<<

(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2017/197056 Al 16 November 2017 (16.11.2017) W !P O PCT

(51) International Patent Classification: AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, A61K 47/48 (2006.01) C07D 471/14 (2006.01) CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, C07D 471/04 (2006.01) DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KH, KN, KP, KR, (21) International Application Number: KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, PCT/US20 17/032053 MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, (22) International Filing Date: PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, 10 May 2017 (10.05.2017) SD, SE, SG, SK, SL, SM, ST, SV, SY,TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every (26) Publication Language: English kind of regional protection available): ARIPO (BW, GH, (30) Priority Data: GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, 62/334,41 1 10 May 2016 (10.05.2016) US UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, (71) Applicant: C4 THERAPEUTICS, INC. [US/US]; 675 W. EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV, Kendall Street, Cambridge, MA 02142 (US). MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, (72) Inventors: PHILLIPS, Andrew, J.; c/o C4 Therapeu TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, tics, Inc., 675 W. Kendall Street, Cambridge, MA 02142 KM, ML, MR, NE, SN, TD, TG). (US). NASVESCHUK, Chris, G.; c/o C4 Therapeutics, Inc., 675 W. Kendall Street, Cambridge, MA 02142 (US). Declarations under Rule 4.17: HENDERSON, James, A.; c/o C4 Therapeutics, Inc., 675 — as to applicant's entitlement to apply for and be granted a W. Kendall Street, Cambridge, MA 02142 (US). LIANG, patent (Rule 4.1 7(H)) Yanke; c/o C4 Therapeutics, Inc., 675 W . Kendall Street, — as to the applicant's entitlement to claim the priority of the Cambridge, MA 02 142 (US). FITZGERALD, Mark, E.; c/ earlier application (Rule 4.17(Hi)) o C4 Therapeutics, Inc., 675 W . Kendall Street, Cambridge, Published: MA 02142 (US). MICHAEL, Ryan, E.; c/o C4 Therapeu — with international search report (Art. 21(3)) tics, Inc., 675 W. Kendall Street, Cambridge, MA 02142 — before the expiration of the time limit for amending the (US). claims and to be republished in the event of receipt of (74) Agent: BELLOWS, Brent, R. et al; Knowles Intellectu amendments (Rule 48.2(h)) al Property Strategies, LLC, 400 Perimeter Center Terrace, Suite 200, Atlanta, GA 30346 (US). (81) Designated States (unless otherwise indicated, for every kind of national protection available): AE, AG, AL, AM,

(54) Title: BROMODOMAIN TARGETING DEGRONIMERS FOR TARGET PROTEIN DEGRADATION

FIG 1J

(57) Abstract: This invention provides a Degronimer that has an E3 Ubiquitin Ligase targeting moiety (Degron) that can be linked to a Targeting Ligand for a bromodomain protein selected for in vivo degradation to achieve a therapeutic effect, and methods of use and compositions thereof as well as methods for their preparation. BROMODOMAIN TARGETING DEGRONIMERS FOR TARGET PROTEIN DEGRADATION

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to U.S. Application No. 62/334,41 1, filed May 10, 2016 which is incorporated by reference herein for all purposes.

FIELD OF THE INVENTION

This invention provides a Degronimer that has an E3 Ubiquitin Ligase targeting moiety (Degron) that can be linked to a Targeting Ligand for a bromodomain protein selected for in vivo degradation to achieve a therapeutic effect, and methods of use and compositions thereof as well as methods for their preparation.

BACKGROUND

Protein degradation is a highly regulated and essential process that maintains cellular homeostasis. The selective identification and removal of damaged, misfolded, or excess proteins is achieved via the ubiquitin-proteasome pathway (UPP). The UPP in fact is central to the regulation of almost all cellular processes, including antigen processing, apoptosis, biogenesis of organelles, cell cycling, DNA transcription and repair, differentiation and development, immune response and inflammation, neural and muscular degeneration, morphogenesis of neural networks, modulation of cell surface receptors, channels and the secretory pathway, the response to stress and extracellular modulators, ribosome biogenesis and viral infection. Covalent attachment of multiple ubiquitin molecules by an E3 ubiquitin ligase to a terminal lysine residue marks the protein for proteasome degradation, where the protein is digested into small and eventually into its constituent amino acids that serve as building blocks for new proteins. Defective proteasomal degradation has been linked to a variety of clinical disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, muscular dystrophies, cardiovascular disease, and cancer among others. There are over 600 E3 ubiquitin ligases which facilitate the ubiquitination of different proteins in vivo, which can be divided into four families: HECT-domain E3s, U-box E3s, monomelic RING E3s and multi-subunit E3s. See generally Li et al. (PLOS One, 2008, 3, 1487) titled "Genome-wide and functional annotation of human E3 ubiquitin ligases identifies MULAN, a mitochondrial E3 that regulates the organelle's dynamics and signaling."; Berndsen et al. (Nat. Struct. Mol. Biol., 2014, 21, 301-307) titled "New insights into ubiquitin E3 ligase mechanism"; Deshaies et al. (Ann. Rev. Biochem., 2009, 78, 399-434) titled "RING domain E3 ubiquitin ligases ."; Spratt et al. (Biochem. 2014, 458, 421-437) titled "RBR E3 ubiquitin ligases: new structures, new insights, new questions ."; and Wang et al. (Nat. Rev. Cancer., 2014, 14, 233-347) titled "Roles of F-box proteins in cancer.". In 1995, Gosink et al. (Proc. Natl. Acad. Sci. USA 1995, 92, 9 117-9121) in a publication titled "Redirecting the Specificity of Ubiquitination by Modifying Ubiquitin-Conjugating Enzymes", provided proof of concept in vitro that engineered peptides can selectively direct ubiquitination of intracellular proteins. The publication by Nawaz et al. (Proc. Natl. Acad. Sci. U. S. A. 1999, 96, 1858-1862) titled "Proteasome-Dependent Degradation of the Human Receptor" describes ER degradation which takes advantage of the ubiquitin-proteasome pathway. Proteinex, Inc. filed a patent application in February 1999 that issued as U.S. Patent 6,306,663 claiming a method of generating a compound for activating the ubiquitination of a Target Protein which comprises covalently linking a Target Protein binding element able to bind specifically to the Target Protein via a ubiquitination recognition element. Proteinex described that the invention can be used to control protein levels in eukaryotes. While the '663 patent may have been based on the first patent application to describe the high level concept of how to manipulate the UPP system to degrade selected proteins in vivo, the patent did not provide sufficient detail to allow persons of skill to easily construct the range of proposed compounds. For example, for the ubiquitination recognition elements, the skilled person was told among other things to use standard methods for drug discovery and screen for appropriate small molecules that would bind to the ligase. Proteinex also emphasized the use of peptides as ubiquitination recognition elements, which can pose significant difficulties for oral drug administration. Since then, harnessing the ubiquitin-proteasome pathway for therapeutic intervention has received significant interest from the scientific community. The publication by Zhou et al. from Harvard Medical School (Mol. Cell 2000, 6, 751-756) titled "Harnessing the Ubiquitination Machinery to Target the Degradation of Specific Cellular Proteins" described an engineered receptor capable of directing ubiquitination in mammalian and yeast cells. Following from these early publications and others in the mid to late 1990s, it was also recognized by Craig Crews and coworkers (Yale University) that a molecule that is capable of binding a Target Protein and a ubiquitin ligase may cause the Target Protein to be degraded. Their first description of such compounds was provided in U.S. Patent 7,041,298 filed in September 2000 by Deshaies et al. and granted in May 2006 titled "Proteolysis Targeting Chimeric Pharmaceutical", which described a "PROTAC" consisting of a small molecule binder of MAP- AP-2 linked to a capable of binding the F-box protein β-TRCP. Information in the '298 patent is also presented in the corresponding publication by Sakamoto et al. (Proc. Natl. Acad. Sci. USA 2001, 98, 8554-8559) titled "Protacs: Chimeric Molecules That Target Proteins to the Skpl- Cullin-F Box Complex for Ubiquitination and Degradation". The publication by Sakamoto et al. (Mol. Cell. Proteomics 2003, 2, 1350-1358) titled "Development of Protacs to Target Cancer- Promoting Proteins for Ubiquitination and Degradation" describes an analogous PROTAC (PROTAC2) that instead of degrading MAP-AP-2 degrades estrogen and receptors. The first E3 ligase successfully targeted with a small molecule was MDM2, which ubiquitinates the tumor suppressor p53. The targeting ligand was an HDM2/MDM2 inhibitor identified in Vassilev et al. (Science 2004, 303, 844-848) titled "In Vivo Activation of the P53 Pathway by Small-Molecule Antagonists of MDM2". Other examples of direct small molecule-induced recruitment of Target Proteins to the proteasome for degradation on addition to cultured cells were described in 2004 (Schneekloth et al. (J. Am. Chem. Soc. 2004, 126, 3748-3754) titled "Chemical Genetic Control of Protein Levels: Selective in Vivo Targeted Degradation"). Schneekloth et al. describe a degradation agent (PROTAC3) that targets the FK506 binding protein (FKBP12) and shows that both PROTAC2 and PROTAC3 hit their respective targets with green fluorescent protein (GFP) imaging. The publication by Schneekloth et al. (ChemBioChem 2005, 6, 40-46) titled "Chemical Approaches to Controlling Intracellular Protein Degradation" described the state of the field at the time. The publication by Schneekloth et al. (Bioorg. Med. Chem. Lett. 2008, 18, 5904-5908) titled "Targeted Intracellular Protein Degradation Induced by a Small Molecule: En Route to Chemical Proteomics" describes a degradation agent that consists of two small molecules linked by PEG that in vivo degrades the by concurrently binding the androgen receptor and ubiquitin E3 ligase. WO 2013/170147 filed by Crews et al. titled "Compounds Useful for Promoting Protein Degradation and Methods of Using Same" describes compounds comprising a protein degradation moiety covalently bound to a linker, wherein the ClogP of the compound is equal to or higher than 1.5. In particular, the specification discloses protein degrading compounds that incorporate certain small molecules that can bind to an E3 ubiquitin ligase. In unrelated parallel research, scientists were investigating toxicity. Ito et al. {Science 2010, 327, 1345-1350) titled "Identification of a Primary Target of Thalidomide Teratogenicity", described that cereblon is a thalidomide binding protein. Cereblon forms part of an E3 ubiquitin ligase protein complex which interacts with damaged DNA binding protein 1, forming an E3 ubiquitin ligase complex with Cullin 4 and the E2-binding protein ROC1 (also known as RBX1) where it functions as a substrate receptor to select proteins for ubiquitination. The study revealed that thalidomide-cereblon binding in vivo may be responsible for thalidomide teratogenicity. After the discovery that thalidomide causes teratogenicity in the mid- 1960' s, the compound and related structures were notwithstanding found to be useful as anti-inflammatory, anti-angiogenic and anti-cancer agents (see Bartlett et al. {Nat. Rev. Cancer 2004, 4, 314-322) titled "The Evolution of Thalidomide and Its Imid Derivatives as Anticancer Agents"). The disclosure that thalidomide binds to the cereblon E3 ubiquitin ligase led to research to investigate incorporating thalidomide and certain derivatives into compounds for the targeted destruction of proteins. Two seminal papers were published in Science in 2014: G . Lu et al., The Myeloma Drug Promotes the Cereblon-Dependent Destruction of Ikaros Proteins, Science, 343, 305-309 (2014); and J . Kronke et al., Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells, Science, 343, 301-305 (2014). U.S. 2014/0356322 assigned to Yale University, GlaxoSmithKline, and Cambridge Enterprise Limited University of Cambridge titled "Compounds and Methods for the Enhanced Degradation of Target Proteins & Other Polypeptides by an E3 Ubiquitin Ligase" describes protein degrading compounds that bind to the VHL E3 Ubiquitin Ligase. See also Buckley et al. (J. Am. Chem. Soc. 2012, 134, 4465-4468) titled "Targeting the Von Hippel-Lindau E3 Ubiquitin Ligase Using Small Molecules to Disrupt the Vhl/Hif-1 alpha Interaction". Additional publications in this area include the following: Lu et al. {Chem. Biol. 2015, 22, 755-763) titled "Hijacking the E3 Ubiquitin Ligase Cereblon to Efficiently Target Brd4"; Bondeson et al. {Nat. Chem. Biol. 2015, 11, 6 11-617) titled "Catalytic in Vivo Protein Knockdown by Small-Molecule Protacs"; Gustafson et al. (Angewandte Chemie, International Edition in English 2015, 54, 9659-9662) titled "Small-Molecule-Mediated Degradation of the Androgen Receptor through Hydrophobic Tagging"; Lai et al. {Angewandte Chemie, International Edition in English 2016, 55, 807-810) titled "Modular Protac Design for the Degradation of Oncogenic Bcr-Abl"; Toure et al. (Angew. Chem. Int. Ed. 2016, 55, 1966-1973) titled "Small-Molecule Protacs: New Approaches to Protein Degradation"; and Winter et al. (Science 2015, 348, 1376- 1381) titled "Drug Development. Phthalimide Conjugation as a Strategy for in Vivo Target Protein Degradation" describes thalidomide based Target Protein degradation technology. WO 2015/160845 assigned to Arvinas Inc. titled "Imide Based Modulators of Proteolysis and Associated Methods of Use" describes protein degradation compounds that incorporate thalidomide and certain derivatives which bind to a cereblon E3 ligase. Additional patent applications filed by Arvinas Inc. directed towards the degradation of a Target Protein using known E3 ligase ligands to direct the Target Protein to the proteasome for degradation include U.S. 2016/0058872 titled "Imide Based Modulators of Proteolysis and Associated Methods of Use"; U.S. 2016/0045607 titled "Estrogen-related Receptor Alpha Based PROTAC Compounds and Associated Methods of Use"; U.S. 2016/0214972 titled "Compounds and Methods for the Targeted Degradation of Androgen Receptor"; U.S. 2016/0272639 titled " Compounds and Methods for the Enhanced Degradation of Target Proteins"; U.S. 2017/0008904 titled "MDM2- Based Modulators of Proteolysis and Associated Methods of Use"; U.S. 2017/0037004 titled "Alanine-Based Modulators of Proteolysis and Associated Methods of Use"; U.S. 2017/0065719 titled "Compounds and Methods for the Targeted Degradation of Bromodomain containing proteins"; WO 2016/036036 titled "Tank Binding Kinase-1 PROTACS and Associated Methods of Use"; and WO 2016/197032 "Imide-Based Modulators and Proteolysis and Associated Methods of Use". Dana-Farber Cancer Institute has also filed several patent applications directed towards the degradation of a Target Protein using known E3 ligase ligands to direct the Target Protein to the proteasome for degradation. These filings include US 2016/0176916 titled "Methods to Induce Target Protein Degradation through Bifunctional Molecules; WO 2017/024318 titled "Target Protein Degradation to Attenuate Adoptive T-Cell Therapy Associated Adverse Inflammatory Responses"; WO 2017/024317 titled "Methods to Induce Target Protein Degradation through Bifunctional Molecules"; and WO 2017/024319 titled "Tunable Endogenous Protein Degradation". While progress has been made in the area of modulation of the UPP for in vivo protein degradation, it would be useful to have additional compounds and approaches to more fully harness the UPP for therapeutic treatments. It is an object of the present invention to provide new compounds, methods, compositions, and methods of manufacture that are useful to degrade selected proteins in vivo.

SUMMARY

Compounds and their uses and manufacture are provided that cause degradation of a bromodomain containing protein via the ubiquitin proteasome pathway (UPP). The compounds of the present invention are referred to below as "Degronimers" because they accomplish degradation of a bromodomain containing protein by the proteasome. The Degronimers include a "Targeting Ligand" that binds to a bromodomain containing protein, a "Degron" which binds (typically non- covalently) to an E3 Ligase (typically the cereblon component) and optionally a linker that covalently links the Targeting Ligand to the Degron. Therefore, in some embodiments a method to treat a host with a disorder mediated by a bromodomain containing protein is provided that includes administering an effective amount of the Degronimer or its pharmaceutically acceptable salt to the host, typically a human. The Degronimer causes the degradation of the bromodomain containing protein in vivo. Degronimers have been discovered that have the ability to ubiquitinate Targeted Proteins by binding both E3 Ubiquitin Ligase (typically through cereblon) and a selected Targeted Protein. In one aspect of the present invention a Degronimer of Formula I, II, III, IV, V, VI, VII, VIII, IX, or X is rovided: L

0 0/Z 0 S l / d 9 0Z6 /Z 0 OAV or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, optionally in a pharmaceutically acceptable carrier to form a composition; wherein: A is independently selected from C(R ), and N wherein in certain embodiments the number of nitrogen atoms is 0, 1, 2, 3, or 4 per ring (as allowed by context) and is selected to produce a stable ring and a pharmaceutically acceptable Degronimer. When A's are in a six- membered ring (unfused or fused), the ring can be, in non-limiting embodiments as allowed by context, a pyridine, diazine, triazine, pyrimidine, pyridazine, pyrazine, triazine or tetrazine.

n is 0, 1, 2, 3, 4, 5, 6 or 7; is a single or double bond; X is independently selected from C(R ), N, N(R 9), N(O), O, and S as permitted by valency and by ring stability under ambient conditions (as appropriate in context); and wherein there is typically not more than 1, 2, or 3 heteroatoms. When X is in a five membered ring, non-limiting examples are pyrrole, furan, thiophene, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, triazole, furazan, oxadiazole, thiadiazole, diazole and . or X is independently selected from C(H), C(R ), N, N(R 9), N(O), O, and S as permitted by valency and by ring stability under ambient conditions; and wherein there is typically not more than 1, 2, or 3 heteroatoms in the X-bearing ring except that the ring may have 4 nitrogens to form a tetrazole; X' is selected from O, S, and -N(R9); 9 Y is selected from CH(R ), C(R )2, O, S, H, N(R ), P=0(aliphatic including alkyl), P=0(Oaliphatic, including alkyl), P=0(Oaryl), P=0(OH), P=0(NHaliphatic, including alkyl),

P=0(NHaryl or heteroaryl), P=0(NH 2), S(0) 2 and S(O); Q is either C or N; T is hydrogen, deuterium, fluorine, chlorine, bromine, iodine, or or T and R together with the carbon to which they are attached form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle; in an alternative embodiment T and R form a 3, 4, 5, or 6 membered spirocarbocycle or a

3, 4, 5, or 6 membered spiroheterocycle that is substituted with R , S0 2heterocycle, or SCtealkyl; W is selected from C(R ) and N; R2 and R3 are independently selected from hydrogen, deuterium, alkyl, hydroxyl, alkoxy, aliphaticoxy, amine, -NH(aliphatic including alkyl), -N(aliphatic including alkyl) 2, fluorine, chlorine, bromine, iodine, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroaliphatic, aliphatic, and carbocyclic; R , R2', R4, R4', R5, R , R6, R6', R7, R , R 3, and R 13' are independently selected from hydrogen, deuterium, alkyl, aliphatic, hydroxyl, alkoxy, aliphaticoxy, amine, cyano, -NH(aliphatic including alkyl), -N(aliphatic, including alkyl) 2, fluorine, chlorine, bromine, iodine, haloalkyl

(including -CFH 2, CHF2, and -CF3), alkenyl, alkynyl, aryl, heteroaryl, heterocyclo and carbocyclic;

or two R substituents can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 4 or two R substituents can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

5 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 6 6 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

7 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 3 or R and R can form C=0, C=S, C=CH2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

or two R4 substituents can form a 1, 2 , 3 or 4 carbon bridged ring; or R4 and R can form a 3, 4, 5, or 6 membered fused ring; or R and R6 can form a 3, 4, 5, or 6 membered fused ring;

or R4' and R can form a 1, 2, 3, or 4 carbon bridged ring;

or R4' and R6 can form a 1, 2, 3, or 4 carbon bridged ring; 8 9 9 R is N(R ), CH2, CHR , C(R )2, O, or S; R9 is selected at each instance from hydrogen, deuterium, alkyl, -C(0)H, -C(0)OH, - C(0)(aliphatic, including alkyl), -C(0)0(aliphatic, including alkyl), alkenyl, and alkynyl; aryl, heteroaryl, heterocyclic, aliphatic and heteroaliphatic; R 0 is Linker-Degron; R and R 11A are independently selected at each instance from hydrogen, alkyl, alkenyl, alkynyl, fluorine, chlorine, bromine, iodine, hydroxyl, alkoxy, cyano, azide, amino, -NH(aliphatic or alkyl), -N(aliphatic or alkyl)2, - H S0 2(aliphatic or alkyl, -N(aliphatic or alkyl)S0 2(aliphatic or alkyl), - H S0 2(aryl or heteroaryl), -N(aliphatic including alkyl)S0 2aryl, - H S0 2alkenyl, -

N(alkyl)S0 2alkenyl, -NHS0 2alkynyl, -N(alkyl)S0 2alkynyl, cyano, nitro, nitroso, -SH, -Salkyl, -

S(0)alkyl, -S(0) 2alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, aliphatic, heteroaliphatic, aryl, heteroaryl, carbocyclic, and haloalkyl; Linker is a chemical group that attaches the Degron to a Targeting Ligand; and Degron is a chemical group that interacts with an E3 ligase to cause ubiquitination of nearby protein(s), and in particular, the Target Protein. In one embodiment when the term alkyl is used in the formula above in some embodiments it may independently refer to a Ci-C 6, C1-C5, C1-C4, C1-C3 or C1-C2 moiety. In one embodiment both R and T are hydrogen or deuterium. In another aspect of the present invention a Degronimer of Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, or Formula XVI is provided: or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, optionally in a pharmaceutically acceptable carrier to form a composition; wherein:

=-=-= is a single or double bond;

Q is either C or N; T is hydrogen, deuterium, fluorine, chlorine, bromine, iodine, or or T and R together with the carbon to which they are attached form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle; in an alternative embodiment T and R form a 3, 4, 5, or 6 membered spirocarbocycle or a

3, 4, 5, or 6 membered spiroheterocycle that is substituted with R , S0 2heterocycle, or SChalkyl; W is selected from C(R ) and N; 9 W2 is selected from C(R )2 and N(R ); R is independently selected from hydrogen, deuterium, alkyl, hydroxyl, alkoxy, amine, -

NH(aliphatic or alkyl), -N(aliphatic or alkyl)2, fluorine, chlorine, bromine, iodine, haloalkyl

(including -CFH 2, CHF2, and -CF3), alkenyl, alkynyl, aryl, heteroaryl, heterocyclo, aliphatic, heteroaliphatic,

or two R substituents can form C=0, C=S, C=CH2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; and the other variables are as described above. In another aspect of the present invention a Degronimer of Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX is provided:

(XVIII)

(XXVIII),

or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, optionally in a pharmaceutically acceptable carrier to form a composition; wherein: X 11 is bond, CH2 or CH(aliphatic including alkyl), and the other variables are as defined above. The Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, is bifunctional with E3 Ubiquitin Ligase targeting moieties (Degrons) linked to Targeting Ligands, which function to recruit Targeted Proteins to E3 Ubiquitin Ligase for degradation. One non-limiting example of a disorder treatable by such Degronimers is abnormal cellular proliferation, such as a tumor or cancer, wherein the Targeted Protein is an oncogenic protein or a signaling mediator of an abnormal cellular proliferative pathway and its degradation decreases abnormal cell growth. Based on this discovery, Degronimers and methods are presented for the treatment of a patient with a disorder mediated by a protein that is targeted for selective degradation that includes administering an effective amount of one or a combination of the Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, described herein to a patient (typically a human) in need thereof, optionally in a pharmaceutically acceptable carrier. In certain embodiments the disorder is selected from a benign growth, neoplasm, tumor, cancer, immune disorder, autoimmune disorder, inflammatory disorder, graft-versus-host rejection, viral infection, bacterial infection, an amyloid- based proteinopathy, a proteinopathy, or fibrotic disorder. In a typical embodiment the patient is a human. In certain embodiments, the present invention provides the administration of an effective amount of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, to treat a patient, for example, a human, having an infectious disease, wherein the therapy targets a Target Protein of the infectious agent or the host optionally in combination with another bioactive agent. The disease state or condition may be caused by a microbial agent or other exogenous agent such as a virus (as non- limiting examples, HIV, HBV, HCV, HSV, HPV, RSV, CMV, Ebola, Flavivirus, Pestivirus, Rotavirus, Influenza, Coronavirus, EBV, viral pneumonia, drug-resistant viruses, Bird flu, RNA virus, DNA virus, adenovirus, poxvirus, Picornavirus, Togavirus, Orthomyxovirus, Retrovirus or Hepadnovirus), bacteria (including but not limited to Gram-negative, Gram-positive, Atypical, Staphylococcus, Streptococcus, E . Coli, Salmonella, Helicobacter pylori, meningitis, gonorrhea, Chlamydiaceae, Mycoplasmataceae, etc), fungus, protozoa, helminth, worms, prion, parasite, or other microbe. The structure of the Degronimer is typically selected such that it is sufficiently stable to sustain a shelf life of at least two, three, four, or five months under ambient conditions. To accomplish this, each of the R groups described herein must be sufficiently stable to sustain the corresponding desired shelf life of at least two, three, four or five months under ambient conditions. One of ordinary skill in the art is well aware of the stability of chemical moieties and can avoid those that are not stable or are too reactive under the appropriate conditions. The Degronimer (Degron, Linker and Targeting Ligand), including any of the "R" groups defined herein, and variables where appropriate, may be optionally substituted as described below in Section I . Definitions, if desired to achieve the target effect, results in a stable R moiety and final Degronimer that makes chemical sense to the routineer, and if the final Degronimer for therapy is pharmaceutically acceptable. Also, all R groups, with or without optional substituents, should be interpreted in a manner that does not include redundancy (i.e., as known in the art, alkyl substituted with alkyl is redundant; however for examples, alkoxy substituted with alkoxy is not redundant). In one embodiment, the present invention provides Degron moieties which are covalently linked to a Targeting Ligand through a Linker which can be of varying length and functionality, as described in more detail below. In one embodiment, the Degron moiety is linked directly to the Targeting Ligand (i.e., the Linker is a bond). In certain embodiments, the Linker can be any chemically stable group that attaches the Degron to the Targeting Ligand. In a typical embodiment the Linker has a chain of 2 to 14, 15, 16, 17, 18 or 20 or more carbon atoms of which one or more carbons can be replaced by a heteroatom such as O, N, S, P, as long as the resulting molecule has a stable shelf life for at least 2 months, 3 months, 6 months or 1 year as part of a pharmaceutically acceptable dosage form, and itself is pharmaceutically acceptable. In certain embodiments the chain has 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 contiguous atoms in the chain. For example, the chain may include 1 or more ethylene glycol units, and in some embodiments, may have at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more contiguous, partially contiguous or non-contiguous ethylene glycol units in the Linker. In certain embodiments the chain has at least 1, 2, 3, 4, 5, 6, 7, or 8 branches which can be independently alkyl, heteroalkyl, aryl, heteroaryl, alkenyl, or alkynyl substituents, which in one embodiment, each branch has 10, 8, 6, 4, 3, 2 carbons or one carbon. In one embodiment, the Target Protein is a protein that is not druggable in the classic sense in that it does not have a binding pocket or an active site that can be inhibited or otherwise bound, and cannot be easily allosterically controlled. In another embodiment, the Targeted Protein is a protein that is druggable in the classic sense. Examples of Targeted Proteins are provided below. Degronimers of the present invention offer important clinical benefits to patients, in particular for the treatment of the disease states and conditions modulated by the selected Target Proteins that are bound to the Targeting Ligand. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this application belongs. In the specification, the singular forms also include the plural unless the context clearly dictates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present application, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting. Other features and advantages of the present application will be apparent from the following detailed description and claims. The present invention thus includes at least the following features: (a) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein, or a pharmaceutically acceptable salt, isotopic derivative (including a deuterated derivative) or prodrug thereof; (b) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, for the treatment of a disorder that is mediated by a Targeted Protein, wherein the Degronimer includes a Targeting Ligand for the Targeted Protein, and wherein the Degron is optionally linked to the Targeting Ligand through a Linker; (c) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, or a pharmaceutically acceptable salt thereof in an effective amount in the treatment of a patient, including a human, with a disorder mediated by a Targeted Protein, including any of the disorders described herein, for example, abnormal cellular proliferation such as a tumor or cancer, an immune disorder, an autoimmune disorder or inflammatory disorder, a cardiovascular disorder, an infectious disease, or other disorder that responds to such treatment; (d) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, or a pharmaceutically acceptable salt thereof, isotopic derivative, and prodrug thereof in the manufacture of a medicament for the treatment of a medical disorder as described herein; (e) A method for manufacturing a medicament intended for the therapeutic treatment of a disorder characterized in that a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein is used in the manufacture; (f A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein, and pharmaceutically acceptable salts and prodrugs thereof that are useful in the treatment of an abnormal cellular proliferation such as cancer, including any of the cancers described herein or a tumor; (g) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for the treatment of an abnormal cellular proliferation such as cancer, including any of the cancers described herein or a tumor; (h) A method for manufacturing a medicament intended for the therapeutic use of treating an abnormal cellular proliferation such as cancer, including any of the cancers described herein, or a tumor characterized in that a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein is used in the manufacture; (i) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein, and pharmaceutically acceptable salts, isotopic derivatives and prodrugs thereof that are useful in the treatment of a tumor, including any of the tumors described herein; (j) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for the treatment of a tumor, including any of the tumors described herein; (k) A method for manufacturing a medicament intended for the therapeutic use of treating a tumor, including any of the tumors described herein, characterized in that a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein is used in the manufacture;

(1) Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein, and pharmaceutically acceptable salts and prodrugs thereof that are useful in the treatment of an immune, autoimmune or inflammatory disorder; (m) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for the treatment of an immune, autoimmune or inflammatory disorder; (n) A method for manufacturing a medicament intended for the therapeutic use of treating an immune, autoimmune or inflammatory disorder, characterized in that a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein is used in the manufacture; (o) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein, and pharmaceutically acceptable salts and prodrugs thereof that are useful in the treatment of an infection by a pathogen, as described futher herein, including a viral infection, such as HIV, HBV, HCV and RSV, HPV, HSV, etc; (p) Use of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for the treatment of a an infection by a pathogen, as described further herein, including a viral infection, including but not limited to HIV, HBV, HCV, RSV, HPV, HSV, etc.; (q) A method for manufacturing a medicament intended for the therapeutic use of treating a viral infection including but not limited to HIV, HBV, HCV and RSV, characterized in that a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein is used in the manufacture; (r) A pharmaceutical formulation comprising an effective host-treating amount of the Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, or a pharmaceutically acceptable salt or prodrug thereof together with a pharmaceutically acceptable carrier or diluent; (s) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein as a mixture of enantiomers or diastereomers (as relevant), including as a racemate; (t) A Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein in enantiomerically or diastereomerically (as relevant) enriched form, including as an isolated enantiomer or diastereomer (i.e., greater than 85, 90, 95, 97 or 99% pure); and, (u) A process for the preparation of therapeutic products that contain an effective amount of a Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as described herein.

