: 807.32; >95% purity.


Cpd. No. 346: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 775.39; calcd: 775.35; >95% purity.


Cpd. No. 347: 2-chloro-4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidin-1-yl)benzoyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 774.36; calcd: 774.32; >95% purity.


Cpd. No. 348: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-3-oxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 775.33; calcd: 775.35; >95% purity.


Cpd. No. 349: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 803.37; calcd: 803.34; >95% purity.


Cpd. No. 350: 2-chloro-4-((1S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.35; calcd: 789.33; >95% purity.


Cpd. No. 351: 2-chloro-4-((1R)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-1-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.36; calcd: 789.33; >95% purity.


Cpd. No. 352: 4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.37; calcd: 823.35; >95% purity.


Cpd. No. 353: 2-(difluoromethyl)-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 805.39; calcd: 805.36; >95% purity.


Cpd. No. 354: 5-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 824.37; calcd: 824.35; >95% purity.


Cpd. No. 150: 2-chloro-4-(8-(4-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1-oxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.81 (d, J=9.2 Hz, 2H), 7.70 (d, J=9.0 Hz, 2H), 7.55-7.50 (m, 2H), 7.37-7.02 (m, 4H), 6.92-6.52 (m, 3H), 6.66-6.41 (m, 2H), 5.17-5.12 (m, 3H), 4.78-4.20 (m, 4H), 3.66-3.30 (m, 10H), 2.98-2.77 (m, 10H), 2.88-2.02 (m, 4H), 2.34-1.32 (m, 5H), ESI-MS: 803.40.


Cpd. No. 144: 2-chloro-4-((2-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-2-azaspiro[3.5]nonan-7-yl)oxy)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.97 (s, 2H), 7.72 (d, J=8.8 Hz, 2H), 7.49-7.40 (m, 2H), 7.22-7.14 (m, 2H), 6.99-6.76 (m, 2H), 3.49-3.30 (m, 13H), 2.87-1.54 (m, 17H), ESI-MS: 761.44.


Cpd. No. 143: 2-chloro-4-((2-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-2-azaspiro[3.5]nonan-7-yl)oxy)benzonitrile; 1H NMR (400 MHz, MeOD) δ 8.00 (s, 2H), 7.74 (d, J=9.0 Hz, 2H), 7.47-7.42 (m, 2H), 7.24-7.10 (m, 2H), 7.01-6.85 (m, 2H), 3.50-3.47 (m, 8H), 3.45-3.22 (m, 5H), 2.66-1.35 (m, 19H), ESI-MS: 775.34.


Cpd. No. 142: 2-chloro-4-((2-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenyl)-2-azaspiro[3.5]nonan-7-yl)oxy)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.74 (d, J=8.8 Hz, 2H), 7.70-7.60 (m, 4H), 7.14-7.13 (m, 2H), 7.05-6.93 (m, 2H), 4.94-4.88 (m, 1H), 3.85-3.11 (m, 13H), 2.94-2.22 (m, 9H), 2.01-1.35 (m, 12H), ESI-MS: 804.44.


Cpd. No. 141: 2-chloro-4-((2-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenyl)-2-azaspiro[3.5]nonan-7-yl)oxy)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.75 (d, J=9.2 Hz, 2H), 7.74-7.62 (m, 4H), 7.13-7.10 (m, 2H), 7.00-6.88 (m, 2H), 4.97-4.90 (m, 1H), 3.83-3.07 (m, 13H), 2.98-2.29 (m, 9H), 2.11-1.29 (m, 14H), ESI-MS: 818.40.


Cpd. No. 145: 2-chloro-4-(7-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)phenoxy)-2-azaspiro[3.5]nonan-2-yl)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.76 (d, J=9.0 Hz, 2H), 7.64-7.55 (m, 2H), 7.42-7.37 (m, 2H), 7.15-7.11 (m, 2H), 7.02-6.80 (m, 2H), 4.99-4.88 (m, 1H), 3.77-3.47 (m, 11H), 3.22-2.41 (m, 13H), 2.22-1.39 (m, 12H), ESI-MS: 818.42.


Cpd. No. 146: 2-chloro-4-(7-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)phenoxy)-2-azaspiro[3.5]nonan-2-yl)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.76 (d, J=8.4 Hz, 2H), 7.76-7.58 (m, 4H), 7.16-7.11 (m, 2H), 6.99-6.88 (m, 2H), 4.95-4.90 (m, 1H), 3.80-3.10 (m, 15H), 3.08-2.44 (m, 7H), 2.01-1.35 (m, 12H), ESI-MS: 804.42.


Cpd. No. 147: 2-chloro-4-(7-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenoxy)-2-azaspiro[3.5]nonan-2-yl)benzonitrile; ESI-MS: 775.30.


Cpd. No. 148: 2-chloro-4-(7-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenoxy)-2-azaspiro[3.5]nonan-2-yl)benzonitrile; ESI-MS: 761.30.


Cpd. No. 137: 2-chloro-4-(8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)bicyclo[2.2.2]octane-1-carbonyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile; 1H NMR (400 MHz, MeOD) δ 7.87 (d, J=9.0 Hz, 1H), 7.56-7.12 (m, 4H), 6.97-6.88 (m, 2H), 4.97-4.90 (m, 1H), 3.80-3.02 (m, 17H), 3.71-2.47 (m, 7H), 2.21-1.35 (m, 22H), ESI-MS: 863.39.


Cpd. No. 138: 2-chloro-4-(8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)bicyclo[2.2.2]octane-1-carbonyl)-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile; ESI-MS: 877.42.


Cpd. No. 152: ESI-MS [M+H]: 817.43.


Cpd. No. 153: ESI-MS [M+H]: 826.54.


Cpd. No. 154: ESI-MS [M+H]: 803.65.


Cpd. No. 155: ESI-MS [M+H]: 812.30.


Cpd. No. 156: ESI-MS [M+H]: 812.36.


Cpd. No. 157: ESI-MS [M+H]: 835.37.


Cpd. No. 158: ESI-MS [M+H]: 835.35.


Cpd. No. 159: ESI-MS [M+H]: 852.39.


Cpd. No. 160: ESI-MS [M+H]: 852.27.


Cpd. No. 161: ESI-MS [M+H]: 857.36.


Cpd. No. 162: ESI-MS [M+H]: 835.47.


Cpd. No. 163: ESI-MS [M+H]: 870.43.


Cpd. No. 164: ESI-MS [M+H]: 870.31.


Cpd. No. 165: ESI-MS [M+H]: 804.35.


Cpd. No. 166: ESI-MS [M+H]: 829.47.


Cpd. No. 167: ESI-MS [M+H]: 802.29.


Cpd. No. 168: ESI-MS [M+H]: 838.47.


Cpd. No. 169: ESI-MS [M+H]: 831.28.


Cpd. No. 170: ESI-MS [M+H]: 866.61.


Cpd. No. 171: ESI-MS [M+H]: 866.47.


Cpd. No. 172: ESI-MS [M+H]: 853.36.


Cpd. No. 173: ESI-MS [M+H]: 888.34.


Cpd. No. 174: ESI-MS [M+H]: 888.20.


Cpd. No. 175: ESI-MS [M+H]: 798.37.


Cpd. No. 176: ESI-MS [M+H]: 798.28.


Cpd. No. 177: ESI-MS [M+H]: 852.32.


Cpd. No. 178: ESI-MS [M+H]: 852.41.


Cpd. No. 179: ESI-MS [M+H]: 864.38.


Cpd. No. 180: ESI-MS [M+H]: 774.29.


Cpd. No. 181: ESI-MS [M+H]: 788.22.


Cpd. No. 182: ESI-MS [M+H]: 802.48.


Cpd. No. 183: ESI-MS [M+H]: 760.35.


Cpd. No. 184: ESI-MS [M+H]: 774.29.


Cpd. No. 185: ESI-MS [M+H]: 788.28.


Cpd. No. 186: ESI-MS [M+H]: 817.39.


Cpd. No. 187: ESI-MS [M+H]: 831.37.


Cpd. No. 188: ESI-MS [M+H]: 867.46.


Cpd. No. 189: ESI-MS [M+H]: 881.37.


Cpd. No. 190: ESI-MS [M+H]: 817.38.


Cpd. No. 191: ESI-MS [M+H]: 831.48.


Cpd. No. 192: ESI-MS [M+H]: 817.28.


Cpd. No. 193: ESI-MS [M+H]: 831.39.


Cpd. No. 194: ESI-MS [M+H]: 835.45.


Cpd. No. 195: ESI-MS [M+H]: 849.63.


Cpd. No. 196: ESI-MS [M+H]: 817.42.


Cpd. No. 197: ESI-MS [M+H]: 849.45.


Cpd. No. 198: ESI-MS [M+H]: 861.41.


Cpd. No. 199: ESI-MS [M+H]: 788.50.


Cpd. No. 200: ESI-MS [M+H]: 817.42.


Cpd. No. 201: ESI-MS [M+H]: 831.28.


Cpd. No. 202: ESI-MS [M+H]: 817.29.


Cpd. No. 203: ESI-MS [M+H]: 831.28.


Cpd. No. 204: ESI-MS [M+H]: 831.35.


Cpd. No. 205: ESI-MS [M+H]: 817.27.


Cpd. No. 206: ESI-MS [M+H]: 788.39.


Cpd. No. 207: ESI-MS [M+H]: 774.26.


Cpd. No. 208: ESI-MS [M+H]: 788.31.


Cpd. No. 209: ESI-MS [M+H]: 774.30.


Cpd. No. 301: ESI-MS [M+H]: 817.32.


Cpd. No. 302: ESI-MS [M+H]: 817.36.


Cpd. No. 306: ESI-MS [M+H]: 774.35.


Cpd. No. 311: ESI-MS [M+H]: 788.39.


Cpd. No. 407: 2-chloro-4-((3S)-8-(4-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.34; calcd: 789.33; >95% purity.


Cpd. No. 408: 2-chloro-4-((3S)-8-(4-(7-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)-2-azaspiro[3.5]nonane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 814.33; calcd: 814.35; >95% purity.


Cpd. No. 409: 2-chloro-4-((3S)-8-(4-(6-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)-2-azaspiro[3.3]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 800.35; calcd: 800.33; >95% purity.


Cpd. No. 410: 2-chloro-4-((3S)-8-(4-((1S,4S)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 801.36; calcd: 801.33; >95% purity.


Cpd. No. 411: 2-chloro-4-((3S)-8-(4-((1R,4R)-5-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)-2,5-diazabicyclo[2.2.1]heptane-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 801.34; calcd: 801.33; >95% purity.


Cpd. No. 412: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-6-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.31; calcd: 790.33; >95% purity.


Cpd. No. 413: 2-chloro-4-((3S)-8-(4-(4-(1-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-3-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.32; calcd: 790.33; >95% purity.


Cpd. No. 414: 5-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-3-(trifluoromethyl)picolinonitrile. LC-MS (ESI) m/z (M+H)+: 809.35; calcd: 809.34; >95% purity.


Cpd. No. 415: 4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.38; calcd: 823.36; >95% purity.


Cpd. No. 416: 2-(difluoromethyl)-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 805.36; calcd: 805.37; >95% purity.


Cpd. No. 417: 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.35; calcd: 790.33; >95% purity.


Cpd. No. 418: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.35; calcd: 789.33; >95% purity.


Cpd. No. 419: 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 791.34; calcd: 791.32; >95% purity.


Cpd. No. 420: 4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.36; calcd: 808.35; >95% purity.


Cpd. No. 421: 2-(difluoromethyl)-4-((3S)-8-(4-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.38; calcd: 790.36; >95% purity.


Cpd. No. 422: 2-chloro-4-((3S)-8-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-1,2,3,5,6,7-hexahydropyrrolo[3,4-f]isoindole-2-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 691.23; calcd: 691.25; >95% purity.


Cpd. No. 423: 4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-(trifluoromethyl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 822.38; calcd: 822.36; >95% purity.


Cpd. No. 424: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.35; calcd: 790.33; >95% purity.


Cpd. No. 425: 2-chloro-4-((3S)-8-(6-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.34; calcd: 790.33; >95% purity.


Cpd. No. 426: 2-chloro-4-((3S)-8-(5-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 775.32; calcd: 775.31; >95% purity.


Cpd. No. 427: 2-chloro-4-((3S)-8-(5-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 789.34; calcd: 789.33; >95% purity.


Cpd. No. 428: 2-chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 788.35; calcd: 788.33; >95% purity.


Cpd. No. 429: 2-chloro-4-((3S)-8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 774.34; calcd: 774.32; >95% purity.


Cpd. No. 430: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 788.35; calcd: 788.33; >95% purity.


Cpd. No. 431: 3-(4-(4-((S)-2-(3-chloro-4-cyanophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-8-yl)benzoyl)piperazin-1-yl)-1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidine-3-carbonitrile. LC-MS (ESI) m/z (M+H)+: 814.35; calcd: 814.33; >95% purity.


Cpd. No. 432: 2-chloro-4-((3S)-8-(6-(4-(6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 776.32; calcd: 776.31; >95% purity.


Cpd. No. 434: 4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-fluorobenzonitrile. LC-MS (ESI) m/z (M+H)+: 773.38; calcd: 773.36; >95% purity.


Cpd. No. 435: 4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)-2-fluoro-3-methylbenzonitrile. LC-MS (ESI) m/z (M+H)+: 787.36; calcd: 787.38; >95% purity.