BRIEF DESCRIPTION OF THE FIGURES FIG. 1A presents examples of BRD1 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 5AME ("the Crystal Structure of the Bromodomain of Human Surface Epitope Engineered Brdl A in Complex with 3D Consortium Fragment 4-Acetyl-Piperazin-2-One Pearce", N.M. et al.); the crystal structure PDB 5AMF ("Crystal Structure of the Bromodomain of Human Surface Epitope Engineered BrdlA in Complex with 3D Consortium Fragment Ethyl 4 5 6 7-Tetrahydro-lH- Indazole-5-Carboxylate", Pearce N.M. et al.); the crystal structure PDB 5FG6 ("the Crystal structure of the bromodomain of human BRD1 (BRPF2) in complex with OF-1 chemical probe.", TallantC. et al.); Filippakopoulos P. etal. "Histone recognition and large-scale structural analysis of the human bromodomain family." Cell, 149: 214-231 (2012). FIG. IB-ID present examples of BRD2 Bromodomain 1 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 2ydw; the crystal structure PDB 2yek; the crystal structure PDB 4a9h; the crystal structure PDB 4a9f; the crystal structure PDB 4a9i; the crystal structure PDB 4a9m; the crystal structure PDB 4akn; the crystal structure PDB 4alg, and the crystal structure PDB 4uyf. FIG. 1E-1G present examples of BRD2 Bromodomain 2 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 3oni; Filippakopoulos P. et al. "Selective Inhibition of BET Bromodomains." Nature 468: 1067-1073 (2010); the crystal structure PDB 4j lp; McLure K.G. et al. "RVX-208: an Inducer of ApoA-I in Humans is a BET Bromodomain Antagonist." Plos One

8 : e83190-e83190 (2013); Baud M.G. et al. "Chemical biology. A bump-and-hole approach to engineer controlled selectivity of BET bromodomain chemical probes" Science 346: 638-641 (2014); Baud M.G. et al. "New Synthetic Routes to Triazolo-benzodiazepine Analogues: Expanding the Scope of the Bump-and-Hole Approach for Selective Bromo and Extra-Terminal (BET) Bromodomain Inhibition" J. Med. Chem. 59: 1492-1500 (2016); Gosmini R . et al. "The Discovery of I-Bet726 (Gskl324726A) a Potent Tetrahydroquinoline Apoal Up-Regulator and Selective Bet Bromodomain Inhibitor" J. Med. Chem. 57: 8 111 (2014); the crystal structure PDB 5EK9 ("Crystal structure of the second bromodomain of human BRD2 in complex with a hydroquinolinone inhibitor", Tallant C . et al); the crystal structure PDB 5BT5; the crystal structure PDB 5dfd; Baud M.G. et al. "New Synthetic Routes to Triazolo-benzodiazepine Analogues: Expanding the Scope of the Bump-and-Hole Approach for Selective Bromo and Extra- Terminal (BET) Bromodomain Inhibition" J. Med. Chem. 59: 1492-1500 (2016). FIG. 1H presents examples of BRD4 Bromodomain 1 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 5WUU and the crystal structure PDB 5F5Z. FIG. ll-lj present examples of BRD4 Bromodomain 2 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, Chung C.W. et al. "Discovery and Characterization of Small Molecule Inhibitors of the Bet Family Bromodomains" J. Med. Chem. 54: 3827 (201 1) and Ran X . et al. "Structure-Based Design of gamma-Carboline Analogues as Potent and Specific BET Bromodomain Inhibitors" J.Med. Chem. 58: 4927-4939 (2015). FIG. IK presents examples of BRDT Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 4flp and the crystal structure PDB 4kcx. FIG. lL-lO present examples of BRD9 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structure PDB 4nqn; the crystal structure PDB 4uit; the crystal structure PDB 4uiu; the crystal structure PDB 4uiv; the crystal structure PDB 4z6h; the crystal structure PDB 4z6i; the crystal structure PDB 5e9v; the crystal structure PDB 5eul; the crystal structure PDB 5flh; and, the crystal structure PDB 5fp2. FIG. 1P-1V present examples of BET Bromodomain-Containing Protein Targeting Ligands wherein R is the point at which the Linker is attached. FIG. 1W-1X present examples of the BRD4 Bromodomains of PCAF and GCN5 receptors 1 Targeting Ligands wherein R is the point at which the Linker is attached. For additional examples and related ligands, see, the PDB crystal structure 5tpx ("Discovery of a PCAF Bromodomain Chemical Probe"); Moustakim, M., et al. Angew. Chem. Int. Ed. Engl. 56: 827 (2017); the PDB crystal structure 5mlj ("Discovery of a Potent, Cell Penetrant, and Selective p300/CBP-Associated Factor (PCAF)/General Control Nonderepressible 5 (GCN5) Bromodomain Chemical Probe"); and, Humphreys, P. G . et al. J. Med. Chem. 60: 695 (2017). FIG. 1Y-1RR present examples of BRD4 Bromodomain 1 Targeting Ligands wherein R or linker- TL is the point at which the Linker is attached. For additional examples and related ligands, see, the crystal structures PDB 3u5k and 3u5 1 and related ligands in Filippakopoulos, P. et al. "Benzodiazepines and benzodiazepines as protein interaction inhibitors targeting bromodomains of the BET family", Bioorg. Med. Chem. 20: 1878-1886 (2012); the crystal structure PDB 3u51; the crystal structure PDB 3zyu and related ligands described in Dawson, M .A . et al. "Inhibition of Bet Recruitment to Chromatin as an Effective Treatment for Mil-Fusion

Leukaemia." Nature 478: 529 (201 1); the crystal structure PDB 4bwl and related ligands described in Mirguet, O . et al. "Naphthyridines as Novel Bet Family Bromodomain Inhibitors."

Chemmedchem 9 : 589 (2014); the crystal structure PDB 4cfl and related ligands described in Dittmann, A . et al. "The Commonly Used Pi3-Kinase Probe Ly294002 is an Inhibitor of Bet

Bromodomains" ACS Chem. Biol. 9 : 495 (2014); the crystal structure PDB 4e96 and related ligands described in Fish, P.V. et al. "Identification of a chemical probe for bromo and extra C- terminal bromodomain inhibition through optimization of a fragment-derived hit." J. Med. Chem. 55: 9831-9837 (2012); the crystal structure PDB 4clb and related ligands described in Atkinson, S.J. et al. "The Structure Based Design of Dual Hdac/Bet Inhibitors as Novel Epigenetic Probes."

Medchemcomm 5 : 342 (2014); the crystal structure PDB 4f3i and related ligands described in Zhang, G . et al. "Down-regulation of NF-{kappa}B Transcriptional Activity in HIV-associated Kidney Disease by BRD4 Inhibition." J. Biol. Chem. 287: 28840-28851 (2012); the crystal structure PDB 4hxl and related ligands described in Zhao, L . "Fragment-Based Drug Discovery of 2-Thiazolidinones as Inhibitors of the Histone Reader BRD4 Bromodomain." J. Med. Chem. 56: 3833-3851 (2013); the crystal structure PDB 4hxs and related ligands described in Zhao, L . et al. "Fragment-Based Drug Discovery of 2-Thiazolidinones as Inhibitors of the Histone Reader BRD4 Bromodomain." J. Med. Chem. 56: 3833-3851 (2013); the crystal structure PDB 41rg and related ligands described in Gehling, V.S. et al. "Discovery, Design, and Optimization of Isoxazole Azepine BET Inhibitors." ACS Med Chem Lett 4 : 835-840 (2013); the crystal structure PDB 4mep and related ligands described in Vidler, L.R. "Discovery of Novel Small-Molecule Inhibitors of BRD4 Using Structure-Based Virtual Screening." et al. J. Med. Chem. 56: 8073-8088 (2013); the crystal structures PDB 4nr8 and PDB 4c77 and related ligands described in Ember, S.W. et al. "Acetyl-lysine Binding Site of Bromodomain-Containing Protein 4 (BRD4) Interacts with Diverse Kinase Inhibitors". ACS Chem.Biol. 9 : 1160-1 171 (2014); the crystal structure PDB 4o7a and related ligands described in Ember, S.W. et al. "Acetyl-lysine Binding Site of Bromodomain- Containing Protein 4 (BRD4) Interacts with Diverse Kinase Inhibitors." ACS Chem. Biol. 9 : 1160- 1171 (2014); the crystal structure PDB 407b and related ligands described in "Acetyl-lysine Binding Site of Bromodomain-Containing Protein 4 (BRD4) Interacts with Diverse Kinase

Inhibitors." Ember, S.W. et al. (2014) ACS Chem. Biol. 9 : 1160-1 171; the crystal structure PDB 4o7c and related ligands described in Ember, S.W. et al. "Acetyl-lysine Binding Site of Bromodomain-Containing Protein 4 (BRD4) Interacts with Diverse Kinase Inhibitors". ACS Chem. Biol. 9 : 1160-1 171 (2014); the crystal structure PDB 4gpj; the crystal structure PDB 4uix and related ligands described in Theodoulou, N.H. et al. "The Discovery of I-Brd9, a Selective Cell Active Chemical Probe for Bromodomain Containing Protein 9 Inhibition". J. Med. Chem. 59: 1425 (2016); the crystal structure PDB 4uiz and related ligands described in Theodoulou, N.H., et al. "The Discovery of I-Brd9, a Selective Cell Active Chemical Probe for Bromodomain Containing Protein 9 Inhibition". J. Med. Chem. 59: 1425 (2016); the crystal structure PDB 4wiv and related ligands described in McKeown, M.R. et al. "Biased multicomponent reactions to develop novel bromodomain inhibitors." J. Med. Chem. 57: 9019-9027 (2014); the crystal structure PDB 4x2i and related ligands described in Taylor, A.M. et al. "Discovery of Benzotriazolo[4,3-d][l,4]diazepines as Orally Active Inhibitors of BET Bromodomains." ACS Med. Chem. Lett. 7 : 145-150 (2016); the crystal structure PDB 4yh3; And related ligands described in Duffy, B.C. "Discovery of a new chemical series of BRD4(1) inhibitors using protein-ligand docking and structure-guided design." Bioorg. Med. Chem. Lett. 25: 2818-2823 (2015); the crystal structure PDB 4yh4 and related ligands described in Duffy, B.C. "Discovery of a new chemical series of BRD4(1) inhibitors using protein-ligand docking and structure-guided design." Bioorg. Med. Chem. Lett. 25: 2818-2823 (2015); the crystal structure PDB 4zlq and related ligands described in Taylor, A.M. "Discovery of Benzotriazolo[4,3- d][l,4]diazepines as Orally Active Inhibitors of BET Bromodomains." ACS Med. Chem. Lett. 7 : 145-150 (2016); the crystal structure PDB 4zwl; the crystal structure PDB 5a5s and related ligands described in Demont, E.H. "Fragment-Based Discovery of Low-Mi cromolar Atad2 Bromodomain Inhibitors. J. Med. Chem. 58: 5649 (2015); the crystal structure PDB 5a85 and related ligands described in Bamborough, P. "Structure-Based Optimization of Naphthyridones Into Potent Atad2 Bromodomain Inhibitors" J. Med. Chem. 58: 6151 (2015); the crystal structure PDB 5acy and related ligands described in Sullivan, J.M. "Autism-Like Syndrome is Induced by Pharmacological Suppression of Bet Proteins in Young Mice." J. Exp. Med. 212: 1771 (2015); the crystal structure PDB 5ad2 and related ligands described in Waring, M.J. et al. "Potent and Selective Bivalent Inhibitors of Bet Bromodomains". Nat. Chem. Biol. 12: 1097 (2016); the crystal structure PDB 5cfw and related ligands described in Chekler, E.L. et al. "Transcriptional Profiling of a Selective CREB Binding Protein Bromodomain Inhibitor Highlights Therapeutic Opportunities." Chem. Biol. 22: 1588-1596 (2015); the crystal structure PDB 5cqt and related ligands described in Xue, X . et al. "Discovery of Benzo[cd]indol-2(lH)-ones as Potent and Specific BET Bromodomain Inhibitors: Structure-Based Virtual Screening, Optimization, and Biological Evaluation". J. Med. Chem. 59: 1565-1579 (2016); the crystal structure PDB 5d3r and related ligands described in Hugle, M . et al. "4-Acyl Pyrrole Derivatives Yield Novel Vectors for Designing Inhibitors of the Acetyl-Lysine Recognition Site of BRD4(1)". J. Med. Chem. 59: 1518-1530 (2016); the crystal structure PDB 5dlx and related ligands described in Milhas, S. et al. "Protein-Protein Interaction Inhibition (2P2I)-Oriented Chemical Library Accelerates Hit Discovery." (2016) ACS Chem.Biol. 11 : 2140-2148; the crystal structure PDB 5dlz and related ligands described in Milhas, S. et al. "Protein-Protein Interaction Inhibition (2P2I)-Oriented Chemical Library Accelerates Hit

Discovery." ACS Chem. Biol. 11 : 2140-2148 (2016); the crystal structure PDB 5dw2 and related ligands described in Kharenko, O.A. et al. "RVX-297- a novel BD2 selective inhibitor of BET bromodomains." Biochem. Biophys. Res. Commun. 477: 62-67 (2016); the crystal structure PDB 5dlx; the crystal structure PDB 5his and related ligands described in Albrecht, B.K. et al. "Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra- Terminal (BET) Family as a Candidate for Human Clinical Trials." J. Med. Chem. 59: 1330-1339 (2016); the crystal structure PDB 5ku3 and related ligands described in Crawford, T.D. et al. "Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of

CBP/EP300". J. Med. Chem. 59: 10549-10563 (2016); the crystal structure PDB 51j2 and related ligands described in Bamborough, P. et al. "A Chemical Probe for the ATAD2 Bromodomain." Angew. Chem. Int. Ed. Engl. 55: 11382-1 1386 (2016); the crystal structure PDB 5dlx and related ligands described in Wang, L . "Fragment-based, structure-enabled discovery of novel pyridones and pyridone macrocycles as potent bromodomain and extra-terminal domain (BET) family bromodomain inhibitors". J. Med. Chem. 10.1021/acs.jmedchem.7b00017 (2017); WO 2015169962 A l titled "Benzimidazole derivatives as BRD4 inhibitors and their preparation and use for the treatment of cancer" assigned to Boehringer Ingelheim International GmbH, Germany; and, WO 201 1143669 A2 titled "Azolodiazepine derivatives and their preparation, compositions and methods for treating neoplasia, inflammatory disease and other disorders" assigned to Dana-Farber Cancer Institute, Inc, USA. FIG. ISS is a dendrogram of the human bromodomain family of proteins organized into eightsubfamilies, which are involved in epigenetic signaling and chromatin biology. Any of the proteins of the bromodomain family in FIG. ISS can be selected as a Target Protein according to the present invention. DETAILED DESCRIPTION

I. DEFINITIONS Compounds are described using standard nomenclature. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. The compounds in any of the Formulas described herein may be in the form of a racemate, enantiomer, mixture of enantiomers, diastereomer, mixture of diastereomers, tautomer, N-oxide, isomer; such as rotamer, as if each is specifically described unless specifically excluded by context. The terms "a" and "an" do not denote a limitation of quantity, but rather denote the presence of at least one of the referenced item. The term "or" means "and/or". Recitation of ranges of values are merely 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 endpoints of all ranges are included within the range and independently combinable. All methods described herein can be performed in a suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of examples, or exemplary language (e.g., "such as"), is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. The present invention includes Degronimers of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, with at least one desired isotopic substitution of an atom, at an amount above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons. Examples of isotopes that can be incorporated into Degronimers of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine and iodine such as 2H, H, C, C, 4C, N, F P, 2P, S, 6C1, and 2 I respectively. In one non-limiting embodiment, isotopically labelled compounds can be used in metabolic studies (with, for example 4C), reaction kinetic studies (with, for example H or H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an F labeled compound may be particularly desirable for PET or SPECT studies. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. Isotopic substitutions, for example deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted with deuterium. In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In one non-limiting embodiment, deuterium is 90, 95 or 99% enriched at a desired location. In one non-limiting embodiment, the substitution of a hydrogen atom for a deuterium atom can be provided in any Degronimer of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX,. Even though reference is given to deuterium at certain locations on the Degronimers of the present invention, this is not a teaching away from positioning deuterium at any other location on the Degronimer, all of which are considered expecitly exemplified. In one non-limiting embodiment the substitution of a hydrogen atom for a deuterium atom can independently occur in one or more groups selected from any of the R groups or variables described herein. For example, when any of the groups are, or contain for example through substitution, methyl, ethyl, or methoxy, the alkyl residue may be deuterated (in non-limiting embodiments, CDH2, CD2H, CD3, CH2CD3, CD2CD3, CHDCHzD,

CH2CD3, CHDCHD2, OCDH2, OCD2H, or OCD3 etc.). In certain other embodiments, when two substituents are combined to form a cycle the unsubstituted carbons may be deuterated. The compound of the present invention may form a solvate with a solvent (including water). Therefore, in one non-limiting embodiment, the invention includes a solvated form of the compound. The term "solvate" refers to a molecular complex of a compound of the present invention (including a salt thereof) with one or more solvent molecules. Non-limiting examples of solvents are water, ethanol, isopropanol, dimethyl sulfoxide, acetone and other common organic solvents. The term "hydrate" refers to a molecular complex comprising a compound of the invention and water. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent may be isotopically substituted, e.g. D2O, d6-acetone, d6-DMSO. A solvate can be in a liquid or solid form. A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -(C=0)NH2 is attached through carbon of the carbonyl (C=0) group. "Alkyl" is a branched or straight chain saturated aliphatic hydrocarbon group. In one non- limiting embodiment, the alkyl group contains from 1 to about 12 carbon atoms, more generally from 1 to about 6 carbon atoms or from 1 to about 4 carbon atoms. In one non-limiting embodiment, the alkyl contains from 1 to about 8 carbon atoms. In certain embodiments, the alkyl is C1-C2, C1-C3, C1-C4, C1-C5, or Ci-C 6. The specified ranges as used herein indicate an alkyl group having each member of the range described as an independent species. For example, the term Ci-

C6 alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species and therefore each subset is considered separately disclosed. For example, the term C1-C4 alkyl as used herein indicates a straight or branched alkyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species. Examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t- butyl, n-pentyl, isopentyl, t rt-pentyl, neopentyl, n-hexyl, 2-methylpentane, 3-methylpentane, 2,2- dimethylbutane, and 2,3-dimethylbutane. In an alternative embodiment, the alkyl group is optionally substituted. The term "alkyl" also encompasses cycloalkyl or carbocyclic groups. For example, when a term is used that includes "alk" then "cycloalkyl" or "carbocyclic" can be considered part of the definition, unless unambiguously excluded by the context. For example and without limitation, the terms alkyl, alkoxy, haloalkyl, etc. can all be considered to include the cyclic forms of alkyl, unless unambiguously excluded by context. "Alkenyl" is a linear or branched aliphatic hydrocarbon groups having one or more carbon- carbon double bonds that may occur at a stable point along the chain. The specified ranges as used herein indicate an alkenyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkenyl radicals include, but are not limited to ethenyl, propenyl, allyl, propenyl, butenyl and 4-methylbutenyl. The term "alkenyl" also embodies "cis" and "trans" alkenyl geometry, or alternatively, "E" and "Z" alkenyl geometry. In an alternative embodiment, the alkenyl group is optionally substituted. The term "Alkenyl" also encompasses cycloalkyl or carbocyclic groups possessing at least one point of unsaturation. "Alkynyl" is a branched or straight chain aliphatic hydrocarbon group having one or more carbon-carbon triple bonds that may occur at any stable point along the chain. The specified ranges as used herein indicate an alkynyl group having each member of the range described as an independent species, as described above for the alkyl moiety. Examples of alkynyl include, but are not limited to, ethynyl, propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3- pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl and 5-hexynyl. In an alternative embodiment, the alkynyl group is optionally substituted. The term "Alkynyl" also encompasses cycloalkyl or carbocyclic groups possessing at least one triple bond.