Cpd. No. 436: 2-chloro-4-((3S)-8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindole-6-carbonyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 437: 2-chloro-4-((3S)-8-(4-(4-(2-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-1,2,3,5,6,7-hexahydrocyclopenta[f]isoindol-6-yl)acetyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 438: 2-chloro-4-((3S)-8-(4-(4-(2′-(2,6-dioxopiperidin-3-yl)-1′,3′-dioxo-2′,3′,5′,7′-tetrahydro-1′H-spiro[azetidine-3,6′-cyclopenta[f]isoindol]-1-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 439: 2-chloro-4-((3S)-8-(4-(4-((2′-(2,6-dioxopiperidin-3-yl)-1′,3′-dioxo-2′,3′,5′,7′-tetrahydro-1′H-spiro[azetidine-3,6′-cyclopenta[f]isoindol]-1-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 440: 2-chloro-4-((3S)-8-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3,5,7-tetrahydro-1H-spiro[cyclopenta[f]isoindole-6,4′-piperidin]-1′-yl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 441: 2-chloro-4-((3S)-8-(4-(4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxo-2,3,5,7-tetrahydro-1H-spiro[cyclopenta[f]isoindole-6,4′-piperidin]-1′-yl)methyl)piperidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile.


Cpd. No. 484: 2-chloro-4-((3S)-8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 805.35; calcd: 805.33; >95% purity.


Cpd. No. 485: 2-chloro-4-((3S)-8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.34; calcd: 823.32; >95% purity.


Cpd. No. 486: 2-chloro-4-((3S)-8-(5-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 800.35; calcd: 823.32; >95% purity.


Cpd. No. 487: 2-chloro-4-((3S)-8-(5-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.34; calcd: 823.32; >95% purity.


Cpd. No. 488: 2-chloro-4-((3S)-8-(5-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.35; calcd: 823.33; >95% purity.


Cpd. No. 489: 2-chloro-4-((3S)-8-(6-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.32; calcd: 833.36; >95% purity.


Cpd. No. 490: 2-chloro-4-((3S)-8-(6-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 851.32; calcd: 851.35; >95% purity.


Cpd. No. 491: 2-chloro-4-((3S)-8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.40; calcd: 833.36; >95% purity.


Cpd. No. 492: 2-chloro-4-((3S)-8-(5-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.41; calcd: 833.36; >95% purity.


Cpd. No. 493: 2-chloro-4-((3S)-8-(5-(4-((4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)methyl)piperidine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 851.39; calcd: 851.35; >95% purity.


Cpd. No. 494: 2-chloro-4-((3S)-8-(5-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)methyl)piperazine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 833.32; calcd: 833.36; >95% purity.


Cpd. No. 495: 2-chloro-4-((3S)-8-(5-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.37; calcd: 790.32; >95% purity.


Cpd. No. 496: 2-chloro-4-((3S)-8-(6-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.34; calcd: 790.32; >95% purity.


Cpd. No. 497: 2-chloro-4-((3S)-8-(5-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.37; calcd: 819.34; >95% purity.


Cpd. No. 498: 2-chloro-4-((3S)-8-(5-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyrazin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 837.38; calcd: 837.34; >95% purity.


Cpd. No. 499: 2-chloro-4-((3S)-8-(6-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.30; calcd: 819.35; >95% purity.


Cpd. No. 500: 2-chloro-4-((3S)-8-(6-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 837.38; calcd: 837.34; >95% purity.


Cpd. No. 501: 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 809.35; calcd: 809.31; >95% purity.


Cpd. No. 502: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.38; calcd: 821.34; >95% purity.


Cpd. No. 503: 2-chloro-4-((3S)-8-(6-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 823.37; calcd: 823.33; >95% purity.


Cpd. No. 504: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.29; calcd: 808.32; >95% purity.


Cpd. No. 505: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.35; calcd: 808.32; >95% purity.


Cpd. No. 506: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.38; calcd: 808.32; >95% purity.


Cpd. No. 507: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.35; calcd: 808.32; >95% purity.


Cpd. No. 508: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 818.32; calcd: 818.36; >95% purity.


Cpd. No. 509: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 818.31; calcd: 818.36; >95% purity.


Cpd. No. 510: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 807.38; calcd: 807.32; >95% purity.


Cpd. No. 511: 2-chloro-4-((3S)-8-(4-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.39; calcd: 835.35; >95% purity.


Cpd. No. 512: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 791.37; calcd: 791.32; >95% purity.


Cpd. No. 513: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.40; calcd: 819.35; >95% purity.


Cpd. No. 514: 2-chloro-4-((3S)-8-(5-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 819.32; calcd: 819.35; >95% purity.


Cpd. No. 515: 2-chloro-4-((3S)-8-(4-((3R)-3-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)pyrrolidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.37; calcd: 821.34; >95% purity.


Cpd. No. 516: 2-chloro-4-((3S)-8-(4-((3S)-3-(4-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)pyrrolidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.40; calcd: 803.35; >95% purity.


Cpd. No. 517: 2-chloro-4-((3S)-8-(4-((3R)-3-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)pyrrolidine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.37; calcd: 821.34; >95% purity.


Cpd. No. 518: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.39; calcd: 835.35; >95% purity.


Cpd. No. 519: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.39; calcd: 835.35; >95% purity.


Cpd. No. 520: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.38; calcd: 835.35; >95% purity.


Cpd. No. 521: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.31; calcd: 835.35; >95% purity.


Cpd. No. 522: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 835.31; calcd: 835.35; >95% purity.


Cpd. No. 523: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 809.37; calcd: 809.31; >95% purity.


Cpd. No. 524: 2-chloro-4-((3S)-8-(5-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperidin-4-yl)piperazine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 837.31; calcd: 837.34; >95% purity.


Cpd. No. 525: 2-chloro-4-((3S)-8-(5-(4-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)piperidine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 837.37; calcd: 837.34; >95% purity.


Cpd. No. 526: 2-chloro-4-((3S)-8-(5-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)pyrimidin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 790.38; calcd: 790.33; >95% purity.


Cpd. No. 527: 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 806.37; calcd: 806.33; >95% purity.


Cpd. No. 528: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.37; calcd: 821.34; >95% purity.


Cpd. No. 529: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)-3-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 821.38; calcd: 821.34; >95% purity.


Cpd. No. 530: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-3-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 825.37; calcd: 825.31; >95% purity.


Cpd. No. 531: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)-3-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 839.37; calcd: 839.33; >95% purity.


Cpd. No. 532: 2-chloro-4-((3S)-8-(4-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 825.35; calcd: 825.31; >95% purity.


Cpd. No. 533: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)-2-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 839.37; calcd: 839.33; >95% purity.


Cpd. No. 534: 2-chloro-4-((3S)-8-(4-(4-((6-(2,6-dioxopiperidin-3-yl)-5,7-dioxo-3,5,6,7-tetrahydropyrrolo[3,4-f]isoindol-2(1H)-yl)methyl)piperidine-1-carbonyl)-3-fluorophenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 806.37; calcd: 806.33; >95% purity.


Cpd. No. 535: 2-chloro-4-((3S)-8-(4-(4-((1-(2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)methyl)piperazine-1-carbonyl)phenyl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 803.37; calcd: 803.35; >95% purity.


Cpd. No. 536: 2-chloro-4-((3S)-8-(6-(4-(1-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)azetidin-3-yl)piperazine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 809.37; calcd: 809.31; >95% purity.


Cpd. No. 537: 2-chloro-4-((3S)-8-(6-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)pyridazin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 809.35; calcd: 809.31; >95% purity.


Cpd. No. 538: 2-chloro-4-((3S)-8-(6-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)pyridin-3-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.36; calcd: 808.32; >95% purity.


Cpd. No. 539: 2-chloro-4-((3S)-8-(5-(3-(4-(2-(2,6-dioxopiperidin-3-yl)-6-fluoro-1,3-dioxoisoindolin-5-yl)piperazin-1-yl)azetidine-1-carbonyl)pyridin-2-yl)-3-methyl-2,8-diazaspiro[4.5]decan-2-yl)benzonitrile. LC-MS (ESI) m/z (M+H)+: 808.38; calcd: 808.32; >95% purity


Example 56
Biological Assays
A. Western Blotting Methods

The appropriate cell line, e.g., prostate cancer LNCaP, Vcap, or 22RV1 cell line, was treated with Compounds of the Disclosure as indicated. The treated cells were lysed with RIPA buffer. The AR level in the cell lysates was examined by western blotting and a specific AR antibody (ab194196, Abcam, Cambridge, Mass. 02139) with concentration of 1:20,000. GAPDH was used as a loading control.


The Western blotting analyses data for representative Compounds of the Disclosure are provided in FIGS. 1-14.


B. Band Quantification and DC50 and DC90 Value Calculation


Bands were quantified with ImageJ software. The relative numbers of each band obtained from normalization with its corresponding GAPDH level were compared with Prism 8 software. The DC50 values were produced from Prism 8, and the DC90 values were calculated with an equation=Bottom+(Top−Bottom)/(1+10{circumflex over ( )}((Log EC50−X)*HillSlope) based on DC50 and Hillslope values.


The DC50 and DC90 for Cpd. No. 307 in prostate cancer Vcap cells is 0.046 nM and 0.199 nM, respectively. See FIG. 1.


The DC50 and DC90 for Cpd. No. 293 in prostate cancer Vcap cells is 0.031 nM and 0.41 nM, respectively. See FIG. 2.


The DC50 and DC90 for Cpd. No. 307 in prostate cancer 22RV1 cells is 0.90 nM and 3.1 nM, respectively. See FIG. 3.


The DC50 and DC90 for Cpd. No. 293 in prostate cancer 22RV1 cells is 0.14 nM and 0.23 nM, respectively. See FIG. 4.


The DC50 and DC90 for Cpd. No. 307 in prostate cancer LNCaP cells is 0.082 nM and 0.11 nM, respectively. See FIG. 5.


The DC50 and DC90 for Cpd. No. 293 in prostate cancer LNCaP cells is 0.3 nM and 0.33 nM, respectively. See FIG. 6.


The degradation in Vcap cells of additional representative Compounds of the Disclosure at the concentrations indicated is presented in Table 4.












TABLE 4










VCap Cells











Cpd. No.
10 nM
100 nM







145
D
C



146
D
B



311
A
A



355
A
A



356
A
A



357
A
A



359
A
A



360
A
A



361
A
A



362
A
A



363
A
A



364
A
A



365
A
A



366
A
A



367
A
A



368
A
A



369
A
A



370
A
A



371
A
A



372
A
A



373
A
A



374
A
A



375
A
A



376
A
A



377
B
A



378
B
A



379
A
A



380
A
A



381
A
A



382
A
A



383
A
A



384
A
A



385
A
A



386
A
A



387
A
A



388
A
A



389
A
A



390
A
A



391
A
A



392
A
A



393
A
A



394
A
A



395
D
C



396
D
B



397
C
A



398
B
A



399
B
A



400
A
A



401
A
A



402
A
A



403
A
A



404
A
A



405
A
A



406
D
C



469
D
B



470
D
B



471
B
A



472
C
A



473
D
B







A: >90% degradation (24 hr treatment)



B: >50% degradation but <90% (24 hr treatment)



C: >10% degradation but <50% (24 hr treatment)



D: No significant degradation (24 hr treatment)






The degradation in MDA-MB-453 cells of additional representative Compounds of the Disclosure at the concentrations indicated is presented in Table 5












TABLE 5










MDA-MB-453











Cpd. No.
10 nM
100 nM







480
B
A



481
B
A



482
D
D



483
D
D







A: >90% degradation (24 hr treatment)



B: >50% degradation but <90% (24 hr treatment)



C: >10% degradation but <50% (24 hr treatment)



D: No significant degradation (24 hr treatment)






The DC50's in VCap cells of representative Compounds of the Disclosure are provided in Table 6.












TABLE 6








VCaP



Cpd. No.
DC50 (nM)









484
 1-10



485
 1-10



486
 1-10



487
 1-10



488
 1-10



489
 1-10



490
 1-10



491
 1-10



492
 1-10



493
 1-10



494
 1-10



495
 1-10



496
<1



497
<1



498
 1-10



499
<1



500
 1-10



501
<1



502
<1



503
 1-10



504
<1



505
<1



506
<1



507
10-100



508
<1



509
<1



510
<1



511
<1



512
<1



513
<1



514
<1



515
10-100



516
 1-10



517
 1-10



518
10-100



519
<1



520
<1



521
<1



522
<1



523
<1



524
<1



525
<1



526
<1



527
<1



528
<1



529
 1-10



530
<1



531
 1-10



532
<1



533
<1



534
<1



535
<1



536
<1



537
 1-10



538
 1-10



539
 1-10










C. VCaP Xenograft Model in SCID Mice

Xenograft tumors were established by injecting 5×106 VCaP cells in 50% Matrigel subcutaneously on the dorsal side of severe combined immunodeficient (SCID) mice, obtained from Charles River, one tumor per mouse. When tumors reached ˜100 mm3, mice were randomly assigned to treatment and vehicle control groups. Animals were monitored for any signs of toxicity. The antitumor activity of Cpd. No. 307 and Cpd. No. 293 is shown in FIG. 15 and FIG. 16, respectively. The antitumor activity of other representative Compounds of the Disclosure is shown in FIGS. 28-35.