"Alkylene" is a bivalent saturated hydrocarbon. Alkylenes, for example, can be a 1, 2, 3, 4, 5, 6, 7 to 8 carbon moiety, 1 to 6 carbon moiety, or an indicated number of carbon atoms, for example Ci-C2alkylene, Ci-C3alkylene, Ci-C4alkylene, Ci-Csalkylene, or Ci-C6alkylene. "Alkenylene" is a bivalent hydrocarbon having at least one carbon-carbon double bond. Alkenylenes, for example, can be a 2 to 8 carbon moiety, 2 to 6 carbon moiety, or an indicated number of carbon atoms, for example C2-C4alkenylene. "Alkynylene" is a bivalent hydrocarbon having at least one carbon-carbon triple bond. Alkynylenes, for example, can be a 2 to 8 carbon moiety, 2 to 6 carbon moiety, or an indicated number of carbon atoms, for example C2-C4alkynylene. "Halo" and "Halogen" refers to fluorine, chlorine, bromine or iodine. "Haloalkyl" is a branched or straight-chain alkyl groupssubstituted with 1 or more halo atoms described above, up to the maximum allowable number of halogen atoms. Examples of haloalkyl groups include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl. "Perhaloalkyl" means an alkyl group having all hydrogen atoms replaced with halogen atoms. Examples include but are not limited to, trifluoromethyl and pentafluoroethyl. "Chain" indicates a linear chain to which all other chains, long or short or both, may be regarded as being pendant. Where two or more chains could equally be considered to be the main chain, "chain" refers to the one which leads to the simplest representation of the molecule. "Haloalkoxy" indicates a haloalkyl group as defined herein attached through an oxygen bridge (oxygen of an alcohol radical). "Heterocycloalkyl" is an alkyl group as defined herein substituted with a heterocyclo group as defined herein. "Arylalkyl" is an alkyl group as defined herein substituted with an aryl group as defined herein. "Heteroarylalkyl" is an alkyl group as defined herein substituted with a heteroaryl group as defined herein. As used herein, "aryl" refers to a radical of a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n+2 aromatic ring system (e.g., having 6, 10, or 14 π electrons shared in a cyclic array) having 6-14 ring carbon atoms and zero heteroatoms provided in the aromatic ring system ("C6-i4 aryl"). In some embodiments, an aryl group has 6 ring carbon atoms ("C6 aryl"; e.g., phenyl). In some embodiments, an aryl group has 10 ring carbon atoms ("Cio aryl"; e.g., naphthyl such as 1- naphthyl and 2-naphthyl). In some embodiments, an aryl group has 14 ring carbon atoms ("Ci4 aryl"; e.g., anthracyl). "Aryl" also includes ring systems wherein the aryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the radical or point of attachment is on the aryl ring, and in such instances, the number of carbon atoms continue to designate the number of carbon atoms in the aryl ring system. The one or more fused carbocyclyl or heterocyclyl groups can be 4 to 7 or 5 to 7-membered saturated or partially unsaturated carbocyclyl or heterocyclyl groups that optionally contain 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, phosphorus, , silicon and boron, to form, for example, a 3,4- methylenedioxyphenyl group. In one non-limiting embodiment, aryl groups are pendant. An example of a pendant ring is a phenyl group substituted with a phenyl group. In an alternative embodiment, the aryl group is optionally substituted as described above. In certain embodiments, the aryl group is an unsubstituted C6-i4 aryl. In certain embodiments, the aryl group is a substituted

C6-i4 aryl. An aryl group may be optionally substituted with one or more functional groups that include but are not limited to, halo, hydroxy, nitro, amino, cyano, haloalkyl, aryl, heteroaryl, and heterocyclo. The term "heterocyclyl" (or "heterocyclo") includes saturated, and partially saturated heteroatom-containing ring radicals, where the heteroatoms may be selected from nitrogen, sulfur and oxygen. Heterocyclic rings comprise monocyclic 3-8 membered rings, as well as 5-16 membered bicyclic ring systems (which can include bridged fused and spiro-fused bicyclic ring systems). It does not include rings containing -O-O-.-O-S- or -S-S- portions. Said "heterocyclyl" group may be optionally substituted, for example, with 1, 2, 3, 4 or more substituents that include but are not limited to, hydroxyl, Boc, halo, haloalkyl, cyano, alkyl, aralkyl, oxo, alkoxy, and amino. Examples of saturated heterocyclo groups include saturated 3- to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms [e.g. pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, piperazinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms [e.g. morpholinyl]; saturated 3 to 6-membered heteromonocyclic group containing 1to 2 sulfur atoms and 1to 3 nitrogen atoms [e.g., thiazolidinyl]. Examples of partially saturated heterocyclyl radicals include but are not limited to, dihydrothienyl, dihydropyranyl, dihydrofuryl, and dihydrothiazolyl. Examples of partially saturated and saturated heterocyclo groups include but are not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, pyrrolinyl, pyrazolidinyl, piperazinyl, morpholinyl, tetrahydropyranyl, thiazolidinyl, dihydrothienyl, 2,3- dihydro-benzo[l,4]dioxanyl, indolinyl, isoindolinyl, dihydrobenzothienyl, dihydrobenzofuryl, isochromanyl, chromanyl, 1,2-dihydroquinolyl, 1,2,3,4- tetrahydro-isoquinolyl, 1 ,2,3,4- tetrahydro-quinolyl, 2,3,4,4a,9,9a-hexahydro-lH-3-aza-fluorenyl, 5,6,7- trihydro-1,2,4- triazolo[3,4-a]isoquinolyl, 3,4-dihydro-2H-benzo[l,4]oxazinyl, benzo[l,4]dioxanyl, 2,3- dihydro- lH-lX'-benzo[d]isothiazol-6-yl, dihydropyranyl, dihydrofuryl and dihydrothiazolyl. Heterocyclo groups also include radicals where heterocyclic radicals are fused/condensed with aryl or heteroaryl radicals: such as unsaturated condensed heterocyclic group containing 1 to 5 nitrogen atoms, for example, indoline, isoindoline, unsaturated condensed heterocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, unsaturated condensed heterocyclic group containing 1to 2 sulfur atoms and 1to 3 nitrogen atoms, and saturated, partially unsaturated and unsaturated condensed heterocyclic group containing 1 to 2 oxygen or sulfur atoms. The term "heteroaryl" denotes aryl ring systems that contain one or more heteroatoms selected from O, N and S, wherein the ring nitrogen and sulfur atom(s) are optionally oxidized, and nitrogen atom(s) are optionally quarternized. Examples include but are not limited to, unsaturated 5 to 6 membered heteromonocyclyl groups containing 1 to 4 nitrogen atoms, such as pyrrolyl, imidazolyl, pyrazolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl [e.g., 4H-l,2,4-triazolyl, IH-1 ,2,3-triazolyl, 2H-l,2,3-triazolyl]; unsaturated 5- to 6- membered heteromonocyclic groups containing an oxygen atom, for example, pyranyl, 2-furyl, 3- furyl, etc.; unsaturated 5 to 6-membered heteromonocyclic groups containing a sulfur atom, for example, 2-thienyl, 3-thienyl, etc.; unsaturated 5- to 6-membered heteromonocyclic groups containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, oxazolyl, isoxazolyl, oxadiazolyl [e.g., 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5- oxadiazolyl]; unsaturated 5 to 6- membered heteromonocyclic groups containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, thiadiazolyl [e.g., 1,2,4-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl]. The term "optionally substituted" denotes the substitution of a group herein by a moiety including, but not limited to, Ci-Cio alkyl, C2-C10 alkenyl, C2-C10 alkynyl, C3-C12 cycloalkyl, C3-

C12 cycloalkenyl, C1-C12 heterocycloalkyl, C3-C12 heterocycloalkenyl, C1-C10 alkoxy, aryl, aryloxy, heteroaryl, heteroaryloxy, amino, C1-C10 alkylamino, C1-C10 dialkylamino, arylamino, diarylamino, C1-C10 alkylsulfonamino, arylsulfonamino, C1-C10 alkylimino, arylimino, C1-C10 alkylsulfonimino, arylsulfonimino, hydroxyl, halo, thio, C1-C10 alkylthio, arylthio, C1-C10 alkylsulfonyl, arylsulfonyl, acylamino, aminoacyl, aminothioacyl, amidino, guanidine, ureido, cyano, nitro, azido, acyl, thioacyl, acyloxy, carboxyl, and carboxylic ester. In one alternative embodiment any suitable group may be present on a "substituted" or "optionally substituted" position if indicated that forms a stable molecule and meets the desired purpose of the invention and includes, but is not limited to, e.g., halogen (which can independently be F , CI, Br or I); cyano; hydroxyl; nitro; azido; alkanoyl (such as a C2-C6 alkanoyl group); carboxamide; alkyl, cycloalkyl, alkenyl, alkynyl, alkoxy, aryloxy such as phenoxy; thioalkyl including those having one or more thioether linkages; alkylsulfinyl; alkylsulfonyl groups including those having one or more sulfonyl linkages; aminoalkyl groups including groups having more than one N atoms; aryl (e.g., phenyl, biphenyl, naphthyl, or the like, each ring either substituted or unsubstituted); arylalkyl having for example, 1to 3 separate or fused rings and from 6 to about 14 or 18 ring carbon atoms, with benzyl being an exemplary arylalkyl group; arylalkoxy, for example, having 1 to 3 separate or fused rings with benzyloxy being an exemplary arylalkoxy group; or a saturated or partially unsaturated heterocycle having 1to 3 separate or fused rings with one or more N , O or S atoms, or a heteroaryl having 1to 3 separate or fused rings with one or more

N , O or S atoms, e.g. coumarinyl, quinolinyl, isoquinolinyl, quinazolinyl, pyridyl, pyrazinyl, pyrimidinyl, furanyl, pyrrolyl, thienyl, thiazolyl, triazinyl, oxazolyl, isoxazolyl, imidazolyl, indolyl, benzofuranyl, benzothiazolyl, tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholinyl, piperazinyl, and pyrrolidinyl. Such groups may be further substituted, e.g. with hydroxy, alkyl, alkoxy, halogen and amino. In certain embodiments "optionally substituted" includes one or more substituents independently selected from halogen, hydroxyl, amino, cyano,

-CHO, -COOH, -CO H 2, alkyl including Ci-Cealkyl, alkenyl including C2-C6alkenyl, alkynyl including C2-C6alkynyl, -Ci-C 6alkoxy, alkanoyl including C2-C6alkanoyl, Ci-C6alkylester, (mono- and di-Ci-C6alkylamino)Co-C2alkyl, haloalkyl including Ci-C6haloalkyl, hydoxyCi-C 6alkyl, ester, carbamate, , sulfonamide,-Ci-C6alkyl(heterocyclo), Ci-C6alkyl(heteroaryl), -Ci-C6alkyl(C3-

C7cycloalkyl), 0-Ci-C6alkyl(C3-C7cycloalkyl), B(OH)2, phosphate, phosphonate and haloalkoxy including Ci-C6haloalkoxy. "Aliphatic" refers to a saturated or unsaturated, straight, branched, or cyclic hydrocarbon. "Aliphatic" is intended herein to include, but is not limited to, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, and cycloalkynyl moieties, and thus incorporates each of these definitions. In one embodiment, "aliphatic" is used to indicate those aliphatic groups having 1-20 carbon atoms. The aliphatic chain be, for example, mono-unsacurated, di-unsaturated, r -unsaturated, or polyunsaturated, or alkynyl. Unsaturated aliphatic groups can be in a cis or trans configuration. In one embodiment, the aliphatic group contains from 1 to about 12 carbon atoms, more generally from 1to about 6 carbon atoms or from 1to about 4 carbon atoms. In one embodiment, the aliphatic group contains from 1to about 8 carbon atoms. In certain embodiments, the aliphatic group is Ci-

C2, C1-C3, C1-C4, C1-C5 or Ci-C6. The specified ranges as used herein indicate an aliphatic group having each member of the range described as an independent species. For example, the term Ci-

C6 aliphatic as used herein indicates a straight or branched alkyl, alkenyl, or alkynyl group having from 1, 2, 3, 4, 5, or 6 carbon atoms and is intended to mean that each of these is described as an independent species. For example, the term C1-C4 aliphatic as used herein indicates a straight or branched alkyl, alkenyl, or alkynyl group having from 1, 2, 3, or 4 carbon atoms and is intended to mean that each of these is described as an independent species. In one embodiment, the aliphatic group is substituted with one or more functional groups that results in the formation of a stable moiety. The term "heteroaliphatic" refers to an aliphatic moiety that contains at least one heteroatom in the chain, for example, an amine, carbonyl, carboxy, oxo, thio, phosphate, phosphonate, nitrogen, phosphorus, silicon, or boron atoms in place of a carbon atom. In one embodiment, the only heteroatom is nitrogen. In one embodiment, the only heteroatom is oxygen. In one embodiment, the only heteroatom is sulfur. "Heteroaliphatic" is intended herein to include, but is not limited to, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyl, heterocycloalkenyl, and heterocycloalkynyl moieties. In one embodiment, "heteroaliphatic" is used to indicate a heteroaliphatic group (cyclic, acyclic, substituted, unsubstituted, branched or unbranched) having 1-20 carbon atoms. In one embodiment, the heteroaliphatic group is optionally substituted in a manner that results in the formation of a stable moiety. Nonlimiting examples of heteroaliphatic moieties are polyethylene glycol, polyalkylene glycol, amide, polyamide, polylactide, polyglycolide, thioether, ether, alkyl-heterocycle-alkyl, -O-alkyl-O-alkyl, alkyl-O- haloalkyl, etc. A "dosage form" means a unit of administration of an active agent. Examples of dosage forms include tablets, capsules, injections, suspensions, liquids, emulsions, implants, particles, spheres, creams, ointments, suppositories, inhalable forms, transdermal forms, buccal, sublingual, topical, gel, mucosal, and the like. A "dosage form" can also include an implant, for example an optical implant. An "effective amount" as used herein, means an amount which provides a therapeutic or prophylactic benefit. As used herein "endogenous" refers to any material from or produced inside an organism, cell, tissue or system. As used herein, the term "exogenous" refers to any material introduced from or produced outside an organism, cell, tissue or system. By the term "modulating," as used herein, is meant mediating a detectable increase or decrease in the level of a response in a subject compared with the level of a response in the subject in the absence of a treatment or compound, and/or compared with the level of a response in an otherwise identical but untreated subject. The term encompasses perturbing and/or affecting a native signal or response thereby mediating a beneficial therapeutic response in a subject, preferably, a human. "Parenteral" administration of an immunogenic composition includes, e.g., subcutaneous (s.c), intravenous (i.v.), intramuscular (i.m.), or intrasternal injection, or infusion techniques. As used herein, the terms "peptide," "polypeptide," and "protein" are used interchangeably, and refer to a compound comprised of amino acid residues covalently linked by peptide bonds. A protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein's or peptide's sequence. Polypeptides include any peptide or protein comprising two or more amino acids j oined to each other by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types. "Polypeptides" include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others. The polypeptides include natural peptides, recombinant peptides, synthetic peptides, or a combination thereof. To "treat" a disease as the term is used herein, means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject. Ranges: throughout this disclosure, various aspects of the invention can be presented in a range format. It should be understood that the description in range format is merely for convenience and should not be construed as a limitation on the scope of the invention. The description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1to 3, from 1to 4, from 1to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6 . This applies regardless of the breadth of the range. As used herein, "pharmaceutical compositions" are compositions comprising at least one active agent, and at least one other substance, such as a carrier. "Pharmaceutical combinations" are combinations of at least two active agents which may be combined in a single dosage form or provided together in separate dosage forms with instructions that the active agents are to be used together to treat any disorder described herein. As used herein, "pharmaceutically acceptable salt" is a derivative of the disclosed compound in which the parent compound is modified by making inorganic and organic, non-toxic, acid or base addition salts thereof. The salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are typical, where practicable. Salts of the present compounds further include solvates of the compounds and of the compound salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts and the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, conventional non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC-(CH2)n- COOH where n is 0-4, and the like, or using a different acid that produces the same counterion. Lists of additional suitable salts may be found, e.g., in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., p . 1418 (1985). The term "carrier" applied to pharmaceutical compositions/combinations of the invention refers to a diluent, excipient, or vehicle with which an active compound is provided. A "pharmaceutically acceptable excipient" means an excipient that is useful in preparing a pharmaceutical composition/combination that is generally safe, non-toxic and neither biologically nor otherwise inappropriate for administration to a host, typically a human. In one embodiment, an excipient is used that is acceptable for veterinary use. A "patient" or "host" or "subject" is a human or non-human animal in need of treatment or prevention of any of the disorders as specifically described herein, for example that is modulated by a natural (wild-type) or modified (non-wild type) protein that can be degraded according to the present invention, resulting in a therapeutic effect. Typically, the host is a human. A "host" may alternatively refer to for example, a mammal, primate (e.g., human), cow, sheep, goat, horse, dog, cat, rabbit, rat, mice, fish, bird and the like. A "therapeutically effective amount" of a pharmaceutical composition/combination of this invention means an amount effective, when administered to a host, to provide a therapeutic benefit such as an amelioration of symptoms or reduction or diminution of the disease itself.

II. COMPOUNDS Formulas I-X In one aspect of the present invention a Degronimer (compound) of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, or Formula X is rovided: or a pharmaceutically acceptable salt, N-oxide, isotopic derivative or prodrug, optionally in a pharmaceutically acceptable carrier to form a pharmaceutically acceptable composition, wherein the variables are as defined above. In an additional embodiment T is hydrogen. In an additional embodiment T is hydrogen and R is methyl. 4 1

In some nonlimiting embodiments alkyl is a small alkyl, in other nonlimiting embodiments alkyl contains a branch point (for example isopropyl).

Examples of the following

haloalkyk , Χ

Examples of the following: Additional examples of the following:

Additional examples of the following:

Non-limitin examples of compounds described by Formula I include:

53

Non-limitin examples of Formula IV include:

additional embodiment the com ound of Formula IV is selected from:

Non-limiting examples of Formula V include:

60 6 1 62

64 65 66

In an additional embodiment the compound of Formula V is selected from the below formulas:

70 In an additional embodiment the compound of Formula VII is selected from the below formulas: Formulas XI-XVI In another aspect of the present invention a compound of Formula XI, Formula XII, Formula XIII Formula XIV, Formula XV, or Formula XVI is provided: or a pharmaceutically acceptable salt, N-oxide, isotopic derivative, prodrug, optionally in a pharmaceutically acceptable carrier to form a composition, wherein the variables are as defined above. In one embodiment when the term alkyl is used in the formula above in some embodiments it may independently refer to a Ci-C 6, C1-C5, C1-C4, C1-C3 or C1-C2 moiety. In one embodiment both R and T are hydrogen or deuterium. In an additional embodiment T is hydrogen. In an additional embodiment T is hydrogen and R is methyl.

or

Embodiments of Degron-Linker-Targeting Ligand In one embodiment the compound of Formula I is selected from:

( ); wherein

1 18' A is CR R , C=0, C=S, C=CH 2, S0 2, S(0); 2 4 14' A is CR R , C=0, C=S, C=CH 2, SO2, S(O); 3 15' A is CR R , C=0, C=S, C=CH 2, SO2, S(O); 9 6 16' X" is independently selected from NH, N(R ), CH2, CH(R ), C(R )2, C(R )(R ), O, and S;

Z is independently selected from O, S, and CH2; m is 0, 1, or 2;

= = is a single or double bond; R 4, R 14', R 5, R , R 6, R 16', R 8, R 18', R 9, R 19' are independently selected from: hydrogen, deuterium, alkyl, hydroxyl, alkoxy, amine, -NHalkyl, -Nalkyh; or R 8 and R 8 form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R 4 and R 4 form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R 5 and R 5 form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R 6 and R 6 form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R 9 and R 9 form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R 4 and R 5 form a 1 or 2 carbon bridged ring; or R 4 and R form a 1 or 2 carbon bridged ring; or R 9 and R 6 form a 1 or 2 carbon bridged ring; in a typical embodiment A1 is C=0; in a typical embodiment A2 is C=0; in a typical embodiment A3 is C=0;

9 6 16' X' is independently selected from NH, N(R ), CH2, CH(R ), C(R )2, C(R )(R ), O, and S; Q Q2 Q3 an d Q4 are independently selected from CH, C(R ), and N; and R 7 in each instance is independently selected from hydrogen, deuterium, alkyl, alkenyl, alkynl, fluorine, chlorine, bromine, iodine, hydroxyl, alkoxy, azide, amino, -NHalkyl, -N(alkyl)2,

-NHSChalkyl, -N(alkyl)S0 2alkyl, -NHS0 2aryl, -N(alkyl)S0 2aryl, -NHSChalkenyl, -

N(alkyl)S0 2alkenyl, -NHSChalkynyl, -N(alkyl)S0 2alkynyl, cyano, nitro, nitroso, -SH, -Salkyl, -

S(0)alkyl, -S(0) 2alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, haloalkyl, aryl, heteroaryl, heteroaliphatic, aliphatic, carbocyclo; or two R 7 substituents together with the carbon atom(s) to which they are bound can form a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or two R 7 substituents together with the carbon atom(s) to which they are bound can form a 1 or 2 carbon bridged ring or a 3, 4, 5, or 6 membered fused ring. In one embodiment the compound of Formula II is selected from:

(me).

In one embodiment the compound of Formula IV is selected from:

(IVa), (IVd), and In one embodiment the com ound of Formula V is selected from:

(Vb), 84 In one embodiment the compound of Formula VI is selected from:

(Vic), (Vie), In an alternative embodiment, the compound of Formula VI is selected from:

(Vlaa), (Vlad), (Vlag), (Vlaj),

(Vlap),

(Vlat),

In one embodiment the compound of Formula VII is selected from:

(Vllb), alternative embodiment, the compound of Formula VII is selected from (Vllaa),

(Vllac), 5 (VHal), (Vllao), (Vllar), (Vllat), embodiment the compound of Formula VIII is selected from:

(Villa) Vllb),

(Vllld), and In one embodiment the com ound of Formula IX is selected from:

(Kb),

104

106 107

109 110 111 112 113 114 115 116 117 wherein R30 is H, S, or O . alternative embodiment the compound of Formula I is selected from:

(I-la), 119 (I- If), and (i-ig); wherein R4 and R42 are independently selected from hydrogen, Ci-C4alkyl, Ci-C4alkenyl, Ci- C4alkynyl, C3-Ciocycloalkyl, aryl, heteroaryl, aliphatic or heteroaliphatic;

43 45 R is hydrogen, CF3, -C2H5, Ci-C 4alkyl, Ci-C 4alkenyl, Ci-C 4alkynyl, CH2R ; 44 R is Ci-Ci 6alkyl, Ci-Ci6alkenyl, Ci-Ci 6alkynyl, C3-Ciocycloalkyl, -(CH2)o-6aryl, or (CH2)o-6heteroary1;

45 R is hydrogen, -OH, F, CI, -CH 3, -CF 3, -CH2CI, -CH2F, or CH2OH; R 5 is hydrogen or Ci-Cioalkyl; R52 is hydrogen or Ci-Cioalkyl; R 53 is hydrogen, Ci-Cioalkyl, or Ci-Ciocycloalkyl; R54 is Ci-Cioalkyl, Ci-Ciocycloalkyl, heterocycloalkyl, cycloalkylalkyl, heterocycloalkylalkyl, aryl, arylalkyl, heteroaryl, or heteroaryl alkyl; R56 is methyl, ethyl, or cyclopropyl; and All of the other variables are as defined above. alternative embodiment the compound of Formula II is selected from:

(II-lc),

II-If), and

(Π-lg)

In an alternative embodiment the compound of Formula III is selected from:

(III-le), (III-lg)

In an alternative embodiment the compound of Formula IV is selected from:

(IV-le), (IV-lg) e embodiment the compound of Formula V is selected from: (V-la), ı 32 (V-lg) alternative embodiment the compound of Formula VI is selected from:

(VI-la), (VI-lc), (VI- If), and ve embodiment the compound of Formula VII is selected from:

(VII-la), (Vll-ld), (Vll-lg) alternative embodiment the compound of Formula VIII is selected from: (XVIIIc), 140 (VIII- If), and

(Vlll-lg) ive embodiment the compound of Formula IX is selected from:

(K-la), IX-lc),

(IX-ld), (IX- If), and (IX-lg) X-l a),

5 (x-ig).

In an additional embodiment the compound of the present invention is selected from:

150 151 ı 52

ı 54 Degron In one aspect of the present invention a Degron of Formula NI, Formula Nil, or Formula sed in the present invention: wherein: c=CH S0 , S(O), P(0)Oalkyl, P(0)NHalkyl, W 2oi i s C R 206 R 207 c = 0 C=S 2, 2

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H 2; 202 0 209 W is CR R , C=0, C=S, C=CH 2, SO2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H 2; in a typical embodiment W 201 is C=0; in another typical embodiment W 202 is C=0;

200 203 203 0 X is independently selected from NH, R , CH 2, CHR , C(R )2, O, and S; n200 is 0, 1, 2, or 3; is a single or double bond;

wherein when represents a single bond, n is 0, 1, 2, or 3;

wherein when =-=-= re resents a double bond, n is 0, 1, or 2;

R is alkyl or hydrogen;

in some embodiments alkyl is Ci-C 6, C1-C5, C1-C4, C1-C3, C1-C2, or methyl; or R201 and R202 are combined to form a 4, 5, 6, 7, 8, 9, or 10 membered heterocyclo or heteroaryl species, wherein the heterocyclo or heteroaryl species is substituted with R212 at any desired position, wherein the heterocyclo or heteroaryl species is optionally further substituted with one or more substituents selected from R205 ; and in an additional alternative embodiment the heterocyclo or heteroaryl species is o tionally further substituted with one or more =0 (oxo) at a position allowed by valence;

or R201 * is selected from: R is selected at each instance from: alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, alkene, and alkyne, and in addition to these can also be selected from aliphatic, heteroaliphatic, aryl, heteroaryl, heteroalkyl; R204 is selected at each instance from: alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -NHalkyl, -N(alkyl)2, - H S0 2alkyl, -N(alkyl)S0 2alkyl, - H S0 2aryl, -

N(alkyl)S0 2aryl, - H S0 2alkenyl, -N(alkyl)S0 2alkenyl, - HS0 2alkynyl, -N(alkyl)S0 2alkynyl, and haloalkyl; and in addition to these can also be selected from aliphatic, heteroaliphatic, aryl, heteroaryl and heteroalkyl; or two R204 substituents together with the carbon atom(s) to which they are bound can form a 3, 4, 5 or 6 membered ring; R205 and R214 are selected at each instance from: hydrogen, alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -NHalkyl, -N(alkyl)2, -NHSChalkyl, -N(alkyl)S0 2alkyl,

-NH S aryl, -N(alkyl)S0 2aryl, -NHS0 2alkenyl, -N(alkyl)S0 2alkenyl, -NHS0 2alkynyl,

-N(alkyl)S0 2alkynyl, and haloalkyl; and in addition to these can also be selected from aliphatic, heteroaliphatic (including heteroalkyl), aryl, heteroaryl, heterocyclic; or in the alternative, R205 is independently selected from C(0)R 204, cyano, aryl, aryloxy, heterocyclo, heteroaryl, arylalkyl, alkoxy, hydroxyl, O-aiylalkyl, or cycloalkyl; each of which R205 can be optionally substituted, for example, with one or more substituents selected from alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -

NHalkyl, -N(alkyl)2, aryl, heterocyclo, heteroaryl, haloalkyl, and cycloalkyl; R206, R207, R208, R209, R210, and R211, are independently selected from hydrogen, alkyl, hydroxyl, alkoxy, amine, -NHalkyl, and -Nalkyl2; or R206 and R207 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R208 and R209 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R210 and R211 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R206 and R208 form a 1 or 2 carbon bridged ring; or R206 and R form a 1 or 2 carbon bridged ring; or R208 and R2 0 form a 1 or 2 carbon bridged ring; or R214 and R206 form a 3, 4, 5, or 6 carbon fused ring; or R214 and R210 form a 3, 4, 5, or 6 carbon fused ring; or R214 and R208 form a 1 or 2 carbon bridged ring; or R214 and R204 form a 3, 4, 5, or 6 carbon fused ring wherein R203 is on the carbon alpha to R2 4 or a 1, 2, 3, or 4 carbon bridged ring wherein R203 is not on the carbon alpha to R2 4; R212 is the point at which the linker is attached; R222 is independently selected at each occurrence from hydrogen, alkyl, alkene, alkyne, haloalkyl, alkoxy, hydroxyl, aryl, heteroaryl, heterocycle, arylalkyl, heteroarylalkyl, heterocycloalkyl, aryloxy, heteroaryloxy, CN, -COOalkyl, COOH, NO2, F, CI, Br, I, CF3, NH2, 225 225 225 204 NHalkyl, N(alkyl) 2, NR R , NHR , aliphatic, heteroaliphatic, and COR ; and in addition to these can also be selected from aliphatic and heteroaliphatic; R225 is selected at each instance from: alkyl, -C(0)H, -C(0)OH. -C(0)alkyl, -C(0)Oalkyl, alkenyl, or alkynyl or alternatively can be aliphatic, heteroaliphatic, aryl, heteroaryl or heterocyclic; and when R212 is bonded to a A that is carbon, then A is CR212;

In another aspect of the present invention a Degron of Formula CI or Formula CII is used in the present invention:

wherein:

401 401 402 W is CR R , C=0, C=S, C=CH 2, S0 2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; 402 40 404 W is CR R , C=0, C=S, C=CH 2, SO2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; in a typical embodiment W401 is C=0; in another typical embodiment W402 is C=0;

400 412 412 412 X is independently selected from NH, NR , CH2, CHR , C(R )2, O, or S; n200 is 0, 1, 2, or 3; is a sin le or double bond; 5 A is independently selected from C(R ), and N wherein in certain embodiments the number of nitrogen atoms is 0, 1, 2, 3, or 4 per ring (as allowed by context) and is selected to produce a stable ring and a pharmaceutically acceptable Degronimer. When A's are in a six- membered ring (unfused or fused), the ring can be, in non-limiting embodiments as allowed by context, a pyridine, diazine, triazine, pyrimidine, pyridazine, pyrazine, triazine or tetrazine.

Z20 is H, O, S, or R 412; and when R410 is bonded to a A that is carbon, then A is CR410, and when R410 is bonded to a Z20 or Z402 that is nitrogen, then Z20 or Z402 is R 410, etc; R is independently selected at each instance from hydrogen, alkyl, alkenyl, alkynyl, fluorine, chlorine, bromine, iodine, hydroxyl, alkoxy, cyano, azide, amino, -NH(aliphatic or alkyl),

-N(aliphatic or alkyl)2, - H S0 2(aliphatic or alkyl), -N(aliphatic or alkyl)S0 2(aliphatic or alkyl), -

H S0 2(aryl or heteroaryl), -N(aliphatic including alkyl)S0 2aryl, -NHSChalkenyl, -

N(alkyl)S0 2alkenyl, -NHS0 2alkynyl, -N(alkyl)S0 2alkynyl, cyano, nitro, nitroso, -SH, -Salkyl, -

S(0)alkyl, -S(0) 2alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, aliphatic, heteroaliphatic, aryl, heteroaryl, carbocyclic, and haloalkyl; R401, R402, R403, R404, R407, R408, and R415 are independently selected from hydrogen, alkyl, aliphatic, heteroaliphatic, heterocyclic, carbocyclic, aryl, heteroaryl, hydroxyl, halo, azide, CN-, alkoxy, amine, -NHalkyl, and -Nalkyh, -NH(aliphatic), and -N(independently aliphatic) 2, each of which may be optionally substituted as described in the Definition Section, if desired to achieve the target effect, results in a stable compound that makes chemical sense to the routineer, and the group is not redundant (i.e., as known in the art, alkyl substituted with alkyl is redundant; however for examples, alkoxy substituted with alkoxy is not redundant); or R401 and R402 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R403 and R404 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R407 and R408 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R401 and R403 form a 1 or 2 carbon bridged ring; or R401 and R407 form a 1 or 2 carbon bridged ring; or R403 and R407 form a 1 or 2 carbon bridged ring; or R415 and R401 form a 3, 4, 5, or 6 carbon fused ring; or R415 and R407 form a 3, 4, 5, or 6 carbon fused ring; or R415 and R403 form a 1 or 2 carbon bridged ring; or R 4 and R405 form a 3, 4, 5, or 6 carbon fused ring wherein R405 is on the carbon alpha to R415 or a 1, 2, 3, or 4 carbon bridged ring wherein R405 is not on the carbon alpha to R415 ; R405 is selected at each instance from: alkyl, alkene, alkyne, aliphatic, heteroaliphatic, heterocyclic, aryl, heteroaryl, halogen, hydroxyl, alkoxy, azide, amino, -NH(alkyl or aliphatic), - N(independently alkyl or aliphatic)2, -NHS02(aliphatic, including alkyl), -N(alkyl or aliphatic)S02(alkyl or aliphatic), - H SC aryl, -N(alkyl or aliphatic)S02aryl, -NHSCtealkenyl, - N(alkyl or aliphatic)S02alkenyl, -NHS02alkynyl, -N(alkyl or aliphatic)S02alkynyl, and halo(alkyl or aliphatic), each of which is provided to form a stable compound as known to those of skill in the art, and can be optionally substituted as described in the Definition Section, if desired to achieve the target effect, results in a stable compound that makes chemical sense to the routineer, and the group is not redundant (i.e., as known in the art, alkyl substituted with alkyl is redundant; however for examples, alkoxy substituted with alkoxy is not redundant); or two R405 substituents together with the carbon atom(s) to which they are bound can form a 3, 4, 5 or 6 membered ring; R410 is the point at which the linker is attached; R411 is selected at each instance from: hydrogen, alkyl, alkenyl, alkynyl, halogen, hydroxyl, heterocyclic, heteroalkyl, carbocyclic, heteroaliphatic, aliphatic, alkoxy, aryl, heteroaryl, alkylamino, alkylhydroxyl, -NHalkyl, -Nalkyh, -NH(aliphatic), -N(independently aliphatic)2, amino, cyano, nitro, nitroso, sulfone, sulfoxide, thioalkyl, thiol and haloalkyl, each of which is optionally substituted as described in the Definition Section, if desired to achieve the target effect, results in a stable compound that makes chemical sense to the routineer; R412 is selected at each instance from: hydrogen, alkyl, aliphatic, heteroaliphatic, heterocyclic, heteroaryl, aryl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, -C(0)(aliphatic, aryl, heteroaliphatic, aryl or heteroaryl), -C(0)0(aliphatic, aryl, heteroaliphatic, aryl or heteroaryl), alkene, and alkyne, each of which is optionally substituted as described in the Definition Section, if desired to achieve the target effect, results in a stable compound that makes chemical sense to the routineer; R413 and R414 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, hydroxy, amino, -NHalkyl, and -N(alkyl)2, each of which is optionally substituted as described in the Definition Section, if desired to achieve the target effect, results in a stable compound that makes chemical sense to the routineer; and or R413 and R414 together with the carbon atom to which they are attached, form C(O), C(S),

C=CH2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O . In another aspect of the present invention a Degron of Formula CIII is used in the present invention:

wherein the R moieities are as described above. In another aspect of the present invention a Degron of Formula CIII is used in the present invention: In another aspect of the present invention a Degron of Formula CIV is used in the present invention:

wherein: R417 is selected from

In any of the fused rings that have an R410, the R410 can be placed on any available ring atom on either of the fused rings, except when excluded by context (such as where valency precludes), for example, as shown in the formulas:

includes compounds of structure

and each is considered specifically and independently described.

Additional Degrons In certain aspects, the Degron covalently or non-covalently binds to an E3 ligase to afford degradation of the targeted protein. In one embodiment the E3 ligase is cereblon. In another embodiment the E3 ligase is VHL. Non-limiting examples of Degrons for use in the present invention include the thalidomide analogues described in WO 2014180882, WO 2014147531, WO 2014068506, WO 2014018866, WO 2013130849, US 20130143922, WO 2012177678, US 20120283292, WO 2012135299, WO 2012079022, WO 2012079075, WO 2012068512, JP 201 1012014, US 20100240651, WO 2010053732, WO 2009145899, JP 2009215195, JP 2009001528, WO 20081 15516, WO 2008033567, WO 2008027542, WO 2007136640, and US 20070197791. In one embodiment the Degron is selected from: thalidomide, lenalidomide, , CC-122, CC-223, CC-292 or analogues thereof. In one embodiment the Degron is thalidomide or an analogue thereof. In another embodiment the Degron is lenalidomide or an analogue thereof. In one embodiment the Degron is pomalidomide or an analogue thereof. In another embodiment the Degron is CC-122 or an analogue thereof. In one embodiment the Degron is CC-223 or an analogue thereof. In another embodiment the Degron is CC-292 or an analogue thereof. In one embodiment the Degron is thalidomide. In another embodiment the Degron is lenalidomide. In one embodiment the Degron is pomalidomide. In another embodiment the Degron is CC-122. In one embodiment the Degron is CC-223. In another embodiment the Degron is CC- 292.

Linker A Linker is included in the Degronimers of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, and Formula XXIX,. Linker is a bond or a chemically stable group that attaches a Degron to a Targeting Ligand. Any of the Linkers described herein can be used in either direction, i.e., either the left end is linked to the Degron and the right end to the Target Linker, or the left end is linked to the Target Linker and the right end is linked to the Degron. According to the invention, any desired linker can be used as long as the resulting compound has a stable shelf life for at least 2 months, 3 months, 6 months or 1 year as part of a pharmaceutically acceptable dosage form, and itself is pharmaceutically acceptable. In a typical embodiment, the Linker has a chain of 2 to 14, 15, 16, 17, 18 or 20 or more carbon atoms of which one or more carbons can be replaced by a heteroatom such as O, N, S, or

P. In certain embodiments the chain has 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 contiguous atoms in the chain. For example, the chain may include 1 or more ethylene glycol units that can be contiguous, partially contiguous or non-contiguous (for example, 2, 3, 4, 5, 6, 7,

8, 9, 10, 11 or 12 ethylene glycol units). In certain embodiments the chain has at least 1, 2, 3, 4, 5, 6, 7, or 8 contiguous chains which can have branches which can be independently alkyl, heteroalkyl, aryl, heteroaryi, alkenyl, or alkynyl, aliphatic, heteroaliphatic, cycloalkyl or heterocyclic substituents. In other embodiments, the linker can include or be comprised of one or more of ethylene glycol, propylene glycol, lactic acid and/or glycolic acid. In general, propylene glycol adds hydrophobicity, while propylene glycol adds hydrophilicity. Lactic acid segments tend to have a longer half-life than glycolic acid segments. Block and random lactic acid-co-glycolic acid moieties, as well as ethylene glycol and propylene glycol, are known in the art to be pharmaceutically acceptable and can be modified or arranged to obtain the desired half-life and hydrophilicity. In certain aspects, these units can be flanked or interspersed with other moieties, such as aliphatic, including alkyl, heteroaliphatic, aryl, heteroaryi, heterocyclic, cycloalkyl, etc., as desired to achieve the appropriate drug properties. In one embodiment, the Linker is a moiety selected from Formula LI, Formula LII, Formula LIII, Formula LIV, Formula LV, Formula LVI, and Formula LVII: i X 1 Heieroary!

p22 20 \ Heteroaryl- R23 R2 X2 X1 (LIV),

wherein:

1 2 25 25 X and X are independently selected from bond, NH, NR , CH2, CHR , C(R )2, O, and S; R20, R2 , R22, R23, and R24 are independently selected from bond, alkyl, -C(O)- -C(0)0-, - OC(O)-, -C(0)alkyl, -C(0)Oalkyl, -C(S)-, -SO2-, -S(O)-, -C(S)-, -C(0)NH-, -NHC(O)-, - N(alkyl)C(0)-, -C(0)N(alkyl)-, -0-, -S-, -NH-, -N(alkyl)-, -CH(-0-R 26)-, -CH(-NHR 25)-, -CH(-

6 NH2)-, -CH(-NR 2)-, -C(-0-R )alkyl-, -C(-NHR )alkyl-, -C(-NH 2)alkyl-, -C(-NR 2)alkyl-, - C(R4R4)-, -alkyl(R )-alkyl(R 28)-, -C(R R28)-, -P(0)(OR 6)0-, -P(0)(OR 26)-, -NHC(0)NH-, - N(R )C(0)N(R 25)-, -N(H)C(0)N(R 25)-, polyethylene glycol, poly(lactic-co-glycolic acid), alkene, haloalkyl, alkoxy, and alkyne; or R20, R2 , R22, R23, and R24 can in addition to those above be independently selected from heteroarylalkyl, aryl, arylalkyl, heterocycle, aliphatic, heteroaliphatic, heteroaryl, polypropylene glycol, lactic acid, glycolic acid, carbocycle, or -0-(CH 2)i-i2-0-, -NH-(CH 2)i-i2-NH-, -NH-(CH 2)i-

12-O-, or -0-(CH 2)i-i2-NH-, -S-(CH 2)i-i2-0-, -0-(CH 2)i-i2-S-, -S-(CH 2)i-i2-S-, -S-(CH 2)i-i2-NH-, -

NH-(CH 2)i-i2-S-, (and wherein the 1-12 can be independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12, and wherein one or more of the CH2 or NH can be modified by substitution of a H for a methyl, ethyl, cyclopropyl, F (if on carbon), etc, as described herein), and optionally, a heteroatom, heteroalkyl, aryl, heteroaryl or cycloaliphatic group is interspersed in the chain). Certain nonlimiting examples include -0-CH(CH 3)-CH(CH 3)CH-0-, -0-CH 2-CH(CH3)CH-0-, -O-

CH(CH3)-CH2CH-0-, etc. each of which R20, R2 , R22, R23, and R24 is optionally substituted with one or more substituents selected from R 101 or alternatively as described in Section 1. Definitions; R25 is selected at each instance from: alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, alkenyl, or alkynyl or alternatively can be aliphatic, heteroaliphatic, aryl, heteroaryl or heterocyclic; R26 is hydrogen, alkyl, silane, arylalkyl, heteroarylalkyl, alkene, and alkyne; or in addition to these can also be selected from aryl, heteroaryl, heterocyclic, aliphatic and heteroaliphatic; R27 and R28 are independently selected from hydrogen, alkyl, amine, or together with the carbon atom to which they are attached, form C(O), C(S), C=CH2, a C3-C6 spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O, or form a 1 or 2 carbon bridged ring; R4 is independently selected from hydrogen, deuterium, alkyl, aliphatic, hydroxyl, alkoxy, aliphaticoxy, amine, cyano, -NH(aliphatic including alkyl), -N(aliphatic, including alkyl)2, fluorine, chlorine, bromine, iodine, haloalkyl (including -CFH 2, CHF2, and -CF3), alkenyl, alkynyl, aryl, heteroaryl, heterocyclo and carbocyclic; and R 101 is independently selected at each occurrence from hydrogen, alkyl, alkene, alkyne, haloalkyl, alkoxy, hydroxyl, aryl, heteroaryl, heterocycle, arylalkyl, heteroarylalkyl, heterocycloalkyl, aryloxy, heteroaryloxy, CN, -COOalkyl, COOH, N0 2, F, CI, Br, I, CF , H 2,

NHalkyl, N(alkyl) 2, aliphatic, and heteroaliphatic. In an additional embodiment, the Linker is a moiety selected from Formula LVIII, LIX, and LX:

(LVIII), wherein each variable is as it is defined in Formula LI. In alternative embodiments of LVIII, LIX and LX, a carbocyclic ring is used in place of the heterocycle. The following are non-limiting examples of Linkers that can be used in this invention. Based on this elaboration, those of skill in the art will understand how to use the full breadth of Linkers that will accomplish the goal of the invention. As certain non-limiting examples, Formula LI, Formula LII, Formula LIII, Formula LIV, Formula LV, Formula LVI, or Formula LVII include:

ı 74

In one embodiment X1 is attached to the Targeting Ligand. In another embodiment X2 is attached to the Targeting Ligand.

Additional non-limiting examples of moieties of R20, R2 , R22, R23, and R24 include Additional non-limiting examples of moieties of R20, R2 , R22, R23, and R24 include:

In additional embodiments, the Linker group is an optionally substituted (poly)ethylene glycol having at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, ethylene glycol units, or optionally substituted alkyl groups interspersed with optionally substituted, O, N, S, P or Si atoms. In certain embodiments, the Linker is flanked, substituted, or interspersed with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group. In certain embodiments, the Linker may be asymmetric or symmetrical. In some embodiments, the Linker is a substituted or unsubstituted polyethylene glycol group ranging in size from about 1 to about 12 ethylene glycol units, between 1 and about 10 ethylene glycol units, about 2 about 6 ethylene glycol units, between about 2 and 5 ethylene glycol units, between about 2 and 4 ethylene glycol units. In any of the embodiments of the compounds described herein, the Linker group may be any suitable moiety as described herein. In additional embodiments, the Linker is selected from:

6 6 - (CH2)ni-(lower alkyl)-, - (CH2)ni-(lower alkoxyl)-, 6 6 - (CH2)ni-(lower alkoxyl)-OCH 2-, - (CH2)ni-(lower alkoxyl)-(lower alkyl)-OCH 2-, 6 6 - (CH2)ni-(cycloalkyl)-(lower alkyl)-OCH 2-, - (CH2)ni-(heterocycloalkyl)-, 6 6 - (CH2CH20)ni-(lower alkyl)-0-CH 2-, - (CH2CH20)ni-(heterocycloalkyl)-0-CH 2-, 6 6 - (CH2CH20)ni-Aryl-0-CH 2-, - (CH2CH20)ni-(heteroaiyl)-0-CH 2-, 6 - (CH2CH20)ni-(cycloalkyl)-0-(heteroaiyl)-0-CH 2-, 6 - (CH2CH20)ni-(cycloalkyl)-0-Aiyl-0-CH 2-, 6 - (CH2CH20)ni-(lower alkyl)-NH-Aryl-0- CH2-, 6 - (CH2CH20)ni-(lower alkyl)-0-Aiyl-CH 2, 6 6 - (CH2CH20)ni-cycloalkyl-0-Aiyl-, - (CH2CH20)ni-cycloalkyl-0-heteroaiyl-, 6 - (CH2CH2)ni-(cycloalkyl)-0-(heterocycle)-CH2, 6 6 - (CH2CH2)ni-(heterocycle)-(heterocycle)-CH2, and - -(heterocycle)-CH 2; wherein n l is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and R6 is H, methyl, or ethyl. In additional embodiments, the Linker is selected from: -N(R 6 )-(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-OCH -, 2 2 1 2 2 2 q 2 2 -0-(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-OCH -, 2 2 1 2 2 2 q 2 2 -0-(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-0-; 2 2 1 2 2 2 q 2 -N(R 6 )-(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-0-; 2 2 1 2 2 2 q 2 -(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-0-; 2 2 1 2 2 2 q 2 -(CH )mi-0(CH ) 2-0(CH )oi-0(CH )pi-0(CH ) i-0(CH )ri-OCH -; 2 2 1 2 2 2 q 2 2 -0(CH )miO(CH ) 20(CH )piO(CH ) iOCH -; 2 2 1 2 2 q 2

-0(CH 2)miO(CH2)n20(CH2)piO(CH2)qiOCH2-; wherein ml, n2, ol, pi, ql, and rl are independently 1, 2, 3, 4, or 5; and R6 is H, methyl, or ethyl. In additional embodiments, the Linker is selected from: 0{CH ) 0(CH } (C )p 0(CH ) OCH 2 2 i2 2 2 q2 2 ml, n2, ol, pi, q2, and rl are independently 1, 2, 3, 4, or 5. In additional embodiments, the Linker is selected from: 181 ı 82 1

ı 86 wherein R 7 is -0-, -NH, Nalkyl, heteroaliphatic, aliphatic or -NMe. In additional embodiments, the Linker is selected from:

190 191 ı 92

In additional embodiments, the Linker is selected from:

In additional embodiments, the Linker is selected from

^0¾, and 0

In ce ı 95 In certain embodiments, Linker can be a 4-24 carbon atom linear chains, wherein one or the carbon atoms in the linear chain can be replaced or substituted with oxygen, nitrogen, amide, fluorinated carbon, etc., such as the following:

In certain embodiments, Linker can be a nonlinear chain, and can be, or include, aliphatic or aromatic or heteroaromatic cyclic moieties. In certain embodiments, the Linker may include contiguous, partially contiguous or non contiguous ethylene glycol unit groups ranging in size from about 1 to about 12 ethylene glycol units, between 1 and about 10 ethylene glycol units, about 2 about 6 ethylene glycol units, between about 2 and 5 ethylene glycol units, between about 2 and 4 ethylene glycol units, for example, 1, 2, 3, 4, 6, 6, 7, 8, 9, 10, 11 or 12 ethylene glycol units.

In certain embodiments, the Linker may have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or

15 fluorine substituents. In another embodiment the Linker is perfluorinated. In yet another embodiment the Linker is a partially or fully fluorinated poly ether. Nonlimiting examples of fl ri nated Linkers include: In certain embodiments, where the Target Ligand binds more than one protein (i.e., is not completely selective), selectivity may be enhanced by varying Linker length where the ligand binds some of its targets in different binding pockets, e.g., deeper or shallower binding pockets than others. Therefore, the length can be adjusted as desired.

Targeted Proteins

Degradation of cellular proteins is required for cell homeostasis and normal cell function, such as proliferation, differentiation and cell death. A primary pathway for degradation of cellular proteins is through ubiquitin-dependent proteolysis. Protein ubiquitination is mediated by sequential reactions of ubiquitin-activating enzymes (El), ubiquitin-conjugating enzymes (E2), and ubiquitin-protein ligases (E3). The bromodomain is currently the only protein domain known to bind specifically to acetylated lysine residues in histone tails. Bromodomains, which are approximately 110 amino acids long, are found in a large number of chromatin-associated proteins and have now been identified in approximately 70 human proteins, often adjacent to other protein motifs. Proteins that contain a bromodomain may contain additional bromodomains, as well as other functional motifs. For example, many HATs also contain a bromodomain. Interactions between bromodomains and modified histones are an important mechanism underlying chromatin structural changes and gene regulation. Bromodomain-containing proteins have been implicated in disease processes including cancer, inflammation and viral replication. In one embodiment, using the compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, or Formula X, this invention targets for ubiquitination and degradation in vivo a bromodomain containing protein that mediates a disorder to be treated. The selected bromodomain containing protein may modulate a disorder in a human via a mechanism of action such as modification of a biological pathway, pathogenic signaling, modulation of a signal cascade or cellular entry. In one embodiment the bromodomain containing protein has histone acetyl transferase activity. In one embodiment, the bromodomain containing protein is BRD2, BRD3, BRD4, BRDT or ASH1L. In one embodiment, the bromodomain containing protein is a non-BET protein. In one embodiment, the non-BET protein is BRD7 or BRD9.