D. Pharmacokinetics in Mice and Rats

The pharmacokinetics of representative Compounds of the Disclosure were determined after oral and IV dosing at the concentrations indicated in mouse (Table 7). These compounds show surprising oral bioavailability and other PK properties. The vehicles used in these studies are either (i) 10% PEG400+90% PBS (adjusted to pH to 8.0 by 0.5 N NaOH); or (ii) 100% PEG200.









TABLE 7







Mouse PK Sudies



















IV




PO







Cpd.
Dose
T1/2
AUC(0 − t)
VSS
Cl
Dose
Tmax
T1/2
Cmax
AUC(0 − t)
F


No.
mg/kg
h
h * ng/mL
L/kg
L/h/kg
mg/kg
h
h
ng/mL
h * ng/mL
%





















307
2
4.34
50538
0.23
0.04
5
2.0
4.4
9124
85243
67


311
2
7.22
13968
1.1
0.13
5
4.0
5.01
2819
28708
82


442
1
9.2
5237
1.7
0.17
3
2.67
5.37
847
6572
42


443
1
7.1
8436
1.02
0.12
3
2.0
5.38
1869
15486
67


444
1
8.3
7805
1.2
0.11
3
4.0
6.9
1251
15952
68


445
1
7.4
5176
1.53
0.18
3
3.33
5.37
1131
11895
77


448
1
2.74
78
25.3
12.2
3
1.67
4.78
16
60
26


492
1
3.3
277
7.71
3.27
3
0.5
2.1
59
123
15


493
1
4.9
840
4.12
1.22
3
0.7
4.8
224
627
25


494
1
5.4
180
16.9
5.24
3
0.5
2.4
36
49
9


486
1
2.3
166
9.16
5.85
3
1.0
2.0
44
78
16


487
1
2.1
364
2.65
2.69
3
0.8
1.7
189
353
32


488
1
5.0
233
7.75
4.21
3
1.0
3.1
99.6
213
31


489
1
7.45
2135
2.72
0.44
3
1.0
7.5
477
2393
37


490
1
8.7
4922
1.74
0.18
3
1.0
7.1
639
3729
25


491
1
7.7
2644
2.09
0.36
3
1.0
5.7
789
2709
34


588
1
9.5
1076
6.76
0.83
3
1.0
5.9
347
1759
54


484
1
6.1
7768
0.62
0.12
3
1.3
4.5
2375
10650
46


485
1
9.8
2346
2.95
0.39
3
1.0
7.2
1105
5153
73


496
1
5.8
3031
1.46
0.32
3
1.0
4.1
1419
6548
72


495
1
2.6
950
2.59
0.97
3
0.8
3.4
722
3046
107


497
1
2.7
849
2.0
1.13
3
0.5
2.0
336
760
30


499
1
3.6
1916
1.73
0.49
3
1.3
2.0
404
1668
29


498
1
1.9
419
3.22
2.21
3
0.8
2.3
191
572
45


500
1
2.6
1882
1.53
0.47
3
1.7
2.0
711
3747
60


302
1
7.0
5033
1.45
0.19
3
1.3
7.3
526
4054
27


540
1
7.3
7993
0.96
0.12
3
3.3
7.8
1130
14527
61


344
1
6.9
4926
1.58
0.19
3
4.0
6.0
841
9467
64


346
1
8.8
3030
2.7
0.3
3
3.3
7.2
260
2953
32


348
1
7.5
10113
0.9
0.1
3
2.0
7.6
334
3739
12


502
1
10.0
3487
3.0
0.2
3
3.3
9.5
289
3393
32


521
1
8.2
2988
3.1
0.3
3
1.7
7.0
344
2726
30


522
1
8.6
1723
4.6
0.5
3
2.0
6.0
196
1731
33


529
1
10.2
5131
2.2
0.2
3
3.3
8.5
507
6917
45


528
1
8.9
1712
5.7
0.5
3
1.7
8.8
149
1682
33


510
1
8.3
4078
2.1
0.2
3
2.7
7.2
518
6269
51


511
1
8.0
9040
0.9
0.1
3
2.7
6.4
668
7522
28









VI. References



  • (1) Hamdy et al., “Outcomes after Monitoring, Surgery, or Radiotherapy for Localized Prostate Cancer,” N Engl J Med, 2016, 375, 1415-1424.

  • (2) Litwin, M. S.; Tan, H. J. The Diagnosis and Treatment of Prostate Cancer. JAMA, 2017, 317, 2532-2542.

  • (3) Karantanos et al., “Prostate cancer progression after androgen deprivation therapy: mechanisms of castrate resistance and novel therapeutic approaches,” Oncogene. 2013, 32, 5501-511.

  • (4) Harris et al., “Androgen deprivation therapy: progress in understanding mechanisms of resistance and optimizing androgen depletion,” Nat Clin Pract Urol, 2009, 6, 76-85.

  • (5) Narayanan et al., “Destroying the androgen receptor (AR)-potential strategy to treat advanced prostate cancer,” Oncoscience. 2017, 4, 175-177.

  • (6) Crowder et al., “Nuclear Androgen Receptor Regulates Testes Organization and Oocyte Maturation in Zebrafish,” Endocrinology. 2018, 159, 980-993.

  • (7) Sundén et al., “Synthesis and Biological Evaluation of Second-Generation Tropanol-Based Androgen Receptor Modulators,” J. Med. Chem. 2015, 58, 1569-1574.

  • (8) Oksala et al., “A Novel Nonsteroidal Compound for the Treatment of Castration-Resistant Prostate Cancer by blocking the Androgen Receptor and Inhibiting CYP17A1,” J Steroid Biochem Mol Biol. 2018, doi: 10.1016/j.jsbmb.2018.02.004.

  • (9) Watson et al., “Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer,” Nat Rev Cancer. 2015, 15, 701-711.

  • (10) Guo et al., “Discovery of Aryloxy Tetramethylcyclobutanes as Novel Androgen Receptor Antagonists,” J. Med. Chem. 2011, 54, 7693-7704.

  • (11) Moilanen et al., “Discovery of ODM-201, a new generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies,” Sci Rep. 2015, 5, 12007.

  • (12) Guerrini et al., “A New Avenue toward Androgen Receptor Pan-antagonists: C2 Sterically Hindered Substitution of Hydroxy-propanamides,” J. Med. Chem. 2014, 57, 7263-7279.

  • (13) Jung et al., “Structure-activity relationship for thiohydantoin androgen receptor antagonists for castration-resistant prostate cancer (CRPC),” J. Med. Chem. 2010, 53, 2779-2796.

  • (14) Yamamoto et al., “Design, synthesis, and biological evaluation of 4-arylmethyl-1-phenylpyrazole and 4-aryloxy-1-phenylpyrazole derivatives as novel androgen receptor antagonists,” Bioorg Med Chem. 2012, 20, 2338-2352.

  • (15) Balbas et al., “Overcoming mutation-based resistance to antiandrogens with rational drug design,” Elife. 2013, 2, e00499.