III. METHODS OF TREATMENT

The compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, can be used in an effective amount to treat a host with any of the disorders described herein, including a human, in need thereof, optionally in a pharmaceutically acceptable carrier. In certain embodiments, the method comprises administering an effective amount of the active compound or its salt as described herein, optionally including a pharmaceutically acceptable excipient, carrier, adjuvant, i.e., a pharmaceutically acceptable composition, optionally in combination or alternation with another bioactive agent or combination of agents. The compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, or a pharmaceutically acceptable salt thereof as described herein can be used to degrade a Target Protein which is a mediator of the disorder affecting the patient, such as a human. The reduction in the Target Protein level afforded by the Formula I, II, V or VII Degronimers of the present invention provides treatment of the implicated disease state or condition, which is modulated through the Target Protein by lowering the level of that protein in the cell, e.g., cell of a patient. The term "disease state or condition" when used in connection with a Formula I, II, V or VII compound is meant to refer to any disease state or condition wherein protein dysregulation occurs that involves the selected Target Protein and where degradation of such protein in a patient may provide beneficial therapy or relief of symptoms to a patient in need thereof. In certain instances, the disease state or condition may be cured. The compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, are useful as therapeutic agents when administered in an effective amount to a host, including a human, to treat a tumor, cancer (solid, non-solid, diffuse, hematological, etc), abnormal cellular proliferation, immune disorder, inflammatory disorder, blood disorder, a myelo- or lymphoproliferative disorder such as B- or T- cell lymphomas, multiple myeloma, breast cancer, prostate cancer, AML, ALL, ACL, lung cancer, pancreatic cancer, colon cancer, skin cancer, melanoma, Waldenstrom's macroglobulinemia, Wiskott-Aldrich syndrome, or a post-transplant lymphoproliferative disorder; an autoimmune disorder, for example, Lupus, Crohn's Disease, Addison disease, Celiac disease, dermatomyositis, Graves disease, thyroiditis, multiple sclerosis, pernicious anemia, reactive arthritis, or type I diabetes; a disease of cardiologic malfunction, including hypercholesterolemia; an infectious disease, including a viral and/or bacterial infection; an inflammatory condition, including asthma, chronic peptic ulcers, tuberculosis, rheumatoid arthritis, periodontitis, ulcerative colitis, or hepatitis. The term "disease state or condition" when used in connection with a Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, compound, for example, refers to any therapeutic indication which can be treated by decreasing the activity of cereblon or a cereblon-containing E3 Ligase, including but not limited to uses known for the cereblon binders thalidomide, pomalidomide or lenalidomide. Nonlimiting examples of uses for cereblon binders are multiple myeloma, a hematological disorder such as myelodysplastic syndrome, cancer, tumor, abnormal cellular proliferation, breast cancer, prostate cancer, AML, ALL, ACL, lung cancer, pancreatic cancer, colon cancer, skin cancer, melanoma, HIV/AIDS, HBV, HCV, hepatitis, Crohn's disease, sarcoidosis, graft-versus-host disease, rheumatoid arthritis, Behcet's disease, tuberculosis, and myelofibrosis. Other indications include a myelo- or lymphoproliferative disorder such as B- or T-cell lymphomas, Waldenstrom's macroglobulinemia, Wiskott-Aldrich syndrome, or a post-transplant lymphoproliferative disorder; an immune disorder, including autoimmune disorders for example as Lupus, Addison disease, Celiac disease, dermatomyositis, Graves disease, thyroiditis, multiple sclerosis, pernicious anemia, arthritis, and in particular rheumatoid arthritis, or type I diabetes; a disease of cardiologic malfunction, including hypercholesterolemia; an infectious disease, including viral and/or bacterial infection, as described generally herein; an inflammatory condition, including asthma, chronic peptic ulcers, tuberculosis, rheumatoid arthritis, periodontitis and ulcerative colitis. In certain embodiments, the present invention provides the administration of an effective amount of a compound to treat a patient, for example, a human, having an infectious disease, wherein the therapy targets a Target Protein of the infectious agent or host (Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX), The disease state or condition may be caused by a microbial agent or other exogenous agent such as a virus (as non-limiting examples, HIV, HBV, HCV, HSV, HPV, RSV, CMV, Ebola, Flavivirus, Pestivirus, Rotavirus, Influenza, Coronavirus, EBV, viral pneumonia, drug-resistant viruses, Bird flu, RNA virus, DNA virus, adenovirus, poxvirus, Picornavirus, Togavirus, Orthomyxovirus, Retrovirus or Hepadnovirus), bacteria (including but not limited to Gram-negative, Gram-positive, Atypical, Staphylococcus, Streptococcus, E . Coli, Salmonella, Helicobacter pylori, meningitis, gonorrhea, Chlamydiaceae, Mycoplasmataceae, etc), fungus, protozoa, helminth, worms, prion, parasite, or other microbe. In certain embodiments, the condition treated with a compound of the present invention is a disorder related to abnormal cellular proliferation. Abnormal cellular proliferation, notably hyperproliferation, can occur as a result of a wide variety of factors, including genetic mutation, infection, exposure to toxins, autoimmune disorders, and benign or malignant tumor induction. There are a number of skin disorders associated with cellular hyperproliferation. Psoriasis, for example, is a benign disease of human skin generally characterized by plaques covered by thickened scales. The disease is caused by increased proliferation of epidermal cells of unknown cause. Chronic eczema is also associated with significant hyperproliferation of the epidermis. Other diseases caused by hyperproliferation of skin cells include atopic dermatitis, lichen planus, warts, pemphigus vulgaris, actinic keratosis, basal cell carcinoma and squamous cell carcinoma. Other hyperproliferative cell disorders include blood vessel proliferation disorders, fibrotic disorders, autoimmune disorders, graft-versus-host rejection, tumors and cancers. Blood vessel proliferative disorders include angiogenic and vasculogenic disorders. Proliferation of smooth muscle cells in the course of development of plaques in vascular tissue cause, for example, , retinopathies and atherosclerosis. Both cell migration and cell proliferation play a role in the formation of atherosclerotic lesions. Fibrotic disorders are often due to the abnormal formation of an extracellular matrix. Examples of fibrotic disorders include hepatic cirrhosis and mesangial proliferative cell disorders. Hepatic cirrhosis is characterized by the increase in extracellular matrix constituents resulting in the formation of a hepatic scar. Hepatic cirrhosis can cause diseases such as cirrhosis of the liver. An increased extracellular matrix resulting in a hepatic scar can also be caused by viral infection such as hepatitis. Lipocytes appear to play a major role in hepatic cirrhosis. Mesangial disorders are brought about by abnormal proliferation of mesangial cells. Mesangial hyperproliferative cell disorders include various human renal diseases, such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic micro angiopathy syndromes, transplant rejection, and glomerulopathies. Another disease with a proliferative component is rheumatoid arthritis. Rheumatoid arthritis is generally considered an autoimmune disease that is thought to be associated with activity of autoreactive T cells, and to be caused by autoantibodies produced against collagen and IgE. Other disorders that can include an abnormal cellular proliferative component include Bechet's syndrome, acute respiratory distress syndrome (ARDS), ischemic heart disease, post- dialysis syndrome, leukemia, acquired immune deficiency syndrome, vasculitis, lipid histiocytosis, septic shock and inflammation in general. Cutaneous contact hypersensitivity and asthma are just two examples of immune responses that can be associated with significant morbidity. Others include atopic dermatitis, eczema, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions. These conditions may result in any one or more of the following symptoms or signs: itching, swelling, redness, blisters, crusting, ulceration, pain, scaling, cracking, hair loss, scarring, or oozing of fluid involving the skin, eye, or mucosal membranes. In atopic dermatitis, and eczema in general, immunologically mediated leukocyte infiltration (particularly infiltration of mononuclear cells, lymphocytes, neutrophils, and eosinophils) into the skin importantly contributes to the pathogenesis of these diseases. Chronic eczema also is associated with significant hyperproliferation of the epidermis. Immunologically mediated leukocyte infiltration also occurs at sites other than the skin, such as in the airways in asthma and in the tear producing gland of the eye in keratoconjunctivitis sicca. In one non-limiting embodiment compounds of the present invention are used as topical agents in treating contact dermatitis, atopic dermatitis, eczematous dermatitis, psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions. The novel method may also be useful in reducing the infiltration of skin by malignant leukocytes in diseases such as mycosis fungoides. These compounds can also be used to treat an aqueous-deficient dry eye state (such as immune mediated keratoconjunctivitis) in a patient suffering therefrom, by administering the compound topically to the eye. Disease states which may be treated according to the present invention include, for example, asthma, autoimmune diseases such as multiple sclerosis, various cancers, ciliopathies, cleft palate, diabetes, heart disease, hypertension, inflammatory bowel disease, mental retardation, mood disorder, obesity, refractive error, infertility, Angelman syndrome, Canavan disease, Coeliac disease, Charcot-Marie-Tooth disease, Cystic fibrosis, Duchenne muscular dystrophy, Haemochromatosis, Haemophilia, Klinefelter's syndrome, Neurofibromatosis, Phenylketonuria, Polycystic kidney disease 1 (PKD1) or 2 (PKD2) Prader-Willi syndrome, Sickle-cell disease, Tay- Sachs disease, Turner syndrome. Further disease states or conditions which may be treated by the disclosed compounds according to the present invention include Alzheimer's disease, Amyotrophic lateral sclerosis (Lou Gehrig's disease), Anorexia nervosa, Anxiety disorder, Atherosclerosis, Attention deficit hyperactivity disorder, Autism, Bipolar disorder, Chronic fatigue syndrome, Chronic obstructive pulmonary disease, Crohn's disease, Coronary heart disease, Dementia, Depression, Diabetes mellitus type 1, Diabetes mellitus type 2, Epilepsy, Guillain-Barre syndrome, Irritable bowel syndrome, Lupus, Metabolic syndrome, Multiple sclerosis, Myocardial infarction, Obesity, Obsessive-compulsive disorder, Panic disorder, Parkinson's disease, Psoriasis, Rheumatoid arthritis, Sarcoidosis, Schizophrenia, Stroke, Thromboangiitis obliterans, Tourette syndrome, Vasculitis. Still additional disease states or conditions which can be treated by the disclosed compounds according to the present invention include aceruloplasminemia, Achondrogenesis type II, achondroplasia, Acrocephaly, Gaucher disease type 2, acute intermittent porphyria, Canavan disease, Adenomatous Polyposis Coli, ALA dehydratase deficiency, adenylosuccinate lyase deficiency, Adrenogenital syndrome, Adrenoleukodystrophy, ALA-D porphyria, ALA dehydratase deficiency, Alkaptonuria, Alexander disease, Alkaptonuric ochronosis, alpha 1- antitrypsin deficiency, alpha-1 proteinase inhibitor, emphysema, amyotrophic lateral sclerosis Alstrom syndrome, Alexander disease, Amelogenesis imperfecta, ALA dehydratase deficiency, Anderson-Fabry disease, androgen insensitivity syndrome, Anemia Angiokeratoma Corporis Diffusum, Angiomatosis retinae (von Hippel-Lindau disease) Apert syndrome, Arachnodactyly (Marfan syndrome), Stickler syndrome, Arthrochalasis multiplex congenital (Ehlers-Danlos syndrome#arthrochalasia type) ataxia telangiectasia, Rett syndrome, primary pulmonary hypertension, Sandhoff disease, neurofibromatosis type II, Beare-Stevenson cutis gyrata syndrome, Mediterranean fever, familial, Benjamin syndrome, beta-thalassemia, Bilateral Acoustic Neurofibromatosis (neurofibromatosis type II), factor V Leiden thrombophilia, Bloch- Sulzberger syndrome (incontinentia pigmenti), Bloom syndrome, X-linked sideroblastic anemia, Bonnevie-Ullrich syndrome (Turner syndrome), Bourneville disease (tuberous sclerosis), prion disease, Birt-Hogg-Dube syndrome, Brittle bone disease (osteogenesis imperfecta), Broad Thumb- Hallux syndrome (Rubinstein-Taybi syndrome), Bronze Diabetes/Bronzed Cirrhosis (hemochromatosis), Bulbospinal muscular atrophy (Kennedy's disease), Burger-Grutz syndrome (lipoprotein lipase deficiency), CGD Chronic granulomatous disorder, Campomelic dysplasia, biotinidase deficiency, Cardiomyopathy (Noonan syndrome), Cri du chat, CAVD (congenital absence of the vas deferens), Caylor cardiofacial syndrome (CBAVD), CEP (congenital erythropoietic porphyria), cystic fibrosis, congenital hypothyroidism, Chondrodystrophy syndrome (achondroplasia), otospondylomegaepiphyseal dysplasia, Lesch-Nyhan syndrome, galactosemia, Ehlers-Danlos syndrome, Thanatophoric dysplasia, Coffin-Lowry syndrome, Cockayne syndrome, (familial adenomatous polyposis), Congenital erythropoietic porphyria, Congenital heart disease, Methemoglobinemia/Congenital methaemoglobinaemia, achondroplasia, X-linked sideroblastic anemia, Connective tissue disease, Conotruncal anomaly face syndrome, Cooley's Anemia (beta-thalassemia), Copper storage disease (Wilson's disease), Copper transport disease (Menkes disease), hereditary coproporphyria, Cowden syndrome, Craniofacial dysarthrosis (Crouzon syndrome), Creutzfeldt-Jakob disease (prion disease), Cockayne syndrome, Cowden syndrome, Curschmann-Batten-Steinert syndrome (myotonic dystrophy), Beare-Stevenson cutis gyrata syndrome, primary hyperoxaluria, spondyloepimetaphyseal dysplasia (Strudwick type), muscular dystrophy, Duchenne and Becker types (DBMD), Usher syndrome, Degenerative nerve diseases including de Grouchy syndrome and Dejerine-Sottas syndrome, developmental disabilities, distal spinal muscular atrophy, type V, androgen insensitivity syndrome, Diffuse Globoid Body Sclerosis (Krabbe disease), Di George's syndrome, receptor deficiency, androgen insensitivity syndrome, Down syndrome, Dwarfism, erythropoietic protoporphyria Erythroid 5-aminolevulinate synthetase deficiency, Erythropoietic porphyria, erythropoietic protoporphyria, erythropoietic uroporphyrin Friedreich's ataxia-familial paroxysmal polyserositis, porphyria cutanea tarda, familial pressure sensitive neuropathy, primary pulmonary hypertension (PPH), Fibrocystic disease of the pancreas, fragile X syndrome, galactosemia, genetic brain disorders, Giant cell hepatitis (Neonatal hemochromatosis), Gronblad-Strandberg syndrome (pseudoxanthoma elasticum), Gunther disease (congenital erythropoietic porphyria), haemochromatosis, Hallgren syndrome, sickle cell anemia, hemophilia, hepatoerythropoietic porphyria (HEP), Hippel-Lindau disease (von Hippel-Lindau disease), Huntington's disease, Hutchinson-Gilford progeria syndrome (progeria), Hyperandrogenism, Hypochondroplasia, Hypochromic anemia, Immune system disorders, including X-linked severe combined immunodeficiency, Insley-Astley syndrome, Jackson-Weiss syndrome, Joubert syndrome, Lesch-Nyhan syndrome, Jackson-Weiss syndrome, Kidney diseases, including hyperoxaluria, Klinefelter's syndrome, Kniest dysplasia, Lacunar dementia, Langer-Saldino achondrogenesis, ataxia telangiectasia, Lynch syndrome, Lysyl-hydroxylase deficiency, Machado-Joseph disease, Metabolic disorders, including Kniest dysplasia, Marfan syndrome, Movement disorders, Mowat-Wilson syndrome, cystic fibrosis, Muenke syndrome, Multiple neurofibromatosis, Nance-Insley syndrome, Nance- Sweeney chondrodysplasia, Niemann-Pick disease, Noack syndrome (Pfeiffer syndrome), Osler-Weber-Rendu disease, Peutz- Jeghers syndrome, Polycystic kidney disease, polyostotic fibrous dysplasia (McCune-Albright syndrome), Peutz-Jeghers syndrome, Prader-Labhart- Willi syndrome, hemochromatosis, primary hyperuricemia syndrome (Lesch-Nyhan syndrome), primary pulmonary hypertension, primary senile degenerative dementia, prion disease, progeria (Hutchinson Gilford Progeria Syndrome), progressive chorea, chronic hereditary (Huntington) (Huntington's disease), progressive muscular atrophy, spinal muscular atrophy, propionic acidemia, protoporphyria, proximal myotonic dystrophy, pulmonary arterial hypertension, PXE (pseudoxanthoma elasticum), Rb (retinoblastoma), Recklinghausen disease (neurofibromatosis type I), Recurrent polyserositis, Retinal disorders, Retinoblastoma, Rett syndrome, RFALS type 3, Ricker syndrome, Riley-Day syndrome, Roussy-Levy syndrome, severe achondroplasia with developmental delay and acanthosis nigricans (SADDAN), Li-Fraumeni syndrome, sarcoma, breast, leukemia, and adrenal gland (SBLA) syndrome, sclerosis tuberose (tuberous sclerosis), SDAT, SED congenital (spondyloepiphyseal dysplasia congenita), SED Strudwick (spondyloepimetaphyseal dysplasia, Strudwick type), SEDc (spondyloepiphyseal dysplasia congenita) SEMD, Strudwick type (spondyloepimetaphyseal dysplasia, Strudwick type), Shprintzen syndrome, Skin pigmentation disorders, Smith-Lemli-Opitz syndrome, South-African genetic porphyria (variegate porphyria), infantile-onset ascending hereditary spastic paralysis, Speech and communication disorders, sphingolipidosis, Tay-Sachs disease, spinocerebellar ataxia, Stickler syndrome, stroke, androgen insensitivity syndrome, tetrahydrobiopterin deficiency, beta-thaiassemia, Thyroid disease, Tomaculous neuropathy (hereditary neuropathy with liability to pressure palsies). The term "neoplasia" or "cancer" is used throughout the specification to refer to the pathological process that results in the formation and growth of a cancerous or malignant neoplasm, i.e., abnormal tissue (solid) or cells (non-solid) that grow by cellular proliferation, often more rapidly than normal and continues to grow after the stimuli that initiated the new growth cease. Malignant neoplasms show partial or complete lack of structural organization and functional coordination with the normal tissue and most invade surrounding tissues, can metastasize to several sites, are likely to recur after attempted removal and may cause the death of the patient unless adequately treated. As used herein, the term neoplasia is used to describe all cancerous disease states and embraces or encompasses the pathological process associated with malignant hematogenous, ascitic and solid tumors. Exemplary cancers which may be treated by the present disclosed compounds either alone or in combination with at least one additional anti-cancer agent include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitt's lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gliobastomas, neuroblastomas, ganglioneuromas, gangliogliomas, medulloblastomas, pineal cell tumors, meningiomas, meningeal sarcomas, neurofibromas, and Schwannomas; bowel cancer, breast cancer, prostate cancer, cervical cancer, uterine cancer, lung cancer, ovarian cancer, testicular cancer, thyroid cancer, astrocytoma, esophageal cancer, pancreatic cancer, stomach cancer, liver cancer, colon cancer, melanoma; carcinosarcoma, Hodgkin's disease, Wilms' tumor and teratocarcinomas. Additional cancers which may be treated using the disclosed compounds according to the present invention include, for example, acute granulocytic leukemia, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, Basal cell carcinoma, B-Cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, breast cancer, triple (estrogen, progesterone and HER-2) negative breast cancer, double negative breast cancer (two of estrogen, progesterone and HER-2 are negative), single negative (one of estrogen, progesterone and HER-2 is negative), estrogen-receptor positive, HER2-negative breast cancer, estrogen receptor-negative breast cancer, estrogen receptor positive breast cancer, metastatic breast cancer, luminal A breast cancer, luminal B breast cancer, Her2-negative breast cancer, HER2-positive or negative breast cancer, progesterone receptor-negative breast cancer, progesterone receptor-positive breast cancer, recurrent breast cancer, carcinoid tumors, cervical cancer, cholangiocarcinoma, chondrosarcoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), colon cancer, colorectal cancer, craniopharyngioma, cutaneous lymphoma, cutaneous melanoma, diffuse astrocytoma, ductal carcinoma in situ (DCIS), , ependymoma, epithelioid sarcoma, esophageal cancer, ewing sarcoma, extrahepatic bile duct cancer, eye cancer, fallopian tube cancer, fibrosarcoma, gallbladder cancer, gastric cancer, gastrointestinal cancer, gastrointestinal carcinoid cancer, gastrointestinal stromal tumors (GIST), germ cell tumor glioblastoma multiforme (GBM), glioma, hairy cell leukemia, head and neck cancer, hemangioendothelioma, Hodgkin lymphoma, hypopharyngeal cancer, infiltrating ductal carcinoma (IDC), infiltrating lobular carcinoma (ILC), inflammatory breast cancer (IBC), intestinal Cancer, intrahepatic bile duct cancer, invasive/infiltrating breast cancer, Islet cell cancer, jaw cancer, Kaposi sarcoma, kidney cancer, laryngeal cancer, leiomyosarcoma, leptomeningeal metastases, leukemia, lip cancer, liposarcoma, liver cancer, lobular carcinoma in situ, low-grade astrocytoma, lung cancer, lymph node cancer, lymphoma, male breast cancer, medullary carcinoma, medulloblastoma, melanoma, meningioma, Merkel cell carcinoma, mesenchymal chondrosarcoma, mesenchymous, mesothelioma metastatic breast cancer, metastatic melanoma metastatic squamous neck cancer, mixed gliomas, monodermal teratoma, mouth cancer mucinous carcinoma, mucosal melanoma, multiple myeloma, Mycosis Fungoides, myelodysplastic syndrome, nasal cavity cancer, nasopharyngeal cancer, neck cancer, neuroblastoma, neuroendocrine tumors ( ETs), non-Hodgkin's lymphoma, non-small cell lung cancer (NSCLC), oat cell cancer, ocular cancer, ocular melanoma, oligodendroglioma, oral cancer, oral cavity cancer, oropharyngeal cancer, osteogenic sarcoma, osteosarcoma, ovarian cancer, ovarian epithelial cancer ovarian germ cell tumor, ovarian primary peritoneal carcinoma, ovarian sex cord stromal tumor, Paget's disease, pancreatic cancer, papillary carcinoma, paranasal sinus cancer, parathyroid cancer, pelvic cancer, penile cancer, peripheral nerve cancer, peritoneal cancer, pharyngeal cancer, pheochromocytoma, pilocytic astrocytoma, pineal region tumor, pineoblastoma, pituitary gland cancer, primary central nervous system (CNS) lymphoma, prostate cancer, rectal cancer, renal cell carcinoma, renal pelvis cancer, rhabdomyosarcoma, salivary gland cancer, soft tissue sarcoma, bone sarcoma, sarcoma, sinus cancer, skin cancer, small cell lung cancer (SCLC), small intestine cancer, spinal cancer, spinal column cancer, spinal cord cancer, squamous cell carcinoma, stomach cancer, synovial sarcoma, T-cell lymphoma, testicular cancer, throat cancer, thymoma/thymic carcinoma, thyroid cancer, tongue cancer, tonsil cancer, transitional cell cancer, tubal cancer, tubular carcinoma, undiagnosed cancer, ureteral cancer, urethral cancer, uterine adenocarcinoma, uterine cancer, uterine sarcoma, vaginal cancer, vulvar cancer, T-cell lineage acute lymphoblastic leukemia (T-ALL), T-cell lineage lymphoblastic lymphoma (T-LL), peripheral T-cell lymphoma, Adult T-cell leukemia, Pre-B ALL, Pre-B lymphomas, large B-cell lymphoma, Burkitts lymphoma, B-cell ALL, Philadelphia chromosome positive ALL, Philadelphia chromosome positive CML, juvenile myelomonocytic leukemia (JMML), acute promyelocytic leukemia (a subtype of AML), large granular lymphocytic leukemia, Adult T-cell chronic leukemia, diffuse large B cell lymphoma, follicular lymphoma; Mucosa-Associated Lymphatic Tissue lymphoma (MALT), small cell lymphocytic lymphoma, mediastinal large B cell lymphoma, nodal marginal zone B cell lymphoma (NMZL); splenic marginal zone lymphoma (SMZL); intravascular large B-cell lymphoma; primary effusion lymphoma; or lymphomatoid granulomatosis;; B-cell prolymphocytic leukemia; splenic lymphoma/1eukemia, unclassifiable, splenic diffuse red pulp small B-cell lymphoma; lymphoplasmacytic lymphoma; heavy chain diseases, for example, Alpha heavy chain disease, Gamma heavy chain disease, Mu heavy chain disease, plasma cell myeloma, solitary plasmacytoma of bone; extraosseous plasmacytoma; primary cutaneous follicle center lymphoma, /histocyte rich large B-cell lymphoma, DLBCL associated with chronic inflammation; Epstein-Barr virus (EBV)+ DLBCL of the elderly; primary mediastinal (thymic) large B-cell lymphoma, primary cutaneous DLBCL, leg type, ALK+ large B-cell lymphoma, plasmablastic lymphoma; large B-cell lymphoma arising in HHV8-associated multicentric, Castleman disease; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma, or B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and classical Hodgkin lymphoma.

IV. COMBINATION THERAPY The disclosed compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent to treat a host such as a human with a disorder as described herein. The disclosed compounds described herein can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent to treat a host such as a human with a disorder as described herein. The term "bioactive agent" is used to describe an agent, other than the selected compound according to the present invention, which can be used in combination or alternation with a compound of the present invention to achieve a desired result of therapy. In one embodiment, the compound of the present invention and the bioactive agent are administered in a manner that they are active in vivo during overlapping time periods, for example, have time-period overlapping Cmax, Tmax, AUC or other pharmacokinetic parameter. In another embodiment, the compound of the present invention and the bioactive agent are administered to a host in need thereof that do not have overlapping pharmacokinetic parameter, however, one has a therapeutic impact on the therapeutic efficacy of the other. In one aspect of this embodiment, the bioactive agent is an immune modulator, including but not limited to a checkpoint inhibitor, including as non-limiting examples, a PD-1 inhibitor,

PD-Ll inhibitor, PD-L2 inhibitor, CTLA-4 inhibitor, LAG-3 inhibitor, TEVI-3 inhibitor, V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, small molecule, peptide, nucleotide, or other inhibitor. In certain aspects, the immune modulator is an , such as a . PD-1 inhibitors that blocks the interaction of PD-1 and PD-Ll by binding to the PD-1 receptor, and in turn inhibit immune suppression include, for example, nivolumab (Opdivo), pembrolizumab (Keytruda), pidilizumab, AMP-224 (AstraZeneca and Medlmmune), PF- 06801591 (Pfizer), MEDI0680 (AstraZeneca), PDR001 (Novartis), REGN2810 (Regeneron), SHR-12-1 (Jiangsu Hengrui Medicine Company and Incyte Corporation), TSR-042 (Tesaro), and the PD-Ll/VISTA inhibitor CA-170 (Curis Inc.). PD-Ll inhibitors that block the interaction of PD-1 and PD-Ll by binding to the PD-Ll receptor, and in turn inhibits immune suppression, include for example, atezolizumab (Tecentriq), durvalumab (AstraZeneca and Medlmmune), KN035 (Alphamab), and BMS-936559 (Bristol-Myers Squibb). CTLA-4 checkpoint inhibitors that bind to CTLA-4 and inhibits immune suppression include, but are not limited to, , (AstraZeneca and Medlmmune), AGEN1884 and AGEN2041 (Agenus). LAG-3 checkpoint inhibitors, include, but are not limited to, BMS-986016 (Bristol-Myers Squibb), GSK2831781 (GlaxoSmithKline), ΓΜΡ321 (Prima BioMed), LAG525 (Novartis), and the dual PD-1 and LAG- 3 inhibitor MGD013 (MacroGenics). An example of a TIM-3 inhibitor is TSR- 022 (Tesaro). In yet another embodiment, one of the active compounds described herein can be administered in an effective amount for the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer, in combination or alternation with an effective amount of an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist or agonist. Partial anti- like raloxifene and retain some estrogen-like effects, including an estrogen-like stimulation of uterine growth, and also, in some cases, an estrogen-like action during breast cancer progression which actually stimulates tumor growth. In contrast, fulvestrant, a complete anti-estrogen, is free of estrogen-like action on the uterus and is effective in tamoxifen- resistant tumors. Non-limiting examples of anti-estrogen compounds are provided in WO 2014/19176 assigned to Astra Zeneca, WO20 13/090921, WO 2014/203129, WO 2014/203132, and US20 13/0 178445 assigned to Olema Pharmaceuticals, and U.S. Patent Nos. 9,078,871, 8,853,423, and 8,703, 810, as well as US 2015/0005286, WO 2014/205136, and WO 2014/205138. Additional non-limiting examples of anti-estrogen compounds include: SERMS such as anordrin, bazedoxifene, broparestriol, chlorotrianisene, clomiphene citrate, cyclofenil, lasofoxifene, ormeloxifene, raloxifene, tamoxifen, toremifene, and fulvestratnt; aromatase inhibitors such as , , anastrozole, , fadrozole, , and letrozole; and such as , , allylestrenol, chloromadinone acetate, acetate, , dydrogesterone, medroxyprogesterone acetate, , acetate, acetate, progesterone, and . Other estrogenic ligands that can be used according to the present invention are described in U.S. Patent Nos. 4,418,068; 5,478,847; 5,393,763; and 5,457,1 17, WO201 1/156518, US Patent Nos. 8,455,534 and 8,299,1 12, U.S. Patent Nos. 9,078,871; 8,853,423; 8,703,810; US 2015/0005286; and WO 2014/205138, US20 16/0 175289,

US2015/0258080, WO 2014/191726, WO 2012/08471 1; WO 2002/013802; WO 2002/004418; WO 2002/003992; WO 2002/003991; WO 2002/003990; WO 2002/003989; WO 2002/003988; WO 2002/003986; WO 2002/003977; WO 2002/003976; WO 2002/003975; WO 2006/078834; US 6821989; US 2002/0128276; US 6777424; US 2002/0016340; US 6326392; US 6756401; US 2002/0013327; US 6512002; US 6632834; US 2001/0056099; US 6583170; US 6479535; WO 1999/024027; US 6005102; EP 0802184; US 5998402; US 5780497, U S 5880137, WO 2012/048058 and WO 2007/087684. In another embodiment, an active compounds described herein can be administered in an effective amount for the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, in combination or alternation with an effective amount of an androgen (such as ) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist. In one embodiment, the prostate or testicular cancer is androgen-resistant. Non-limiting examples of anti-androgen compounds are provided in WO 201 1/156518 and US Patent Nos. 8,455,534 and 8,299,1 12. Additional non- limiting examples of anti-androgen compounds include: , , , , spironolactone, , , , topilutamide, , and . In one embodiment, the bioactive agent is an ALK inhibitor. Examples of ALK inhibitors include but are not limited to , , , TAE684 (NVP-TAE684), GSK1838705A, AZD3463, ASP3026, PF-06463922, (RXDX-101), and AP261 13,. In one embodiment, the bioactive agent is an EGFR inhibitor. Examples of EGFR inhibitors include (Tarceva), (Iressa), (Gilotrif), (CO- 1686), (Tagrisso), (Olita), naquotinib (ASP8273), nazartinib (EGF816), PF- 06747775 (Pfizer), (BPI-2009), (HKI-272; PB272); avitinib (ACOOIO), EAI045, tarloxotinib (TH-4000; PR-610), PF-06459988 (Pfizer), (XL647; EXEL-7647; KD- 019), transtinib, WZ-3146, WZ8040, CNX-2006, and (PF-00299804; Pfizer). In one embodiment, the bioactive agent is an HER-2 inhibitor. Examples of HER-2 inhibitors include , , ado-, and pertuzumab. In one embodiment, the bioactive agent is a CD20 inhibitor. Examples of CD20 inhibitors include , , fatumumab, ibritumomab, , and . In one embodiment, the bioactive agent is a JAK3 inhibitor. Examples of JAK3 inhibitors include tasocitinib. In one embodiment, the bioactive agent is a BCL-2 inhibitor. Examples of BCL-2 inhibitors include , ABT-199 (4-[4-[[2-(4-Chlorophenyl)-4,4-dimethylcyclohex-l-en- l-yl]methyl]piperazin-l-yl]-N-[[3-nitro-4-[[(tetrahydro-2H-pyran-4- yl)methyl]amino]phenyl]sulfonyl]-2-[(lH- pyrrolo[2,3-b]pyridin-5-yl)oxy]benzamide), ABT-737 (4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-l-yl]-N-[4- [[(2R)-4-(dimethylamino)-l- phenylsulfanylbutan-2-yl] amino]-3- nitrophenyljsulfonylbenzamide) (navitoclax), ABT-263 ((R)-4-(4-((4'-chloro-4,4-dimethyl-3,4,5,6-tetrahydro-[l, l'-biphenyl]-2-yl)methyl)piperazin-l-yl)- N-((4-((4-morpholino- 1-(phenylthio)butan-2-yl)amino)- 3((trifluoromethyl)sulfonyl)phenyl)sulfonyl)benzamide), GX1 5-070 (obatoclax mesylate, (2Z)-2- [(5Z)-5-[(3,5- dimethyl-lH-pyrrol-2-yl)methylidene]-4-methoxypyrrol-2-ylidene]indole; methanesulfonic acid))), 2-methoxy-antimycin A3, YC137 (4-(4,9-dioxo-4,9- dihydronaphtho[2,3-d]thiazol-2-ylamino)-phenyl ester), pogosin, ethyl 2-amino-6-bromo-4-(l- cyano-2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate, -d3, TW-37 (N-[4-[[2-(l,l- Dimethylethyl)phenyl]sulfonyl]phenyl]-2,3,4-trihydroxy-5-[[2-(l- methylethyl)phenyl]methyl]benzamide), Apogossypolone (ApoG2), HA14-1, AT101, sabutoclax, gambogic acid, or G3139 (Oblimersen). In one embodiment, the bioactive agent is a kinase inhibitor. In one embodiment, the kinase inhibitor is selected from a phosphoinositide 3-kinase (PI3K) inhibitor, a Bruton's tyrosine kinase (BTK) inhibitor, or a spleen tyrosine kinase (Syk) inhibitor, or a combination thereof. Examples of PI3 kinase inhibitors include but are not limited to Wortmannin, demethoxyviridin, perifosine, idelalisib, Pictilisib , Palomid 529, ZSTK474, PWT33597, CUDC- 907, and AEZS-136, duvelisib, GS-9820, BKM120, GDC-0032 (Taselisib) (2-[4-[2-(2-Isopropyl- 5-methyl- 1,2,4-triazol-3 -yl)-5,6-dihydroimidazo[ 1,2-d][1,4]benzoxazepin-9-yl]pyrazol- 1-yl]-2- methylpropanamide), MLN-1 117 ((2R)-l-Phenoxy-2-butanyl hydrogen (S)-methylphosphonate; or Methyl(oxo) {[(2R)-l-phenoxy-2-butanyl]oxy}phosphonium)), BYL-719 ((2S)-Nl-[4-Methyl- 5-[2-(2,2,2-trifluoro- 1,1-dimethylethyl)-4-pyridinyl]-2-thiazolyl]- 1,2-pyrrolidinedicarboxamide), GSK2126458 (2,4-Difluoro-N-{2-(methyloxy)-5-[4-(4-pyridazinyl)-6-quinolinyl]-3- pyridinyl}benzenesulfonamide) (omipalisib), TGX-221 ((±)-7-Methyl-2-(morpholin-4-yl)-9-(l- phenylaminoethyl)-pyrido[l,2-a]-pyrimidin-4-one), GSK2636771 (2-Methyl-l-(2-methyl-3- (trifluoromethyl)benzyl)-6-mo holino-lH-benzo[d]imidazole-4-carboxylic acid dihydrochloride), KIN- 193 ((R)-2-((l-(7-methyl-2-mo holino-4-oxo-4H-pyrido[l,2-a]pyrimidin- 9-yl)ethyl)amino)benzoic acid), TGR-1202/RP5264, GS-9820 ((S)- l-(4-((2-(2-aminopyrimidin- 5-yl)-7-methyl-4-mohydroxypropan- 1 -one), GS-1 101 (5-fluoro-3-phenyl-2-([S)]-l-[9H-purin-6- ylamino]-propyl)-3H-quinazolin-4-one), AMG-319, GSK-2269557, SAR245409 (N-(4-(N-(3- ((3,5-dimethoxyphenyl)amino)quinoxalin-2-yl)sulfamoyl)phenyl)-3-methoxy-4 methylbenzamide), BAY80-6946 (2-amino-N-(7-methoxy-8-(3-morpholinopropoxy)-2,3- dihydroimidazo[l,2-c]quinaz), AS 252424 (5-[l-[5-(4-Fluoro-2-hydroxy-phenyl)-furan-2-yl]- meth-(Z)-ylidene]-thiazolidine-2,4-dione), CZ 24832 (5-(2-amino-8-fluoro-[l,2,4]triazolo[l,5- a]pyridin-6-yl)-N-tert-butylpyridine-3-sulfonamide), Buparlisib pyrimidinyl]-4-(trifluoromethyl)-2-pyridinamine), GDC-0941 (2-(lH-Indazol-4-yl)-6-[[4- (methylsulfonyl)-l-piperazinyl]methyl]-4-(4-mo holinyl)thieno[3,2-d]pyrimidine), GDC-0980 ((S)-l-(4-((2-(2-aminopyrimidin-5-yl)-7-methyl-4-morpholinothieno[3,2-d]pyrimidin-6 yl)methyl)piperazin-l-yl)-2-hydroxypropan-l-one (also known as RG7422)), SF1 126 ((8S,14S,17S)-14-(carboxymethyl)-8-(3-guanidinopropyl)-17-(hydroxymethyl)-3,6,9,12,15- pentaoxo- 1-(4-(4-oxo-8-phenyl-4H-chromen-2-yl)mo holino-4-ium)-2-oxa-7, 10,13,16- tetraazaoctadecan-18-oate), PF-05212384 (N-[4-[[4-(Dimethylamino)-l- piperidinyl]carbonyl]phenyl]-N'-[4-(4,6-di-4-morpholinyl-l,3,5-triazin-2-yl)phenyl]urea) (gedatolisib), LY3023414, BEZ235 (2-Methyl-2-{4-[3-methyl-2-oxo-8-(quinolin-3-yl)-2,3- dihydro-lH-imidazo[4,5-c]quinolin-l-yl]phenyl}propanenitrile) (dactolisib), XL-765 (N-(3-(N-(3- (3,5-dimethoxyphenylamino)quinoxalin-2-yl)sulfamoyl)phenyl)-3-methoxy-4- methylbenzamide), and GSK1059615 (5-[[4-(4-Pyridinyl)-6-quinolinyl]methylene]-2,4- thiazolidenedione), PX886 ([(3aR,6E,9S,9aR,10R,l laS)-6-[[bis(prop-2- enyl)amino]methylidene]-5-hydroxy-9-(methoxymethyl)-9a, 1la-dimethyl-l,4,7-trioxo- 2,3,3a,9,10,ll-hexahydroindeno[4,5h]isochromen- 10-yl] acetate (also known as sonolisib)), LY294002, AZD8186, PF-4989216, pilaralisib, GNE-317, PI-3065, PI-103, NU7441 (KU- 57788), HS 173, VS-5584 (SB2343), CZC24832, TGI 00-1 15, A66, YM201636, CAY10505, PIK- 75, PIK-93, AS-605240, BGT226 (NVP-BGT226), AZD6482, voxtalisib, alpelisib, IC-871 14, TGI100713, CH5 132799, PKI-402, copanlisib (BAY 80-6946), XL 147, PIK-90, PIK-293, PIK- 294, 3-MA (3-methyladenine), AS-252424, AS-604850, apitolisib (GDC-0980; RG7422), and the structure described in WO2014/071 109 having the formula: Compound 292

Examples of BTK inhibitors include (also known as PCI-32765)(Imbruvica™)(l- [(3R)-3-[4-amino-3-(4-phenoxy-phenyl)pyrazolo[3,4-d]pyrimidin-l-yl]piperidin-l-yl]prop-2-en-

1-one), dianilinopyrimidine-based inhibitors such as AVL-101 and AVL-291/292 (N-(3-((5- fluoro-2-((4-(2-methoxyethoxy)phenyl)amino)pyrimidin-4-yl)amino)phenyl)acrylamide) (Avila Therapeutics) (see US Patent Publication No 201 1/01 17073, incorporated herein in its entirety), ([N-(2-chloro-6-methylphenyl)-2-(6-(4-(2-hydroxyethyl)piperazin- 1-yl)-2- methylpyrimidin-4-ylamino)thiazole-5-carboxamide], LFM-A13 (alpha-cyano-beta-hydroxy- beta-methyl-N-(2,5-ibromophenyl) propenamide), GDC-0834 ([R-N-(3-(6-(4-(l,4-dimethyl-3- oxopiperazin-2-yl)phenylamino)-4-methyl-5-oxo-4,5-dihydropyrazin-2-yl)-2-methylphenyl)- 4,5,6,7-tetrahydrobenzo[b]thiophene-2-carboxamide], CGI-560 4-(tert-butyl)-N-(3-(8- (phenylamino)imidazo[ 1,2-a]pyrazin-6-yl)phenyl)benzamide, CGI- 1746 (4-(tert-butyl)-N-(2- methyl-3-(4-methyl-6-((4-(mo holine-4-carbonyl)phenyl)amino)-5-oxo-4,5-dihydropyrazin-2- yl)phenyl)benzamide), CNX-774 (4-(4-((4-((3-acrylamidophenyl)amino)-5-fluoropyrimidin-2- yl)amino)phenoxy)-N-methylpicolinamide), CTA056 (7-benzyl-l-(3-(piperidin-l-yl)propyl)-2- (4-(pyridin-4-yl)phenyl)-lH-imidazo[4,5-g]quinoxalin-6(5H)-one), GDC-0834 ((R)-N-(3-(6-((4- (l,4-dimethyl-3-oxopiperazin-2-yl)phenyl)amino)-4-methyl-5-oxo-4,5-dihydropyrazin-2-yl)-2- methylphenyl)-4,5,6,7-tetrahydrobenzo[b]thiophene-2-carboxamide), GDC-0837 ((R)-N-(3-(6- ((4-(l,4-dimethyl-3-oxopiperazin-2-yl)phenyl)amino)-4-methyl-5-oxo-4,5-dihydropyrazin-2-yl)- 2-methylphenyl)-4,5,6,7-tetrahydrobenzo[b]thiophene-2-carboxamide), HM-71224, ACP-196, ONO-4059 (Ono Pharmaceuticals), PRT062607 (4-((3-(2H-l,2,3-triazol-2-yl)phenyl)amino)-2- (((lR,2S)-2-aminocyclohexyl)amino)pyrimidine-5-carboxamide hydrochloride), QL-47 (1-(1- acryloylindolin-6-yl)-9-(l-methyl-lH-pyrazol-4-yl)benzo[h][l,6]naphthyridin-2(lH)-one), and RN486 (6-cyclopropyl-8-fluoro-2-(2-hydroxymethyl-3-{l-methyl-5-[5-(4-methyl-piperazin-l- yl)-pyridin-2-ylamino]-6-oxo-l,6-dihydro-pyridin-3-yl}-phenyl)-2H-isoquinolin-l-one), and other molecules capable of inhibiting BTK activity, for example those BTK inhibitors disclosed in Akinleye et ah, Journal of Hematology & Oncology, 2013, 6:59, the entirety of which is incorporated herein by reference. Syk inhibitors include, for example, Cerdulatinib (4-(cyclopropylamino)-2-((4-(4- (ethylsulfonyl)piperazin-l-yl)phenyl)amino)pyrimidine-5-carboxamide), entospletinib (6-(lH- indazol-6-yl)-N-(4-mo holinophenyl)imidazo[l,2-a]pyrazin-8-amine), fostamatinib ([6-({5- Fluoro-2-[(3,4,5-trimethoxyphenyl)amino]-4-pyrimidinyl}amino)-2,2-dimethyl-3-oxo-2,3- dihydro-4H-pyrido[3,2-b][l,4]oxazin-4-yl]methyl dihydrogen phosphate), fostamatinib disodium salt (sodium (6-((5-fluoro-2-((3,4,5-trimethoxyphenyl)amino)pyrimidin-4-yl)amino)-2,2- dimethyl-3-oxo-2H-pyrido[3,2-b][l,4]oxazin-4(3H)-yl)methyl phosphate), BAY 61-3606 (2-(7- (3,4-Dimethoxyphenyl)-imidazo[l,2-c]pyrimidin-5-ylamino)-nicotinamide HC1), RO9021 (6- [(lR,2S)-2-Amino-cyclohexylamino]-4-(5,6-dimethyl-pyridin-2-ylamino)-pyridazine-3- carboxylic acid amide), (Gleevac; 4-[(4-methylpiperazin-l-yl)methyl]-N-(4-methyl-3- {[4-(pyridin-3-yl)pyrimidin-2-yl]amino}phenyl)benzamide), staurosporine, GSK143 (2- (((3R,4R)-3-aminotetrahydro-2H-pyran-4-yl)amino)-4-(p-tolylamino)pyrimidine-5- carboxamide), PP2 (l-(tert-butyl)-3-(4-chlorophenyl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine), PRT-0603 18 (2-(((lR,2S)-2-aminocyclohexyl)amino)-4-(m-tolylamino)pyrimidine-5- carboxamide), PRT-062607 (4-((3-(2H-l,2,3-triazol-2-yl)phenyl)amino)-2-(((lR,2S)-2- aminocyclohexyl)amino)pyrimidine-5-carboxamide hydrochloride), R 112 (3,3'-((5- fluoropyrimidine-2,4-diyl)bis(azanediyl))diphenol), R348 (3-Ethyl-4-methylpyridine), R406 (6- ((5-fluoro-2-((3,4,5-trimethoxyphenyl)amino)pyrimidin-4-yl)amino)-2,2-dimethyl-2H- pyrido[3,2-b][l,4]oxazin-3(4H)-one), piceatannol (3-Hydroxyresveratol), YM193306(see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643), 7-azaindole, piceatannol, ER-27319 (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), Compound D (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), PRT060318 (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), luteolin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), apigenin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), quercetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), fisetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), myricetin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein), morin (see Singh et al. Discovery and Development of Spleen Tyrosine Kinase (SYK) Inhibitors, J . Med. Chem. 2012, 55, 3614-3643 incorporated in its entirety herein). In one embodiment, the bioactive agent is a MEK inhibitor. MEK inhibitors are well known, and include, for example, /GSK1120212 (N-(3-{3-Cyclopropyl-5-[(2-fluoro-4- iodophenyl)amino]-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin-l(2H- yl}phenyl)acetamide), (6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2 -hydroxyethoxy)- 3-methylbenzimidazole-5-carboxamide), pimasertib/AS703026/MSC 1935369 ((S)-N-(2,3- dihydroxypropyl)-3-((2-fluoro-4- iodophenyl)amino)isonicotinamide), XL-518/GDC-0973 (1- ({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2- yl]azetidin-3-ol), refametinib/BAY869766/RDEAl 19 (N-(3,4-difluoro-2-(2-fluoro-4- iodophenylamino)-6-methoxyphenyl)-l -(2,3-dihydroxypropyl)cyclopropane-l -sulfonamide), PD-0325901 (N-[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]- benzamide), TAK733 ((R)-3-(2,3-Dihydroxypropyl)-6-fluoro-5-(2-fluoro-4-iodophenylamino)-8- methylpyrido[2,3-d]pyrimidine-4,7(3H,8H)-dione), MEK162/ARRY438162 (5-[(4-Bromo-2- fluorophenyl)amino]-4-fluoro-N-(2- hydroxyethoxy)-l -methyl- lH-benzimidazole-6- carboxamide), R05 126766 (3-[[3-Fluoro-2- (methylsulfamoylamino)-4-pyridyl]methyl]-4- methyl-7-pyrimidin-2-yloxychromen-2-one), WX-554, R04987655/CH4987655 (3,4-difluoro-2- ((2-fluoro-4-iodophenyl)amino)-N-(2 -hydroxyethoxy)-5-((3-oxo-l,2-oxazinan- 2yl)methyl)benzamide), or AZD8330 (2-((2-fluoro-4-iodophenyl)amino)-N-(2 hydroxyethoxy)-l ,5-dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide), U0126-EtOH, PD184352 (CI-1040), GDC-0623, BI-847325, , PD98059, BIX 02189, BIX 02188, , SL-327, TAK-733, PD3 18088. In one embodiment, the bioactive agent is a Raf inhibitor. Raf inhibitors are known and include, for example, Vemurafinib (N-[3-[[5-(4-Chlorophenyl)-lH-pyrrolo[2,3-b]pyridin-3- yl]carbonyl]-2,4-difluorophenyl]-l-propanesulfonamide), tosylate (4-[4-[[4-chloro-3- (trifluoromethyl)phenyl]carbamoylamino]phenoxy]-N-methylpyridine-2-carboxamide;4- methylbenzenesulfonate), AZ628 (3-(2-cyanopropan-2-yl)-N-(4-methyl-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)benzamide), NVP-BHG712 (4-methyl-3-(l-methyl-6- (pyridin-3-yl)-lH-pyrazolo[3,4-d]pyrimidin-4-ylamino)-N-(3- (trifluoromethyl)phenyl)benzamide), RAF-265 (l-methyl-5-[2-[5-(trifluoromethyl)-lH-imidazol- 2-yl]pyridin-4-yl]oxy-N-[4-(trifluoromethyl)phenyl]benzimidazol-2-amine), 2-Bromoaldisine (2-Bromo-6,7-dihydro-lH,5H-pyrrolo[2,3-c]azepine-4,8-dione), Raf Kinase Inhibitor IV (2- chloro-5-(2-phenyl-5-(pyridin-4-yl)-lH-imidazol-4-yl)phenol), Sorafenib N-Oxide (4-[4-[[[[4- Chloro-3(trifluoroMethyl)phenyl]aMino]carbonyl]aMino]phenoxy]-N-Methyl- 2pyridinecarboxaMide 1-Oxide), PLX-4720, (GSK21 18436), GDC-0879, RAF265, AZ 628, SB590885, ZM336372, GW5074, TAK-632, CEP-32496, LY3009120, and GX818 (). In one embodiment, the bioactive agent is an AKT inhibitor, including but not limited to, MK-2206, GSK690693, Perifosine, (KRX-0401), GDC-0068, Triciribine, AZD5363, Honokiol, PF-04691502, and Miltefosine, a FLT-3 inhibitor, including but not limited to, P406, Dovitinib, Quizartinib (AC220), Amuvatinib (MP-470), Tandutinib (MLN518), ENMD-2076, and KW- 2449, or a combination thereof. In one embodiment, the bioactive agent is an mTOR inhibitor. Examples of mTOR inhibitors include but are not limited to rapamycin and its analogs, (Afinitor), , , , and deforolimus. Examples of MEK inhibitors include but are not limited to tametinib/GSK1120212 (N-(3-{3-Cyclopropyl-5-[(2-fluoro-4- iodophenyl)amino]-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin-l(2H- yl}phenyl)acetamide), selumetinob (6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2 -hydroxyethoxy)- 3-methylbenzimidazole-5-carboxamide), pimasertib/AS703026/MSC1935369 ((S)-N-(2,3- dihydroxypropyl)-3-((2-fluoro-4-iodophenyl)amino)isonicotinamide), XL-518/GDC-0973 (1- ({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2- yl]azetidin-3-ol) (cobimetinib), refametinib/BAY869766/RDEA119 (N-(3,4-difluoro-2-(2-fluoro- 4-iodophenylamino)-6-methoxyphenyl)-l-(2,3-dihydroxypropyl)cyclopropane-l-sulfonamide), PD-0325901 (N-[(2R)-2,3-Dihydroxypropoxy]-3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]- benzamide), TAK733 ((R)-3-(2,3-Dihydroxypropyl)-6-fluoro-5-(2-fluoro-4-iodophenylamino)-8- methylpyrido[2,3d]pyrimidine-4,7(3H,8H)-dione), MEK162/ARRY438162 (5-[(4-Bromo-2- fluorophenyl)amino]-4-fluoro-N-(2-hydroxyethoxy)- 1-methyl- lH-benzimidazole-6 carboxamide), R05 126766 (3-[[3-Fluoro-2-(methylsulfamoylamino)-4-pyridyl]methyl]-4- methyl-7-pyrimidin-2-yloxychromen-2-one), WX-554, R04987655/CH4987655 (3,4-difluoro-2- ((2-fluoro-4-iodophenyl)amino)-N-(2 -hydroxy ethoxy)-5-((3-oxo-l,2-oxazinan-2 yl)methyl)benzamide), or AZD8330 (2-((2-fluoro-4-iodophenyl)amino)-N-(2-hydroxyethoxy)- l,5-dimethyl-6-oxo-l,6-dihydropyridine-3-carboxamide). In one embodiment, the bioactive agent is a RAS inhibitor. Examples of RAS inhibitors include but are not limited to Reolysin and siG12D LODER. In one embodiment, the bioactive agent is a HSP inhibitor. HSP inhibitors include but are not limited to Geldanamycin or 17-N-Allylamino-17-demethoxygeldanamycin (17AAG), and Radicicol. Additional bioactive compounds include, for example, everolimus, trabectedin, abraxane, TLK 286, AV-299, DN-101, , GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY- 142886), AMN-107, TKI-258, GSK461364, AZD 1152, enzastaurin, , ARQ-197, MK- 0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, a VEGFR inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, an HDAC inhbitor, a c-MET inhibitor, a PARP inhibitor, a Cdk inhibitor, an IGFR-TK inhibitor, an anti-HGF antibody, a focal adhesion kinase inhibitor, a Map kinase kinase (mek) inhibitor, a VEGF trap antibody, pemetrexed, , amrubicin, oregovomab, Lep-etu, nolatrexed, azd2171, batabulin, , , edotecarin, tetrandrine, rubitecan, tesmilifene, oblimersen, ticilimumab, ipilimumab, gossypol, Bio 111, 131- I-TM-601, ALT-1 10, BIO 140, CC 8490, cilengitide, gimatecan, IL13-PE38QQR, INO 1001, IPdRi KRX-0402, lucanthone, LY3 17615, neuradiab, vitespan, Rta 744, Sdx 102, talampanel, , Xr 3 11, romidepsin, ADS-100380, , 5-fluorouracil, vorinostat, etoposide, gemcitabine, doxorubicin, liposomal doxorubicin, 5'-deoxy-5-fluorouridine, vincristine, temozolomide, ZK-304709, seliciclib; PD0325901, AZD-6244, capecitabine, L-Glutamic acid, N- [4-[2-(2-amino-4,7-dihydro-4-oxo-lH-pyrrolo[2,3-d]pyrimidin-5-yl)ethyl]benzoyl]-, disodium salt, heptahydrate, camptothecin, PEG-labeled irinotecan, tamoxifen, toremifene citrate, anastrazole, exemestane, letrozole, DES(), , estrogen, conjugated estrogen, , IMC-1C1 1, CHIR-258); 3-[5-(methylsulfonylpiperadinemethyl)-indolyl- quinolone, vatalanib, AG-013736, AVE-0005, goserelin acetate, leuprolide acetate, triptorelin pamoate, medroxyprogesterone acetate, hydroxyprogesterone caproate, megestrol acetate, raloxifene, , , , megestrol acetate, CP-724714; TAK-165, HKI- 272, erlotinib, lapatanib, canertinib, ABX-EGF antibody, erbitux, EKB-569, PKI-166, GW- 572016, Ionafarnib, BMS-214662, tipifarnib; amifostine, NVP-LAQ824, suberoyl analide hydroxamic acid, valproic acid, trichostatin A, FK-228, SU1 1248, sorafenib, KRN951, aminoglutethimide, arnsacrine, anagrelide, L-asparaginase, Bacillus Calmette-Guerin (BCG) vaccine, adriamycin, bleomycin, buserelin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, , clodronate, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, diethylstilbestrol, epirubicin, fludarabine, fludrocortisone, , flutamide, gleevec, gemcitabine, hydroxyurea, idarubicin, ifosfamide, imatinib, leuprolide, levamisole, lomustine, mechlorethamine, melphalan, 6-mercaptopurine, mesna, , mitomycin, mitotane, mitoxantrone, nilutamide, octreotide, oxaliplatin, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, teniposide, testosterone, thalidomide, thioguanine, thiotepa, tretinoin, vindesine, 13-cis-retinoic acid, phenylalanine mustard, uracil mustard, , altretamine, floxuridine, 5-deooxyuridine, cytosine arabinoside, 6-mecaptopurine, deoxycoformycin, calcitriol, valrubicin, mithramycin, vinblastine, vinorelbine, topotecan, razoxin, marimastat, COL-3, neovastat, BMS-275291, squalamine, endostatin, SU5416, SU6668, EMD121974, interleukin-12, EVI862, angiostatin, vitaxin, droloxifene, idoxyfene, spironolactone, , cimitidine, trastuzumab, , gefitinib, bortezimib, paclitaxel, cremophor-free paclitaxel, docetaxel, epithiloneB, BMS-247550, BMS-3 10705, droloxifene, 4-hydroxytamoxifen, pipendoxifene, ERA-923, arzoxifene, fulvestrant, acolbifene, lasofoxifene, idoxifene, TSE-424, HMR-3339, ZK 1866 19, topotecan, PTK787/ZK 222584, VX-745, PD 184352, rapamycin, 40-O-(2-hydroxyethyl)-rapamycin, temsirolimus, AP-23573, RADOOl, ABT-578, BC-210, LY294002, LY292223, LY292696, LY293684, LY293646, wortmannin, ZM336372, L-779,450, PEG-filgrastim, darbepoetin, erythropoietin, granulocyte colony-stimulating factor, zolendronate, prednisone, , granulocyte macrophage colony-stimulating factor, histrelin, pegylated alfa-2a, interferon alfa-2a, pegylated interferon alfa-2b, interferon alfa-2b, azacitidine, PEG-L- asparaginase, lenalidomide, gemtuzumab, , interleukin-1 1, dexrazoxane, , all-transretinoic acid, ketoconazole, interleukin-2, megestrol, immune globulin, nitrogen mustard, methylprednisolone, ibritgumomab tiuxetan, , decitabine, hexamethylmelamine, bexarotene, tositumomab, arsenic trioxide, cortisone, editronate, mitotane, cyclosporine, liposomal daunorubicin, Edwina-asparaginase, 89, casopitant, netupitant, an K-1 , palonosetron, aprepitant, diphenhydramine, hydroxyzine, , lorazepam, alprazolam, , droperidol, dronabinol, dexamethasone, methylprednisolone, prochlorperazine, granisetron, ondansetron, dolasetron, tropisetron, pegfilgrastim, erythropoietin, epoetin alfa, darbepoetin alfa and mixtures thereof. In one embodiment, the bioactive agent is selected from, but are not limited to, Imatinib mesylate (Gleevac®), Dasatinib (Sprycel®), Nilotinib (Tasigna®), (Bosulif®), Trastuzumab (Herceptin®), trastuzumab-DMl, Pertuzumab (PerjetaTM), Lapatinib (Tykerb®), Gefitinib (Iressa®), Erlotinib (Tarceva®), Cetuximab (Erbitux®), Panitumumab (Vectibix®), Vandetanib (Caprelsa®), (Zelboraf®), Vorinostat (Zolinza®), Romidepsin (Istodax®), Bexarotene (Tagretin®), (Panretin®), Tretinoin (Vesanoid®), Carfilizomib (KyprolisTM), Pralatrexate (Folotyn®), Bevacizumab (Avastin®), Ziv- (Zaltrap®), Sorafenib (Nexavar®), Sunitinib (Sutent®), Pazopanib (Votrient®), (Stivarga®), and (CometriqTM). In certain aspects, the bioactive agent is an anti-inflammatory agent, a chemotherapeutic agent, a radiotherapeutic, an additional therapeutic agent, or an immunosuppressive agent. Suitable chemotherapeutic bioactive agents include, but are not limited to, a radioactive molecule, a toxin, also referred to as cytotoxin or cytotoxic agent, which includes any agent that is detrimental to the viability of cells, and liposomes or other vesicles containing chemotherapeutic compounds. General anticancer pharmaceutical agents include: Vincristine (Oncovin®) or liposomal vincristine (Marqibo®), Daunorubicin (daunomycin or Cerubidine®) or doxorubicin (Adriamycin®), Cytarabine (cytosine arabinoside, ara-C, or Cytosar®), L-asparaginase (Elspar®) or PEG-L-asparaginase (pegaspargase or Oncaspar®), Etoposide (VP- 16), Teniposide (Vumon®), 6-mercaptopurine (6-MP or Purinethol®), Methotrexate, Cyclophosphamide (Cytoxan®), Prednisone, Dexamethasone (Decadron), imatinib (Gleevec®), dasatinib (Sprycel®), nilotinib (Tasigna®), bosutinib (Bosulif®), and (Iclusig™). Examples of additional suitable chemotherapeutic agents include but are not limited to 1-dehydrotestosterone, 5-fluorouracil decarbazine, 6-mercaptopurine, 6-thioguanine, actinomycin D, adriamycin, aldesleukin, an alkylating agent, allopurinol sodium, altretamine, amifostine, anastrozole, anthramycin (AMC)), an anti-mitotic agent, cis-dichlorodiamine platinum (II) (DDP) cisplatin), diamino dichloro platinum, anthracycline, an antibiotic, an , asparaginase, BCG live (intravesical), betamethasone sodium phosphate and betamethasone acetate, bicalutamide, bleomycin sulfate, busulfan, leucouorin, calicheamicin, capecitabine, carboplatin, lomustine (CCNU), carmustine (BSNU), Chlorambucil, Cisplatin, Cladribine, Colchicin, conjugated estrogens, Cyclophosphamide, Cyclothosphamide, Cytarabine, Cytarabine, cytochalasin B, Cytoxan, Dacarbazine, Dactinomycin, dactinomycin (formerly actinomycin), daunirubicin HCL, daunorucbicin citrate, denileukin diftitox, Dexrazoxane, Dibromomannitol, dihydroxy anthracin dione, Docetaxel, dolasetron mesylate, doxorubicin HCL, dronabinol, E. coli L-asparaginase, emetine, epoetin-a, Erwinia L-asparaginase, esterified estrogens, estradiol, sodium, ethidium bromide, ethinyl estradiol, etidronate, etoposide citrororum factor, etoposide phosphate, filgrastim, floxuridine, fluconazole, fludarabine phosphate, fluorouracil, flutamide, folinic acid, gemcitabine HCL, glucocorticoids, goserelin acetate, gramicidin D, granisetron HCL, hydroxyurea, idarubicin HCL, ifosfamide, interferon a-2b, irinotecan HCL, letrozole, leucovorin calcium, leuprolide acetate, levamisole HCL, lidocaine, lomustine, maytansinoid, mechlorethamine HCL, medroxyprogesterone acetate, megestrol acetate, melphalan HCL, mercaptipurine, mesna, methotrexate, , mithramycin, mitomycin C, mitotane, mitoxantrone, nilutamide, octreotide acetate, ondansetron HCL, paclitaxel, pamidronate disodium, pentostatin, pilocarpine HCL, plimycin, polifeprosan 20 with carmustine implant, porfimer sodium, procaine, procarbazine HCL, propranolol, rituximab, sargramostim, streptozotocin, tamoxifen, taxol, teniposide, tenoposide, testolactone, tetracaine, thioepa chlorambucil, thioguanine, thiotepa, topotecan HCL, toremifene citrate, trastuzumab, tretinoin, valrubicin, vinblastine sulfate, vincristine sulfate, and vinorelbine tartrate. Additional therapeutic agents that can be administered in combination with a degronimer disclosed herein can include bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, finasunate, vatalanib, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, dovitinib, figitumumab, atacicept, rituximab, alemtuzumab, aldesleukine, atlizumab, , temsirolimus, everolimus, lucatumumab, dacetuzumab, HLLl, huN901-DMl, atiprimod, , bortezomib, carfilzomib, marizomib, tanespimycin, saquinavir mesylate, ritonavir, nelfinavir mesylate, indinavir sulfate, belinostat, panobinostat, mapatumumab, lexatumumab, dulanermin, ABT-737, oblimersen, plitidepsin, talmapimod, P276-00, enzastaurin, tipifarnib, perifosine, imatinib, dasatinib, lenalidomide, thalidomide, simvastatin, celecoxib, bazedoxifene, AZD4547, rilotumumab, oxaliplatin (Eloxatin), PD0332991, ribociclib (LEE01 1), amebaciclib (LY2835219), HDM201, fulvestrant (Faslodex), exemestane (Aromasin), PIM447, (INC424), BGJ398, necitumumab, pemetrexed (Alimta), and ramucirumab (IMC-1 121B). In one aspect of the invention, the disclosed compound is administered in combination with an anti-infective agent, for example but not limited to an anti-HIV agent, anti-HCV agent, anti- HBV agent, or other anti-viral or anti-bacterial agent. In one embodiment, the anti-HIV agent can be, but is not limited to, for example, a nucleoside reverse transcriptase inhibitor ( RTI), other non-nucloeoside reverse transcriptase inhibitor, protease inhibitor, fusion inhibitor, among others. Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTIs) include, but are not limited to, Abacavir or ABC (Ziagen), Didanosine or ddl (Videx), Emtricitabine or FTC (Emtriva), Lamivudine or 3TC (Epivir), ddC (zalcitabine), Stavudine or d4T (Zerit), Tenofovircor TDF (Viread), D-D4FC (Reverset), and Zidovudine or AZT or ZDV (Retrovir). Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs) include, but are not limited to, Delavirdine (Rescriptor), Efavirenz (Sustiva), Etravirine (Intelence), Nevirapine (Viramune), and Rilpivirine (Edurant). Anti-HIV Protease Inhibitors (Pis) include, but are not limited to, Atazanavir or ATV (Reyataz), Darunavir or DRV (Prezista), Fosamprenavir or FPV (Lexiva), Indinavir or IDV (Crixivan), Lopinavir + ritonavir, or LPV/r (Kaletra), Nelfinavir or NFV (Viracept), Ritonavir or RTV (Norvir), Saquinavir or SQV (Invirase), Tipranavir, or TPV (Aptivus), Cobicistat (Tybost), Atazanavir + cobicistat, or ATV/COBI (Evotaz), Darunavir + cobicistat, or DRV/COBI (Prezcobix). Anti-HIV Fusion Inhibitors include, but are not limited to, Enfuvirtide or ENF or T- 20 (Fuzeon). Anti-HIV also include, but are not limited to, Maraviroc or MVC (Selzentry). Anti- HIV Integrase Inhibitors include, but are not limited to Dolutegravir (Tivicay), Elvitegravir (Vitekta), Raltegravir (Isentress). Anti-HIV combinations agents include Abacavir + Dolutegravir + lamivudine,or ABC/DTG/3TC (Triumeq), Abacavir +lamivudine or ABC/3TC (Epzicom), Abacavir + lamivudine + zidovudine, or ABC/3TC/ZDV (Trizivir), Efavirenz + emtricitabine + tenofovir or EFV/FTC/TDF (Atripla, Tribuss), elvitegravir, cobicistat, emtricitabine, tenofovir alafenamide or EVG/COBI/FTC/TAF or ECF/TAF (Genvoya; (Stribild), emtricitabine + rilpivirine + tenofovir or FTC/RPV/TAF (Odefsey); Emtricitabine + rilpivirine + tenofovir or FTC/RPV/TDF (Complera), Emtricitabine + tenofovir or TAF/FTC (Descovy), emtricitabine and tenofovir disoproxil fumarate (Truvada), and Lamivudine + zidovudine or 3TC/ZDV (Combivir). Other anti-HIV compounds include, but are not limited to Racivir, L-FddC, L-FD4C, SQVM (Saquinavir mesylate), IDV (Indinavir), SQV (Saquinavir), APV (Amprenavir), LPV (Lopinavir), fusion inhibitors such as T20, among others, fuseon and mixtures thereof, including anti-HIV compounds presently in clinical trials or in development. Other anti-HIV agents which may be used in co-administration with the disclosed compounds according to the present invention. NNRTIs may be selected from the group consisting of nevirapine (BI-R6-587), delavirdine (U-90152S/T), efavirenz (DMP-266), UC-781 (N-[4- chloro-3-(3-methyl-2-butenyloxy)phenyl]-2methyl3-furancarbothiamide), etravirine (TMC125), Trovirdine (Ly300046.HCl), HI-236, HI-240, HI-280, HI-281, rilpivirine (TMC-278), MSC-127, HBY 097, DMP266, Baicalin (TJN-151) ADAM-II (Methyl 3',3'-dichloro-4',4"-dimethoxy-5',5"- bis(methoxycarbonyl)-6,6-diphenylhexenoate), Methyl 3-Bromo-5-(l-5-bromo-4-methoxy-3- (methoxycarbonyl)phenyl)hept- 1-enyl)-2-methoxybenzoate (Alkenyldiarylmethane analog, Adam analog), (5-chloro-3-(phenylsulfinyl)-2'-indolecarboxamide), AAP-BHAP (U-104489 or PNU-104489), Capravirine (AG-1549, S-1 153), atevirdine (U-87201E), aurin tricarboxylic acid