  • (16) Lottrup et al., “Identification of a novel androgen receptor mutation in a family with multiple components compatible with the testicular dysgenesis syndrome,” J Clin Endocrinol Metab. 2013, 98, 2223-2229.

  • (17) Zhu et al., “BMI1 regulates androgen receptor in prostate cancer independently of the polycomb repressive complex 1,” Nat Commun. 2018, 9, 500.

  • (18) Munuganti et al., “Identification of a potent antiandrogen that targets the BF3 site of the androgen receptor and inhibits enzalutamide-resistant prostate cancer,” Chem Biol. 2014, 21, 1476-485.

  • (19) Raina et al., “PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer,” Proc Nat Acad Sci USA. 2016, 113, 7124-7129.

  • (20) Zhou et al., “Discovery of a Small-Molecule Degrader of Bromodomain and Extra-Terminal (BET) Proteins with Picomolar Cellular Potencies and Capable of Achieving Tumor Regression,” J. Med. Chem. 2018, 61, 462-481.

  • (21) Gadd et al., “Structural basis of PROTAC cooperative recognition for selective protein degradation,” Nat Chem. Biol. 2017, 13, 514-521.

  • (22) Toure et al., “Small-molecule PROTACS: new approaches to protein degradation,” Angew. Chem. Int. Edn. 2016, 55, 1966-1973.

  • (23) Qin et al., “Discovery of QCA570 as an Exceptionally Potent and Efficacious Proteolysis Targeting Chimera (PROTAC) Degrader of the Bromodomain and Extra-Terminal (BET) Proteins Capable of Inducing Complete and Durable Tumor Regression,” J. Med. Chem. 2018, 61, 6685-6704.

  • (24) Hatcher et al., “Development of Highly Potent and Selective Steroidal Inhibitors and Degraders of CDK8,” ACS Med. Chem. Lett. 2018, 9, 540-545.

  • (25) Gollavilli et al., “EWS/ETS-Driven Ewing Sarcoma Requires BET Bromodomain Proteins,” Cancer Res. 2018, 78, 4760-4773.

  • (26) Bondeson et al., “Targeted Protein Degradation by Small Molecules. Annu Rev Pharmacol Toxicol,” 2017, 57, 107-123.

  • (27) Salami et al., “Androgen receptor degradation by the proteolysis-targeting chimera ARCC-4 outperforms enzalutamide in cellular models of prostate cancer drug resistance,” Commun Biol. 2018, 1, 100.

  • (28) Pal et al., “Identification of mechanisms of resistance to treatment with abiraterone acetate or enzalutamide in patients with castration-resistant prostate cancer (CRPC),” Cancer. 2018, 124, 1216-1224.

  • (29) Wang et al., “Blocking the Feedback Loop between Neuroendocrine Differentiation and Macrophages Improves the Therapeutic Effects of Enzalutamide (MDV3100) on Prostate Cancer,” Clin Cancer Res. 2018, 24, 708-723.

  • (30) Gustafson et al., “Small-Molecule-Mediated Degradation of the Androgen Receptor through Hydrophobic Tagging,” Angew. Chem. Int. Ed. 2015, 54, 9659-9662.

  • (31) Shibata et al., “Development of Protein Degradation Inducers of Androgen Receptor by Conjugation of Androgen Receptor Ligands and Inhibitor of Apoptosis Protein Ligands,” J. Med. Chem. 2018, 61, 543-575.

  • (32) Crew et al., US 20170327469 A1

  • (33) Pereira de Jésus-Tran et al., “Comparison of crystal structures of human androgen receptor ligand-binding domain complexed with various agonists reveals molecular determinants responsible for binding affinity,” Protein Sci. 2006, 15, 987-999.

  • (34) Galdeano et al., “Structure-guided design and optimization of small molecules targeting the protein-protein interaction between the von Hippel-Lindau (VHL) E3 ubiquitin ligase and the hypoxia inducible factor (HIF) alpha subunit with in vitro nanomolar affinities,” J. Med. Chem. 2014, 57, 8657-8663.

  • (35) Soares et al., “Group-Based Optimization of Potent and Cell-Active Inhibitors of the von Hippel-Lindau (VHL) E3 Ubiquitin Ligase: Structure-Activity Relationships Leading to the Chemical Probe (2S,4R)-1-((S)-2-(1-Cyanocyclopropanecarboxamido)-3,3-dimethylbutanoyl)-4-hydroxy-N-(4-(4-methylthiazol-5-yl)benzyl)pyrrolidine-2-carboxamide (VH298),” J. Med. Chem. 2018, 61, 599-618.

  • (36) Buckley et al., “Targeting the von Hippel-Lindau E3 ubiquitin ligase using small molecules to disrupt the VHL/HIF-1α interaction,” J. Am. Chem. Soc. 2012, 134, 4465-4468.

  • (37) Frost et al., “Potent and selective chemical probe of hypoxic signalling downstream of HIF-alpha hydroxylation via VHL inhibition,” Nat Commun, 2016, 7, 13312-13312.

  • (38) Berlin et al., WO2016149668A1

  • (39) Ishoey et al., “Translation Termination Factor GSPT1 Is a Phenotypically Relevant Off-Target of Heterobifunctional Phthalimide Degraders,” ACS Chem. Biol. 2018, 13, 553-560.

  • (40) Powell et al., “Chemically Induced Degradation of Anaplastic Lymphoma Kinase (ALK),” J. Med. Chem. 2018, 61, 4249-4255.

  • (41) Liu et al., “Melatonin Inhibits Androgen Receptor Splice Variant-7 (AR-V7)-Induced Nuclear Factor-Kappa B (NF-κB) Activation and NF-κB Activator-Induced AR-V7 Expression in Prostate Cancer Cells: Potential Implications for the Use of Melatonin in Castration-Resistant Prostate Cancer (CRPC) Therapy,” Int J Mol Sci. 2017, 18, E1130.

  • (42) Sun et al., “Design, synthesis, and characterization of a potent, nonpeptide, cell-permeable, bivalent Smac mimetic that concurrently targets both the BIR2 and BIR3 domains in XIAP,” J. Am. Chem. Soc. 2007, 129, 15279-15294.

  • (43) Lu et al., “SM-164: a novel, bivalent Smac mimetic that induces apoptosis and tumor regression by concurrent removal of the blockade of cIAP-1/2 and XIAP,” Cancer Res. 2008, 68, 9384-9393.

  • (44) Bai et al., “Targeted Degradation of BET Proteins in Triple-Negative Breast Cancer,” Cancer Res. 2017, 77, 2476-2487.

  • (45) Stols et al., “A new vector for high-throughput, ligation-independent cloning encoding a tobacco etch virus protease cleavage site,” Protein Expr Purif. 2002, 25, 8-15.