(SD-095345), l-[(6-cyano-2-indolyl)carbonyl]-4-[3-(isopropylamino)-2-pyridinyl]piperazine, 1- [5-[[N-(methyl)methylsulfonylamino]-2-indolylcarbonyl-4-[3-(isopropylamino)-2- pyridinyljpiperazine, l-[3-(Ethylamino)-2-[pyridinyl]-4-[(5-hydroxy-2- indolyl)carbonyl]piperazine, l-[(6-Formyl-2-indolyl)carbonyl]-4-[3-(isopropylamino)-2- pyridinyljpiperazine, l-[[5-(Methylsulfonyloxy)-2-indoyly)carbonyl]-4-[3-(isopropylamino)-2- pyridinyl]piperazine, U88204E, Bis(2-nitrophenyl)sulfone (NSC 633001), Calanolide A (NSC675451), Calanolide B, 6-Benzyl-5-methyl-2-(cyclohexyloxy)pyrimidin-4-one (DABO- 546), DPC 961, E-EBU, E-EBU-dm, E-EPSeU, E-EPU, Foscarnet (Foscavir), HEPT (l-[(2-

Hydroxyethoxy)methyl]-6-(phenylthio)thymine), HEPT-M ( 1-[(2-Hydroxyethoxy)methyl]-6-(3 - methylphenyl)thio)thymine), HEPT-S(1 -[(2-Hydroxyethoxy)methyl]-6-(phenylthio)-2- thiothymine), Inophyllum P, L-737,126, Michellamine A (NSC650898), Michellamine B (NSC649324), Michellamine F, 6-(3,5-Dimethylbenzyl)-l-[(2-hydroxyethoxy)methyl]-5- isopropyluracil, 6-(3,5-Dimethylbenzyl)-l-(ethyoxymethyl)-5-isopropyluracil, NPPS, E-BPTU (NSC 648400), Oltipraz (4-Methyl-5-(pyrazinyl)-3H-l,2-dithiole-3-thione), N-{2-(2-Chloro-6- fluorophenethyl]-N'-(2-thiazolyl)thiourea (PETT CI, F derivative), N-{2-(2,6-Difluorophenethyl]- N'-[2-(5-bromopyridyl)]thiourea {PETT derivative), N-{2-(2,6-Difluorophenethyl]-N'-[2-(5- methylpyridyl]thiourea {PETT Pyridyl derivative), N-[2-(3-Fluorofuranyl)ethyl]-N'-[2-(5- chloropyridyl)]thiourea, N-[2-(2-Fluoro-6-ethoxyphenethyl)]-N'-[2-(5-bromopyridyl)]thiourea, N-(2-Phenethyl)-N'-(2-thiazolyl)thiourea (LY-73497), L-697,639, L-697,593, L-697,661, 342- (4,7-Difluorobenzoxazol-2-yl)ethyl}-5-ethyl-6-methyl(pypridin-2(lH)-thione (2-Pyridinone Derivative), 3-[[(2-Methoxy-5,6-dimethyl-3-pyridyl)methyl]amine]-5-ethyl-6-methyl(pypridin- 2(lH)-thione, R82150, R82913, R87232, R88703, R89439 (Loviride), R90385, S-2720, Suramin Sodium, TBZ (Thiazolobenzimidazole, NSC 625487), Thiazoloisoindol-5-one, (+)(R)-9b-(3,5- Dimethylphenyl-2,3-dihydrothiazolo[2,3-a]isoindol-5 (9bH)-one, Tivirapine (R86183), UC-38 and UC-84, among others. In one aspect of the invention, the disclosed compound when used to treat an HCV infection can be administered in combination with another anti-HCV agent. Anti-HCV agents are known in the art. To date, a number of fixed dose drug combinations have been approved for the treatment of HCV. Harvoni® (Gilead Sciences, Inc.) contains the NS5A inhibitor ledipasvir and the NS5B inhibitor sofosbuvir. TechnivieTM (AbbVie, Inc.) is a fixed-dose combination containing ombitasvir, an NS5A inhibitor; paritaprevir, an NS3/4A protease inhibitor; and ritonavir, a CYP3A inhibitor. DaklinzaTM (daclatasvir, Bristol-Myers Squibb) is a HCV NS5A inhibitor indicated for use with sofosbuvir for the treatment of chronic genotype 3 infection. ZepatierTM (Merck & Co.) has recently been approved for the treatment of chronic HCV genotypes 1 and 4 . ZepatierTM is a fixed-dose combination product containing elbasvir, an HCV NS5A inhibitor, and grazoprevir, an HCV NS3/4A protease inhibitor. ZepatierTM is indicated with or without ribavirin. Epclusa® (Gilead Sciences, Inc.) is a fixed-dose combination tablet containing sofosbuvir and velpatasvir. Additional anti-HCV agents and combinations thereof include those described in U.S. Patent Nos: 9,382,218; 9,321,753; 9,249,176; 9,233,974; 9,221,833; 9,21 1,315; 9,194,873; 9,186,369; 9,180,193; 9,156,823; 9,138,442; 9,133,170; 9,108,999; 9,090,559; 9,079,887; 9,073,943; 9,073,942; 9,056,090; 9,051,340; 9,034,863; 9,029,413; 9,01 1,938; 8,987,302; 8,945,584; 8,940,718; 8,927,484; 8,921,341; 8,884,030; 8,841,278; 8,822,430; 8,772,022; 8,765,722; 8,742,101; 8,741,946; 8,674,085; 8,673,288; 8,669,234; 8,663,648; 8,618,275; 8,580,252; 8,575,195; 8,575,135; 8,575,1 18; 8,569,302; 8,524,764; 8,513,298; 8,501,714; 8,404,651; 8,273,341; 8,257,699; 8,197,861; 8,158,677; 8,105,586; 8,093,353; 8,088,368; 7,897,565; 7,871,607; 7,846,431; 7,829,081; 7,829,077; 7,824,851; 7,572,621; and 7,326,536; Patents assigned to Alios: U.S. Patent Nos: 9,365,605; 9,346,848; 9,328,1 19; 9,278,990; 9,249,174;

9,243,022; 9,073,960; 9,012,427; 8,980,865; 8,895,723; 8,877,73 1; 8,871,737; 8,846,896 and 8,772,474; Achillion 9,273,082; 9,233,136; 9,227,952; 9,133,1 15; 9,125,904; 9,1 15,175; 9,085,607; 9,006,423; 8,946,422; 8,835,456; 8,809,313; 8,785,378; 8,614,180; 8,445,430; 8,435,984; 8,183,263; 8,173,636; 8,163,693; 8,138,346; 8,1 14,888; 8,106,209; 8,088,806; 8,044,204; 7,985,541; 7,906,619; 7,902,365; 7,767,706; 7,741,334; 7,718,671; 7,659,399; 7,476,686; 7,439,374; 7,365,068; 7,199,128; and 7,094,807; Cocrystal Pharma Inc. 9,181,227; 9,173,893; 9,040,479 and 8,771,665; Gilead Sciences 9,353,423; 9,346,841; 9,321,800; 9,296,782; 9,296,777; 9,284,342; 9,238,039; 9,216,996; 9,206,217; 9,161,934; 9,145,441; 9,139,604; 9,090,653; 9,090,642; 9,085,573; 9,062,092; 9,056,860; 9,045,520; 9,045,462; 9,029,534; 8,980,878; 8,969,588; 8,962,652; 8,957,046; 8,957,045; 8,946,238; 8,933,015; 8,927,741; 8,906,880; 8,889,159; 8,871,785; 8,841,275; 8,815,858; 8,809,330; 8,809,267; 8,809,266; 8,779,141; 8,765,710; 8,759,544; 8,759,510; 8,735,569; 8,735,372; 8,729,089; 8,722,677; 8,716,264; 8,716,263; 8,716,262; 8,697,861; 8,664,386; 8,642,756; 8,637,531; 8,633,309; 8,629,263; 8,618,076; 8,592,397; 8,580,765; 8,569,478; 8,563,530; 8,551,973; 8,536,187; 8,513,186; 8,513,184; 8,492,539; 8,486,938; 8,481,713; 8,476,225; 8,420,597; 8,415,322; 8,338,435; 8,334,270; 8,329,926; 8,329,727; 8,324,179; 8,283,442; 8,263,612; 8,232,278; 8,178,491; 8,173,621; 8,163,718; 8,143,394; patents assigned to Idenix, acquired by Merck, include U.S. Patent Nos.: 9,353,100; 9,309,275; 9,296,778; 9,284,307; 9,249,173; 9,243,025; 9,21 1,300; 9,187,515; 9,187,496, 9,109,001; 8,993,595; 8,951,985; 8,691,788; 8,680,071; 8,637,475; 8,507,460; 8,377,962; 8,362,068; 8,343,937; 8,299,038; 8,193, 372; 8,093,379; 7,951,789; 7,932,240; 7,902,202; 7,662,798; 7,635,689; 7,625,875; 7,608,600; 7,608,597; 7,582,618; 7,547,704; 7,456,155; 7,384,924; 7,365,057; 7,192,936; 7,169,766; 7,163,929; 7,157,441; 7,148,206; 7,138,376; 7,105,493; 6,914,054 and 6,812,219; patents assigned to Merck include U.S. Patent Nos: 9,364,482; 9,339,541; 9,328,138; 9,265,773; 9,254,292; 9,243,002; 9,242,998; 9,242,988; 9,242,917; 9,238,604; 9,156,872; 9,150,603; 9,139,569; 9,120,818; 9,090,661; 9,073,825; 9,061,041; 8,987,195; 8,980,920; 8,927,569; 8,871,759; 8,828,930; 8,772,505; 8,715,638; 8,697,694; 8,637,449; 8,609,635; 8,557,848; 8,546,420; 8,541,434; 8,481,712; 8,470,834; 8,461,107; 8,404,845; 8,377,874; 8,377,873; 8,354,518; 8,309,540; 8,278,322; 8,216,999; 8,148,349; 8,138,164; 8,080,654; 8,071,568; 7,973,040; 7,935,812; 7,915,400; 7,879,815; 7,879,797; 7,632,821; 7,569,374; 7,534,767; 7,470,664 and 7,329,732; patent application publication U S 2013/0029904 to Boehringer Ingelheim GMBH and US 2014/01 13958 to Stella Aps. In one embodiment, the additional therapy is a monoclonal antibody (MAb). Some MAbs stimulate an immune response that destroys cancer cells. Similar to the produced naturally by B cells, these MAbs may "coat" the cancer cell surface, triggering its destruction by the immune system. For example, bevacizumab targets vascular endothelial growth factor(VEGF), a protein secreted by tumor cells and other cells in the tumor's microenvironment that promotes the development of tumor blood vessels. When bound to bevacizumab, VEGF cannot interact with its cellular receptor, preventing the signaling that leads to the growth of new blood vessels. Similarly, cetuximab and panitumumab target the epidermal growth factor receptor (EGFR), and trastuzumab targets the human epidermal growth factor receptor 2 (HER-2). MAbs that bind to cell surface growth factor receptors prevent the targeted receptors from sending their normal growth-promoting signals. They may also trigger apoptosis and activate the immune system to destroy tumor cells. In one aspect of the present invention, the bioactive agent is an immunosuppressive agent. The immunosuppressive agent can be a inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (NEORAL®), FK506 (), , a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g. Sirolimus (RAPAMUNE®), Everolimus (Certican®), temsirolimus, , biolimus-7, biolimus-9, a rapalog, e.g.ridaforolimus, , campath 1H, a SIP receptor modulator, e.g. or an analogue thereof, an anti IL-8 antibody, or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g. Mycophenolate Mofetil (CELLCEPT®), OKT3 (ORTHOCLONE OKT3®), Prednisone, ATGAM®, THYMOGLOBULIN®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15- deoxyspergualin, tresperimus, ARAVA®, CTLAI-Ig, anti-CD25, anti-IL2R, (SIMULECT®), (ZENAPAX®), mizorbine, methotrexate, dexamethasone, ISAtx-247, SDZ ASM 981 (pimecrolimus, Elidel®), CTLA41g (), , LFA31g„ (sold as Enbrel® by Immunex), (Humira®), (Remicade®), an anti-LFA-1 antibody, natalizumab (Antegren®), Enlimomab, , antithymocyte immunoglobulin, , , pentasa, mesalazine, asacol, codeine phosphate, benorylate, fenbufen, naprosyn, diclofenac, etodolac and indomethacin, aspirin and ibuprofen.