  • (46) Benoit, et al., “Seamless Insert-Plasmid Assembly at High Efficiency and Low Cost,” PLoS One. 2016, 11, e0153158.



It is to be understood that the foregoing embodiments and exemplifications are not intended to be limiting in any respect to the scope of the disclosure, and that the claims presented herein are intended to encompass all embodiments and exemplifications whether or not explicitly presented herein


All patents and publications cited herein are fully incorporated by reference in their entirety.

Claims
  • 1. A compound of Formula I:
  • 2. The compound of claim 1, wherein E is selected from the group consisting of:
  • 3. The compound of claim 2, wherein E is E-1, or a pharmaceutically acceptable salt or solvate thereof.
  • 4. The compound of claim 3, wherein E-1 is selected from the group consisting of:
  • 5. The compound of claim 4, wherein E-1 is E-1-1, or a pharmaceutically acceptable salt or solvate thereof.
  • 6. The compound of claim 5, wherein R1a and R1b are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 7. The compound of claim 6, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 8. The compound of claim 6, wherein q is 2; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 9. The compound of claim 6, wherein q is 1; r is 0; s is 0; and t is 2, or a pharmaceutically acceptable salt or solvate thereof.
  • 10. The compound of claim 6, wherein q is 0; r is 1; s is 1; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 11. The compound of claim 6, wherein q is 1; r is 1; s is 0; and t is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 12. The compound of claim 5, wherein R1a and R1b are independently C1-C3 alkyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 13. The compound of claim 12, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 14. The compound of claim 5, wherein R1a is C1-C3 alkyl; and R1b is hydrogen or a pharmaceutically acceptable salt or solvate thereof.
  • 15. The compound of claim 14, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 16. The compound of claim 15 of Formula III:
  • 17. The compound of claim 15 of Formula IV:
  • 18. The compound of claim 5, wherein R1a and R1b taken together with the carbon atom to which they are attached form an —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.
  • 19. The compound of claim 18, wherein q, r, s, and t are 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 20. The compound of claim 4, wherein: E-1 is E-1-2;R1c is C1-C3 alkyl;R1d is selected from the group consisting of hydrogen and C1-C3 alkyl; orR1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group,or a pharmaceutically acceptable salt or solvate thereof.
  • 21. The compound of claim 20, wherein R1d is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 22. The compound of claim 21 of Formula V:
  • 23. The compound of claim 21 of Formula VI:
  • 24. The compound of claim 20, wherein R1c and R1d taken together with the carbon atom to which they are attached form an —C(═O)— group, or a pharmaceutically acceptable salt or solvate thereof.
  • 25. The compound of claim 2, wherein E is E-2, or a pharmaceutically acceptable salt or solvate thereof.
  • 26. The compound of claim 25, wherein E-2 is:
  • 27. The compound of claim 2, wherein E is E-3, or a pharmaceutically acceptable salt or solvate thereof.
  • 28. The compound of claim 27, wherein E-3 is:
  • 29. The compound of any one of claims 1-26, wherein X1 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 30. The compound of any one of claims 1-26, wherein X1 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 31. The compound of any one of claims 1-26, wherein X1 is —CR4aR4b—, or a pharmaceutically acceptable salt or solvate thereof.
  • 32. The compound of claim 31, wherein R4a and R4b are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 33. The compound of any one of claims 1-28, wherein X1 is absent, or a pharmaceutically acceptable salt or solvate thereof.
  • 34. The compound of any one of claims 1-33, wherein: A1 is selected from the group consisting of:
  • 35. The compound of claim 34, wherein A1 is A1-2, or a pharmaceutically acceptable salt or solvate thereof.
  • 36. The compound of claim 34 or 35, wherein R5a, R5b, R5c, and R5d are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 37. The compound of any one of claims 1-36, wherein X2 is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 38. The compound of any one of claims 1-36, wherein X2 is —S(═O)2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 39. The compound of any one of claims 1-36, wherein X2 is —O—, or a pharmaceutically acceptable salt or solvate thereof.
  • 40. The compound of any one of claims 1-36, wherein X2 is —CR4cR4d—, or a pharmaceutically acceptable salt or solvate thereof.
  • 41. The compound of claim 40, wherein R4c and R4d are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 42. The compound of any one of claims 1-36, wherein X2 is absent, or a pharmaceutically acceptable salt or solvate thereof.
  • 43. The compound of claim 1 of Formula VII:
  • 44. The compound of any one of claims 1-43, wherein J1 is cycloalkylenyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 45. The compound of any one of claims 1-43, wherein J1 is heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 46. The compound of claim 45, wherein J1 is selected from the group consisting of:
  • 47. The compound of any one of claims 1-46, wherein J1 is absent, or a pharmaceutically acceptable salt or solvate thereof.
  • 48. The compound of any one of claims 1-42 or 44-47, wherein J2 is selected from the group consisting of —(CH2)m1— and —C≡C—; and m1 is 0, 1, or 2, or a pharmaceutically acceptable salt or solvate thereof.
  • 49. The compound of claim 48, wherein J2 is —(CH2)m1—; and m1 is 0, or a pharmaceutically acceptable salt or solvate thereof.
  • 50. The compound of claim 48, wherein J2 is —(CH2)m1—; and m1 is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 51. The compound of claim 48, wherein J2 is —C≡C—, or a pharmaceutically acceptable salt or solvate thereof.
  • 52. The compound of any one of claims 1-42 or 44-51, wherein J3 is selected from the group consisting of cycloalkylenyl and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 53. The compound of any one of claims 1-42 or 44-51, wherein J3 is absent, or a pharmaceutically acceptable salt or solvate thereof.
  • 54. The compound of any one of claims 1-53, wherein J4 is selected from the group consisting of alkylenyl, cycloalkylenyl, and heterocyclenyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 55. The compound of any one of claims 1-54, wherein J4 is absent, or a pharmaceutically acceptable salt or solvate thereof.
  • 56. The compound of any one of claims 1-55, wherein: J5 is selected from the group consisting of —O— and —N(H)—; andB1 is selected from the group consisting of B1-1, B1-2, B1-3, B1-4, B1-15, B1-16, B1-17, B1-18, B1-19, B1-20, B1-21, B1-22, B1-23, B1-24, B1-25, and B1-26, or a pharmaceutically acceptable salt or solvate thereof.
  • 57. The compound of any one of claims 1-54, wherein: J5 is selected from the group consisting of —(CH2)m2— and —O—;m2 is 0;J4 is selected from the group consisting of:
  • 58. The compound of claim 56 or 57, wherein B1 is B1-1, or a pharmaceutically acceptable salt or solvate thereof.
  • 59. The compound of claim 58, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 60. The compound of claim 58, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 61. The compound of claim 56 or 57, wherein B1 is B1-2, or a pharmaceutically acceptable salt or solvate thereof.
  • 62. The compound of claim 61, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 63. The compound of claim 61, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 64. The compound of claim 56 or 57, wherein B1 is B1-3, or a pharmaceutically acceptable salt or solvate thereof.
  • 65. The compound of claim 56 or 57, wherein B1 is B1-4, or a pharmaceutically acceptable salt or solvate thereof.
  • 66. The compound of any one of claims 56-65, wherein R2a, R2b, and R2c are independently selected from the group consisting of hydrogen and fluoro, or a pharmaceutically acceptable salt or solvate thereof.
  • 67. The compound of 66, wherein R2a, R2b, and R2c are hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 68. The compound of any one of claims 1-55, wherein: J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;m2 is 0, 1, 2, or 3; andB1 is selected from the group consisting of B1-5, B1-6, B1-7, B1-27, and B1-28, or a pharmaceutically acceptable salt or solvate thereof.
  • 69. The compound of claim 68, wherein B1 is B1-5, or a pharmaceutically acceptable salt or solvate thereof.
  • 70. The compound of claim 68, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 71. The compound of claim 68 wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 72. The compound of any one of claims 69-71, wherein m is 1 or 2; and n is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 73. The compound of claim 68, wherein B1 is B1-6, or a pharmaceutically acceptable salt or solvate thereof.
  • 74. The compound of claim 73, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 75. The compound of claim 73, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 76. The compound of any one of claims 73-75, wherein m is 1 or 2; and n is 1 or 2, or a pharmaceutically acceptable salt or solvate thereof.
  • 77. The compound of claim 68, wherein B1 is B1-7, or a pharmaceutically acceptable salt or solvate thereof.
  • 78. The compound of claim 77, wherein m is 1 or 2; and n is 1 or 2, or a pharmaceutically acceptable salt or solvate thereof.
  • 79. The compound any one of claims 1-54, wherein: J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;m2 is 0, 1, 2, or 3; andB1 is selected from the group consisting of B1-9, B1-10, and B1-11,or a pharmaceutically acceptable salt or solvate thereof.
  • 80. The compound of claim 79, wherein B1 is B1-9, or a pharmaceutically acceptable salt or solvate thereof.
  • 81. The compound of claim 79, wherein B1 is B1-10, or a pharmaceutically acceptable salt or solvate thereof.
  • 82. The compound of claim 79, wherein B1 is B1-11, or a pharmaceutically acceptable salt or solvate thereof.
  • 83. The compound of any one of claims 79-82, wherein o is 1 or 2; and p is 1 or 2, or a pharmaceutically acceptable salt or solvate thereof.
  • 84. The compound any one of claims 1-54, wherein: J5 is selected from the group consisting of —(CH2)m2— and —C(═O)—;m2 is 0, 1, 2, or 3; andB1 is selected from the group consisting of B1-12, B1-13, and B1-14,or a pharmaceutically acceptable salt or solvate thereof.
  • 85. The compound of claim 84, wherein B1 is B1-12, or a pharmaceutically acceptable salt or solvate thereof.
  • 86. The compound of claim 84, wherein B1 is B1-13, or a pharmaceutically acceptable salt or solvate thereof.
  • 87. The compound of claim 84, wherein B1 is B1-14, or a pharmaceutically acceptable salt or solvate thereof.
  • 88. The compound of any one of claims 79-87, wherein m is 1 or 2; and n is 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 89. The compound of any one of claims 79-81, 83-86, or 88, wherein Z is —CH2—, or a pharmaceutically acceptable salt or solvate thereof.
  • 90. The compound of any one of claims 79-81, 83-86, or 88, wherein Z is —C(═O)—, or a pharmaceutically acceptable salt or solvate thereof.
  • 91. The compound of any one of claims 68-90, wherein R2d and R2e are independently selected from the group consisting of hydrogen and fluoro, or a pharmaceutically acceptable salt or solvate thereof.
  • 92. The compound of any one of claims 56-91, wherein R3 is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 93. The compound of any one of claims 1-55, wherein B1 is selected from the group consisting of:
  • 94. The compound of claim 93, wherein B1 is:
  • 95. The compound of claim 93, wherein B1 is:
  • 96. The compound of any one of claims 1-92, wherein R8 is hydrogen, or a pharmaceutically acceptable salt or solvate thereof.
  • 97. The compound of any one of claims 1-96, wherein R3a is halo, or a pharmaceutically acceptable salt or solvate thereof.
  • 98. The compound of any one of claims 1-96, wherein R3a is C1-C4 alkyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 99. The compound of any one of claims 1-96, wherein R3a is C1-C4 haloalkyl, or a pharmaceutically acceptable salt or solvate thereof.
  • 100. The compound of any one of claims 1-96, wherein R3a is selected from the group consisting of —Cl, —CH3, and —CF3, or a pharmaceutically acceptable salt or solvate thereof.
  • 101. The compound of any one of claims 1-100, wherein Z1 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.
  • 102. The compound of any one of claims 1-101, wherein Z2 is —C(H)═, or a pharmaceutically acceptable salt or solvate thereof.
  • 103. The compound of claim 1 of Formula VIII:
  • 104. The compound of claim 34 of Formula XV:
  • 105. The compound of claim 104, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is selected from the group consisting of:
  • 106. The compound of claim 34 of Formula XVI:
  • 107. The compound of claim 106, or a pharmaceutically acceptable salt or solvate thereof, wherein B1 is selected from the group consisting of:
  • 108. The compound of claim 1 selected from any one or more of the compounds of Table 1, or a pharmaceutically acceptable salt or solvate thereof.
  • 109. A pharmaceutical composition comprising the compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
  • 110. A method of treating cancer in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof.
  • 111. The method of claim 110, wherein the cancer is breast cancer or prostate cancer.
  • 112. The pharmaceutical composition of claim 109 for use in treating cancer.
  • 113. The pharmaceutical composition of claim 112, wherein the cancer is breast cancer or prostate cancer.
  • 114. A compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof, for use in treating of cancer.
  • 115. The compound for use of claim 114, wherein the cancer is breast cancer or prostate cancer.
  • 116. Use of a compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof, for the manufacture of a medicament for treatment of cancer.
  • 117. The use of claim 116, wherein the cancer is breast cancer or prostate cancer.
  • 118. A method of reducing androgen receptor protein within a cell of a patient in need thereof, the method comprising administering to the subject a compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof.
  • 119. A kit comprising the compound of any one of claims 1-108, or a pharmaceutically acceptable salt or solvate thereof, and instructions for administering the compound, or a pharmaceutically acceptable salt or solvate thereof, to a subject having cancer.
  • 120. A compound of Formula II:
GOVERNMENT SUPPORT

This invention was made with government support under CA186786 awarded by the National Institutes of Health. The government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US20/51503 9/18/2020 WO
Provisional Applications (2)
Number Date Country
62902714 Sep 2019 US
63024697 May 2020 US