V. PHARMACEUTICAL COMPOSITIONS The compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, as disclosed herein can be administered as the neat chemical, but are more typically administered as a pharmaceutical composition, that includes an effective amount for a host, typically a human, in need of such treatment for any of the disorders described herein. Accordingly, the disclosure provides pharmaceutical compositions comprising an effective amount of the disclosed compound or pharmaceutically acceptable salt thereof together with at least one pharmaceutically acceptable carrier for any of the uses described herein. The pharmaceutical composition may contain the disclosed compound or salt as the only active agent, or, in an alternative embodiment, the disclosed compound and at least one additional active agent. Compounds disclosed herein may be administered by any suitable route desired by the healthcare provider, including orally, topically, systemically, parenterally, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, as an ophthalmic solution, injection, including ocular injection, intraveneous, intra arterial, intra-aortal, intracranial, subdermal, intraperitioneal, subcutaneous, transnasal, sublingual, or rectal or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers. In general, the compositions of the disclosure will be administered in a therapeutically effective amount by the desired mode of administration. Suitable dosage ranges depend upon numerous factors such as the severity of the disease to be treated, the age and relative health of the subject, the potency of the compound used, the route and form of administration, the indication towards which the administration is directed, and the preferences and experience of the medical practitioner involved. One of ordinary skill in the art of treating such diseases will be able, without undue experimentation and in reliance upon personal knowledge and the disclosure of this application, to ascertain a therapeutically effective amount of the compositions of the disclosure for a given disease. In certain embodiments the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10, 25, 50 or 100 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 50 to 500, 75 to 500, or 200 mg to about 600 mg of the active compounds and optionally for example from about 0.1 mg to about 2000 mg, from about 10, 25, 50 or 100 mg to about 1000 mg, from about 50 to 500, 75 to 500,from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of an additional active agent in a unit dosage form. Examples are dosage forms with at least 0.1, 1, 5, 10, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, 750 or 800 mg of active compound, or its salt. The therapeutically effective dosage of any active compound described herein will be determined by the health care practitioner depending on the condition, size and age of the patient as well as the route of delivery. In one non-limited embodiment, a dosage from about 0 .1 to about 200 mg/kg, from about 0.01 mg/kg to about 250 mg/kg body weight, more preferably about 0.1 mg/kg to up to about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20 or 30 mg/kg, in at least one dose. In some embodiments, the dosage may be the amount of compound needed to provide a serum concentration of the active compound of up to about 10 nM, 50 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, 1 µΜ, 5 µΜ , 10 µΜ, 20 µΜ, 30 µΜ , or 40 µΜ . The pharmaceutical composition may be formulated as any pharmaceutically useful form, e.g., as an aerosol, a cream, a gel, a pill, an injection or infusion solution, a capsule, a tablet, a syrup, a transdermal patch, a subcutaneous patch, a dry powder, an inhalation formulation, in a medical device, suppository, buccal, or sublingual formulation, parenteral formulation, or an ophthalmic solution. Some dosage forms, such as tablets and capsules, are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose. "Pharmaceutically acceptable carriers" for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, 18th Edition (Easton, Pennsylvania: Mack Publishing Company, 1990). For example, sterile saline and phosphate-buffered saline at physiological pH can be used. Preservatives, stabilizers, dyes and even flavoring agents can be provided in the pharmaceutical composition. For example, sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid can be added as preservatives. Id. at 1449. In addition, antioxidants and suspending agents can be used. Id. Carriers include excipients must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated. The carrier can be inert or it can possess pharmaceutical benefits of its own. The amount of carrier employed in conjunction with the disclosed compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound, as decribed in more detail herein. Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidents, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents. Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others. Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils. Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the disclosed compounds of the present invention. Additionally, auxiliary substances, such as wetting or emulsifying agents, biological buffering substances, surfactants, and the like, can be present in such vehicles. A biological buffer can be any solution which is pharmacologically acceptable and which provides the formulation with the desired pH, i.e., a pH in the physiologically acceptable range. Examples of buffer solutions include saline, phosphate buffered saline, Tris buffered saline, Hank's buffered saline, and the like. Depending on the intended mode of administration, the pharmaceutical compositions can be in the form of solid, semi-solid or liquid dosage forms, such as, for example, tablets, suppositories, pills, capsules, powders, liquids, suspensions, creams, ointments, lotions or the like, preferably in unit dosage form suitable for single administration of a precise dosage. The compositions will include an effective amount of the selected drug in combination with a pharmaceutically acceptable carrier and, in addition, can include other pharmaceutical agents, adjuvants, diluents, buffers, and the like. Thus, the compositions of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, pulmonary, vaginal or parenteral (including intramuscular, intra-arterial, intrathecal, subcutaneous and intravenous) administration or in a form suitable for administration by inhalation or insufflation. The preferred manner of administration is intravenous or oral using a convenient daily dosage regimen which can be adjusted according to the degree of affliction. For solid compositions, conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like. Liquid pharmaceutically administrable compositions can, for example, be prepared by dissolving, dispersing, and the like, an active compound as described herein and optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol, and the like, to thereby form a solution or suspension. If desired, the pharmaceutical composition to be administered can also contain minor amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like, for example, sodium acetate, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, and the like. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, referenced above. In yet another embodiment is the use of permeation enhancer excipients including polymers such as: polycations (chitosan and its quaternary ammonium derivatives, poly-L- arginine, aminated gelatin); polyanions (N -carboxymethyl chitosan, poly-acrylic acid); and, thiolated polymers (carboxymethyl cellulose-cysteine, polycarbophil-cysteine, chitosan- thiobutylamidine, chitosan-thiogly colic acid, chitosan-glutathione conjugates). For oral administration, the composition will generally take the form of a tablet, capsule, a softgel capsule or can be an aqueous or nonaqueous solution, suspension or syrup. Tablets and capsules are preferred oral administration forms. Tablets and capsules for oral use can include one or more commonly used carriers such as lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. Typically, the compositions of the disclosure can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like. When liquid suspensions are used, the active agent can be combined with any oral, non toxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like and with emulsifying and suspending agents. If desired, flavoring, coloring and/or sweetening agents can be added as well. Other optional components for incorporation into an oral formulation herein include, but are not limited to, preservatives, suspending agents, thickening agents, and the like. Parenteral formulations can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solubilization or suspension in liquid prior to injection, or as emulsions. Preferably, sterile injectable suspensions are formulated according to techniques known in the art using suitable carriers, dispersing or wetting agents and suspending agents. The sterile injectable formulation can also be a sterile injectable solution or a suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils, fatty esters or polyols are conventionally employed as solvents or suspending media. In addition, parenteral administration can involve the use of a slow release or sustained release system such that a constant level of dosage is maintained. Parenteral administration includes intraarticular, intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, and include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. Administration via certain parenteral routes can involve introducing the formulations of the disclosure into the body of a patient through a needle or a catheter, propelled by a sterile syringe or some other mechanical device such as an continuous infusion system. A formulation provided by the disclosure can be administered using a syringe, injector, pump, or any other device recognized in the art for parenteral administration. Preferably, sterile injectable suspensions are formulated according to techniques known in the art using suitable carriers, dispersing or wetting agents and suspending agents. The sterile injectable formulation can also be a sterile injectable solution or a suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils, fatty esters or polyols are conventionally employed as solvents or suspending media. In addition, parenteral administration can involve the use of a slow release or sustained release system such that a constant level of dosage is maintained. Preparations according to the disclosure for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, or emulsions. Examples of non-aqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms can also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They can be sterilized by, for example, filtration through a bacteria retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They can also be manufactured using sterile water, or some other sterile injectable medium, immediately before use. Sterile injectable solutions are prepared by incorporating one or more of the compounds of the disclosure in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof. Thus, for example, a parenteral composition suitable for administration by injection is prepared by stirring 1.5% by weight of active ingredient in 10% by volume propylene glycol and water. The solution is made isotonic with sodium chloride and sterilized. Formulations suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active disclosed compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture. Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof. Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis {see, for example, Pharmaceutical Research 3 (6):3 18 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound. In one embodiment, microneedle patches or devices are provided for delivery of drugs across or into biological tissue, particularly the skin. The microneedle patches or devices permit drug delivery at clinically relevant rates across or into skin or other tissue barriers, with minimal or no damage, pain, or irritation to the tissue. Formulations suitable for administration to the lungs can be delivered by a wide range of passive breath driven and active power driven single/-multiple dose dry powder inhalers (DPI). The devices most commonly used for respiratory delivery include nebulizers, metered-dose inhalers, and dry powder inhalers. Several types of nebulizers are available, including jet nebulizers, ultrasonic nebulizers, and vibrating mesh nebulizers. Selection of a suitable lung delivery device depends on parameters, such as nature of the drug and its formulation, the site of action, and pathophysiology of the lung. Additional non-limiting examples of drug delivery devices and methods include, for example, US20090203709 titled "Pharmaceutical Dosage Form For Oral Administration Of Tyrosine Kinase Inhibitor" (Abbott Laboratories); US20050009910 titled "Delivery of an active drug to the posterior part of the eye via subconjunctival or periocular delivery of a prodrug", US 20130071349 titled "Biodegradable polymers for lowering intraocular pressure", US 8,481,069 titled "Tyrosine kinase microspheres", US 8,465,778 titled "Method of making tyrosine kinase microspheres", US 8,409,607 titled "Sustained release intraocular implants containing tyrosine kinase inhibitors and related methods", US 8,512,738 and US 2014/003 1408 titled "Biodegradable intravitreal tyrosine kinase implants", US 2014/0294986 titled "Microsphere Drug Delivery System for Sustained Intraocular Release", US 8,91 1,768 titled "Methods For Treating Retinopathy With Extended Therapeutic Effect" (Allergan, Inc.); US 6,495, 164 titled "Preparation of injectable suspensions having improved injectability" (Alkermes Controlled Therapeutics, Inc.); WO 2014/047439 titled "Biodegradable Microcapsules Containing Filling Material" (Akina, Inc.); WO 2010/132664 titled "Compositions And Methods For Drug Delivery" (Baxter International Inc. Baxter Healthcare SA); US20 120052041 titled "Polymeric nanoparticles with enhanced drug loading and methods of use thereof (The Brigham and Women's Hospital, Inc.); US20140178475, US20140248358, and US20140249158 titled "Therapeutic Nanoparticles Comprising a Therapeutic Agent and Methods of Making and Using Same" (BIND Therapeutics, Inc.); US 5,869,103 titled "Polymer microparticles for drug delivery" (Danbiosyst UK Ltd.); US 8628801 titled "Pegylated Nanoparticles" (Universidad de Navarra); US20 14/0 107025 titled "Ocular drug delivery system" (Jade Therapeutics, LLC); US 6,287,588 titled "Agent delivering system comprised of microparticle and biodegradable gel with an improved releasing profile and methods of use thereof, US 6,589,549 titled "Bioactive agent delivering system comprised of microparticles within a biodegradable to improve release profiles" (Macromed, Inc.); US 6,007,845 and US 5,578,325 titled "Nanoparticles and microparticles of non-linear hydrophilic hydrophobic multiblock copolymers" (Massachusetts Institute of Technology); US2004023461 1, US20080305172, US20120269894, and US20130122064 titled "Ophthalmic depot formulations for periocular or subconjunctival administration (Novartis Ag); US 6,413,539 titled "Block polymer" (Poly-Med, Inc.); US 20070071756 titled "Delivery of an agent to ameliorate inflammation" (Peyman); US 2008016641 1 titled "Injectable Depot Formulations And Methods For Providing Sustained Release Of Poorly Soluble Drugs Comprising Nanoparticles" (Pfizer, Inc.); US 6,706,289 titled "Methods and compositions for enhanced delivery of bioactive molecules" (PR Pharmaceuticals, Inc.); and US 8,663,674 titled "Microparticle containing matrices for drug delivery" (Surmodics).

VI. COMBINATION THERAPY The compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, Formula VIII, Formula IX, Formula X, Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI, Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX, can be used in an effective amount alone or in combination to treat a host such as a human with any of the disorders as described herein. In one aspect of this embodiment, the second active compound is an immune modulator, including but not limited to a checkpoint inhibitor such as an anti-PDl, anti-CTLA, anti-LAG-3, anti-Tim, etc antibody, small molecule, peptide, nucleotide or other inhibitor (including but not limited to ipilimumab (Yervoy), Pembrolizumab (Keytruda) and nivolumab (Opdivo). In yet another embodiment, one of the active compounds described herein is administered in an effective amount for the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer, in combination or alternation with an effective amount of an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist. Partial anti-estrogens like raloxifene and tamoxifen retain some estrogen-like effects, including an estrogen-like stimulation of uterine growth, and also, in some cases, an estrogen-like action during breast cancer progression which actually stimulates tumor growth. In contrast, fulvestrant, a complete anti-estrogen, is free of estrogen-like action on the uterus and is effective in tamoxifen-resistant tumors. Non-limiting examples of anti-estrogen compounds are provided in WO 2014/19176 assigned to Astra Zeneca, WO20 13/090921, WO 2014/203129, WO 2014/203132, and US20 13/0 178445 assigned to Olema Pharmaceuticals, and U.S. Patent Nos. 9,078,871, 8,853,423, and 8,703, 810, as well as US 2015/0005286, WO 2014/205136, and WO 2014/205138. Additional non-limiting examples of anti-estrogen compounds include: SERMS such as anordrin, bazedoxifene, broparestriol, chlorotrianisene, clomiphene citrate, cyclofenil, lasofoxifene, ormeloxifene, raloxifene, tamoxifen, toremifene, and fulvestratnt; aromatase inhibitors such as aminoglutethimide, testolactone, anastrozole, exemestane, fadrozole, formestane, and letrozole; and antigonadotropins such as leuprorelin, cetrorelix, allylestrenol, chloromadinone acetate, cyproterone acetate, delmadinone acetate, dydrogesterone, medroxyprogesterone acetate, megestrol acetate, , , progesterone, and spironolactone. In another embodiment, one of the active compounds described herein is administered in an effective amount for the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, in combination or alternation with an effective amount of an androgen (such as testosterone) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist. In one embodiment, the prostate or testicular cancer is androgen-resistant. Non-limiting examples of anti-androgen compounds are provided in WO 201 1/156518 and US Patent Nos. 8,455,534 and 8,299,1 12. Additional non- limiting examples of anti-androgen compounds include: enzalutamide, apalutamide, cyproterone acetate, chlormadinone acetate, spironolactone, canrenone, drospirenone, ketoconazole, topilutamide, abiraterone acetate, and cimetidine. In one aspect, a treatment regimen is provided comprising the administration of a compound of the present invention in combination with at least one additional chemotherapeutic agent. The combinations disclosed herein can be administered for beneficial, additive, or synergistic effect in the treatment of abnormal cellular proliferative disorders. In specific embodiments, the treatment regimen includes the administration of a compound of the present invention in combination with at least one kinase inhibitor. In one embodiment, the at least one kinase inhibitor is selected from a phosphoinositide 3-kinase (PI3K) inhibitor, a Bruton's tyrosine kinase (BTK) inhibitor, or a spleen tyrosine kinase (Syk) inhibitor, or a combination thereof. Additional anti-cancer agents include, for example, everolimus, trabectedin, abraxane, TLK 286, AV-299, DN-101, pazopanib, GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY- 142886), AMN-107, TKI-258, GSK461364, AZD 1152, enzastaurin, vandetanib, ARQ-197, MK- 0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, a VEGFR inhibitor, an EGFR TK inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, a Be1-2 inhibitor, an HDAC inhbitor, a c-MET inhibitor, a PARP inhibitor, a Cdk inhibitor, an EGFR TK inhibitor, an IGFR-TK inhibitor, an anti-HGF antibody, aPI3 kinase inhibitor, an AKT inhibitor, an mTORCl/2 inhibitor, a JAK/STAT inhibitor, a checkpoint-1 or 2 inhibitor, a focal adhesion kinase inhibitor, a Map kinase kinase (mek) inhibitor, a VEGF trap antibody, pemetrexed, erlotinib, dasatanib, nilotinib, decatanib, panitumumab, amrubicin, oregovomab, Lep-etu, nolatrexed, azd2171, batabulin, ofatumumab, zanolimumab, edotecarin, tetrandrine, rubitecan, tesmilifene, oblimersen, ticilimumab, ipilimumab, gossypol, Bio 111, 131-I-TM-601, ALT-1 10, BIO 140, CC 8490, cilengitide, gimatecan, IL13-PE38QQR, INO 1001, IPdRi KRX-0402, lucanthone, LY3 17615, neuradiab, vitespan, Rta 744, Sdx 102, talampanel, atrasentan, Xr 3 11, romidepsin, ADS-100380, sunitinib, 5-fluorouracil, vorinostat, etoposide, gemcitabine, doxorubicin, liposomal doxorubicin, 5'-deoxy-5-fluorouridine, vincristine, temozolomide, ZK-304709, seliciclib; PD0325901, AZD- 6244, capecitabine, L-Glutamic acid, N-[4-[2-(2-amino-4,7-dihydro-4-oxo-lH-pyrrolo[2,3- d]pyrimidin-5-yl)ethyl]benzoyl]-, disodium salt, heptahydrate, camptothecin, PEG-labeled irinotecan, tamoxifen, toremifene citrate, anastrazole, exemestane, letrozole,

DES(diethylstilbestrol), estradiol, estrogen, conjugated estrogen, bevacizumab, IMC-1C1 1, CHIR-258); 3-[5-(methylsulfonylpiperadinemethyl)-indolyl-quinolone, vatalanib, AG-013736, AVE-0005, goserelin acetate, leuprolide acetate, triptorelin pamoate, medroxyprogesterone acetate, hydroxyprogesterone caproate, megestrol acetate, raloxifene, bicalutamide, flutamide, nilutamide, megestrol acetate, CP-724714; TAK-165, HKI-272, erlotinib, lapatanib, canertinib, ABX-EGF antibody, erbitux, EKB-569, PKI-166, GW-572016, Ionafarnib, BMS-214662, tipifarnib; amifostine, NVP-LAQ824, suberoyl analide hydroxamic acid, valproic acid, trichostatin A, FK-228, SU1 1248, sorafenib, KRN95 1, aminoglutethimide, arnsacrine, anagrelide, L-asparaginase, Bacillus Calmette-Guerin (BCG) vaccine, adriamycin, bleomycin, buserelin, busulfan, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, diethylstilbestrol, epirubicin, fludarabine, fludrocortisone, fluoxymesterone, flutamide, gleevec, gemcitabine, hydroxyurea, idarubicin, ifosfamide, imatinib, leuprolide, levamisole, lomustine, mechlorethamine, melphalan, 6- mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, octreotide, oxaliplatin, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, teniposide, testosterone, thalidomide, thioguanine, thiotepa, tretinoin, vindesine, 13- cis-retinoic acid, phenylalanine mustard, uracil mustard, estramustine, altretamine, floxuridine, 5- deooxyuridine, cytosine arabinoside, 6-mecaptopurine, deoxycoformycin, calcitriol, valrubicin, mithramycin, vinblastine, vinorelbine, topotecan, razoxin, marimastat, COL-3, neovastat, BMS- 275291, squalamine, endostatin, SU5416, SU6668, EMD121974, interleukin-12, EVI862, angiostatin, vitaxin, droloxifene, idoxyfene, spironolactone, finasteride, cimitidine, trastuzumab, denileukin diftitox, gefitinib, bortezimib, paclitaxel, cremophor-free paclitaxel, docetaxel, epithilone B, BMS-247550, BMS-3 10705, droloxifene, 4-hydroxytamoxifen, pipendoxifene, ERA-923, arzoxifene, fulvestrant, acolbifene, lasofoxifene, idoxifene, TSE-424, HMR-3339, ZK186619, topotecan, PTK787/ZK 222584, VX-745, PD 184352, rapamycin, 40-O-(2- hydroxyethyl)-rapamycin, temsirolimus, AP-23573, RAD001, ABT-578, BC-210, LY294002, LY292223, LY292696, LY293684, LY293646, wortmannin, ZM336372, L-779,450, PEG- filgrastim, darbepoetin, erythropoietin, granulocyte colony-stimulating factor, zolendronate, prednisone, cetuximab, granulocyte macrophage colony-stimulating factor, histrelin, pegylated interferon alfa-2a, interferon alfa-2a, pegylated interferon alfa-2b, interferon alfa-2b, azacitidine,

PEG-L-asparaginase, lenalidomide, gemtuzumab, hydrocortisone, interleukin-1 1, dexrazoxane, alemtuzumab, all-transretinoic acid, ketoconazole, interleukin-2, megestrol, immune globulin, nitrogen mustard, methylprednisolone, ibritgumomab tiuxetan, androgens, decitabine, hexamethylmelamine, bexarotene, tositumomab, arsenic trioxide, cortisone, editronate, mitotane, cyclosporine, liposomal daunorubicin, Edwina-asparaginase, strontium 89, casopitant, netupitant, an K-1 receptor antagonist, palonosetron, aprepitant, diphenhydramine, hydroxyzine, metoclopramide, lorazepam, alprazolam, haloperidol, droperidol, dronabinol, dexamethasone, methylprednisolone, prochlorperazine, granisetron, ondansetron, dolasetron, tropisetron, pegfilgrastim, erythropoietin, epoetin alfa, darbepoetin alfa and mixtures thereof. The term "anti-HIV agent" or "additional anti-HIV agent" includes, for example, nucleoside reverse transcriptase inhibitors ( RTI), other non-nucloeoside reverse transcriptase inhibitors (i.e., those which are not representative of the present invention), protease inhibitors, fusion inhibitors, among others, exemplary compounds of which may include, for example, 3TC (Lamivudine), AZT (Zidovudine), (-)-FTC, ddl (Didanosine), ddC (zalcitabine), abacavir (ABC), tenofovir (PMPA), D-D4FC (Reverset), D4T (Stavudine), Racivir, L-FddC, L-FD4C, NVP (Nevirapine), DLV (Delavirdine), EFV (Efavirenz), SQVM (Saquinavir mesylate), RTV (Ritonavir), IDV (Indinavir), SQV (Saquinavir), FV (Nelfinavir), APV (Amprenavir), LPV (Lopinavir), fusion inhibitors such as T20, among others, fuseon and mixtures thereof, including anti-HIV compounds presently in clinical trials or in development. Other anti-HIV agents which may be used in coadministration with compounds according to the present invention include, for example, other NNRTI's (i.e., other than the NNRTI's according to the present invention) may be selected from the group consisting of nevirapine (BI- R6-587), delavirdine (U-90152S/T), efavirenz (DMP-266), UC-781 (N-[4-chloro-3-(3-methyl-2- butenyloxy)phenyl]-2methyl3-furancarbothiamide), etravirine (TMC125), Trovirdine (Ly300046.HCl), HI-236, HI-240, HI-280, HI-281, rilpivirine (TMC-278), MSC-127, HBY 097, DMP266, Baicalin (TJN-151) ADAM-II (Methyl 3',3'-dichloro-4',4"-dimethoxy-5',5"- bis(methoxycarbonyl)-6,6-diphenylhexenoate), Methyl 3-Bromo-5-(l-5-bromo-4-methoxy-3- (methoxycarbonyl)phenyl)hept- 1-enyl)-2-methoxybenzoate (Alkenyldiarylmethane analog, Adam analog), (5-chloro-3-(phenylsulfinyl)-2'-indolecarboxamide), AAP-BHAP (U-104489 or PNU-104489), Capravirine (AG-1549, S-1 153), atevirdine (U-87201E), aurin tricarboxylic acid

(SD-095345), l-[(6-cyano-2-indolyl)carbonyl]-4-[3-(isopropylamino)-2-pyridinyl]piperazine, 1- [5-[[N-(methyl)methylsulfonylamino]-2-indolylcarbonyl-4-[3-(isopropylamino)-2- pyridinyljpiperazine, l-[3-(Ethylamino)-2-[pyridinyl]-4-[(5-hydroxy-2- indolyl)carbonyl]piperazine, l-[(6-Formyl-2-indolyl)carbonyl]-4-[3-(isopropylamino)-2- pyridinyljpiperazine, l-[[5-(Methylsulfonyloxy)-2-indoyly)carbonyl]-4-[3-(isopropylamino)-2- pyridinyl]piperazine, U88204E, Bis(2-nitrophenyl)sulfone (NSC 633001), Calanolide A (NSC675451), Calanolide B, 6-Benzyl-5-methyl-2-(cyclohexyloxy)pyrimidin-4-one (DABO- 546), DPC 961, E-EBU, E-EBU-dm, E-EPSeU, E-EPU, Foscarnet (Foscavir), HEPT (l-[(2- Hydroxyethoxy)methyl]-6-(phenylthio)thymine), HEPT-M ( 1-[(2-Hydroxyethoxy)methyl]-6-(3- methylphenyl)thio)thymine), HEPT-S(1 -[(2-Hydroxyethoxy)methyl]-6-(phenylthio)-2- thiothymine), Inophyllum P, L-737,126, Michellamine A (NSC650898), Michellamine B (NSC649324), Michellamine F, 6-(3,5-Dimethylbenzyl)-l-[(2-hydroxyethoxy)methyl]-5- isopropyluracil, 6-(3,5-Dimethylbenzyl)-l-(ethyoxymethyl)-5-isopropyluracil, NPPS, E-BPTU (NSC 648400), Oltipraz (4-Methyl-5-(pyrazinyl)-3H-l,2-dithiole-3-thione), N-{2-(2-Chloro-6- fluorophenethyl]-N'-(2-thiazolyl)thiourea (PETT CI, F derivative), N-{2-(2,6-Difluorophenethyl]- N'-[2-(5-bromopyridyl)]thiourea {PETT derivative), N-{2-(2,6-Difluorophenethyl]-N'-[2-(5- methylpyridyl]thiourea {PETT Pyridyl derivative), N-[2-(3-Fluorofuranyl)ethyl]-N'-[2-(5- chloropyridyl)]thiourea, N-[2-(2-Fluoro-6-ethoxyphenethyl)]-N'-[2-(5-bromopyridyl)]thiourea, N-(2-Phenethyl)-N'-(2-thiazolyl)thiourea (LY-73497), L-697,639, L-697,593, L-697,661, 342- (4,7-Difluorobenzoxazol-2-yl)ethyl}-5-ethyl-6-methyl(pypridin-2(lH)-thione (2-Pyridinone Derivative), 3-[[(2-Methoxy-5,6-dimethyl-3-pyridyl)methyl]amine]-5-ethyl-6-methyl(pypridin- 2(lH)-thione, R82150, R82913, R87232, R88703, R89439 (Loviride), R90385, S-2720, Suramin Sodium, TBZ (Thiazolobenzimidazole, NSC 625487), Thiazoloisoindol-5-one, (+)(R)-9b-(3,5- Dimethylphenyl-2,3-dihydrothiazolo[2,3-a]isoindol-5 (9bH)-one, Tivirapine (R86183), UC-38 and UC-84, among others. In one embodiment, a combination described herein can be further combined with an additional therapeutic to treat the cancer. The second therapy can be an immunotherapy. As discussed in more detail below, the compound of the present invention can be conjugated to an antibody, radioactive agent, or other targeting agent that directs the compound to the diseased or abnormally proliferating cell. In another embodiment, the combination is used in combination with another pharmaceutical or a biologic agent (for example an antibody) to increase the efficacy of treatment with a combined or a synergistic approach. In an embodiment, combination can be used with T-cell vaccination, which typically involves immunization with inactivated autoreactive T cells to eliminate a cancer cell population as described herein. In another embodiment, the combination is used in combination with a bispecific T-cell Engager (BiTE), which is an antibody designed to simultaneously bind to specific antigens on endogenous T cells and cancer cells as described herein, linking the two types of cells. In one embodiment, the additional therapy is a monoclonal antibody (MAb). Some MAbs stimulate an immune response that destroys cancer cells. Similar to the antibodies produced naturally by B cells, these MAbs "coat" the cancer cell surface, triggering its destruction by the immune system. For example, bevacizumab targets vascular endothelial growth factor(VEGF), a protein secreted by tumor cells and other cells in the tumor's microenvironment that promotes the development of tumor blood vessels. When bound to bevacizumab, VEGF cannot interact with its cellular receptor, preventing the signaling that leads to the growth of new blood vessels. Similarly, cetuximab and panitumumab target the epidermal growth factor receptor (EGFR), and trastuzumab targets the human epidermal growth factor receptor 2 (HER-2). MAbs that bind to cell surface growth factor receptors prevent the targeted receptors from sending their normal growth-promoting signals. They may also trigger apoptosis and activate the immune system to destroy tumor cells. Another group of cancer therapeutic MAbs are the immunoconjugates. These MAbs, which are sometimes called immunotoxins or antibody-drug conjugates, consist of an antibody attached to a cell-killing substance, such as a plant or bacterial toxin, a drug, or a radioactive molecule. The antibody latches onto its specific antigen on the surface of a cancer cell, and the cell-killing substance is taken up by the cell. FDA-approved conjugated MAbs that work this way include ado-trastuzumab emtansine, which targets the FIER-2 molecule to deliver the drug DM1, which inhibits cell proliferation, to FIER-2 expressing metastatic breast cancer cells. Immunotherapies with T cells engineered to recognize cancer cells via bispecific antibodies (bsAbs) or chimeric antigen receptors (CARs) are approaches with potential to ablate both dividing and non/slow-dividing subpopulations of cancer cells. In one aspect of the present invention, a compound described herein can be combined with at least one immunosuppressive agent. The immunosuppressive agent is preferably selected from the group consisting of a calcineurin inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (NEORAL®), FK506 (tacrolimus), pimecrolimus, a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g. Sirolimus (RAPAMUNE®), Everolimus (Certican®), temsirolimus, zotarolimus, biolimus-7, biolimus-9, a rapalog, e.g.ridaforolimus, azathioprine, campath 1H, a SIP receptor modulator, e.g. fingolimod or an analogue thereof, an anti IL-8 antibody, mycophenolic acid or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g. Mycophenolate Mofetil (CELLCEPT®), OKT3 (ORTHOCLONE OKT3®), Prednisone, ATGAM®, THYMOGLOBULIN®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15-deoxyspergualin, tresperimus, Leflunomide ARAVA®, CTLAI-Ig, anti-CD25, anti-IL2R, Basiliximab (SIMULECT®), Daclizumab (ZENAPAX®), mizorbine, methotrexate, dexamethasone, ISAtx- 247, SDZ ASM 981 (pimecrolimus, Elidel®), CTLA41g (Abatacept), belatacept, LFA31g„ etanercept (sold as Enbrel® by Immunex), adalimumab (Humira®), infliximab (Remicade®), an anti-LFA-1 antibody, natalizumab (Antegren®), Enlimomab, gavilimomab, antithymocyte immunoglobulin, siplizumab, Alefacept efalizumab, pentasa, mesalazine, asacol, codeine phosphate, benorylate, fenbufen, naprosyn, diclofenac, etodolac and indomethacin, aspirin and ibuprofen. In certain embodiments, a compound described herein is administered to the subject prior to treatment with another chemotherapeutic agent, during treatment with another chemotherapeutic agent, after administration of another chemotherapeutic agent, or a combination thereof. In some embodiments, the compound of the present invention can be administered to the subject such that the other chemotherapeutic agent can be administered either at higher doses (increased chemotherapeutic dose intensity) or more frequently (increased chemotherapeutic dose density). Dose-dense chemotherapy is a chemotherapy treatment plan in which drugs are given with less time between treatments than in a standard chemotherapy treatment plan. Chemotherapy dose intensity represents unit dose of chemotherapy administered per unit time. Dose intensity can be increased or decreased through altering dose administered, time interval of administration, or both. The term "pharmaceutically acceptable salt" is used throughout the specification to describe, where applicable, a salt form of one or more of the compounds described herein which are presented to increase the solubility of the compound in the gastic juices of the patient's gastrointestinal tract in order to promote dissolution and the bioavailability of the compounds. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable inorganic or organic bases and acids, where applicable. Suitable salts include those derived from alkali metals such as potassium and sodium, alkaline earth metals such as calcium, magnesium and ammonium salts, among numerous other acids and bases well known in the pharmaceutical art. Sodium and potassium salts are particularly preferred as neutralization salts of the phosphates according to the present invention.

VII. GENERAL SYNTHESIS

The compounds described herein can be prepared by methods known by those skilled in the art. In one non-limiting example the disclosed compounds can be made by the schemes below.

Compounds of the present invention with stereocenters may be drawn without stereochemistry for convenience. One skilled in the art will recognize that pure enantiomers and diastereomers can be prepared by methods known in the art. Examples of methods to obtain optically active materials include at least the following.

i) Physical separation of crystals—a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct;

ii) Simultaneous crystallization—a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;

iii) Enzymatic resolutions—a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme;

iv) Enzymatic asymmetric synthesis—a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer; v) Chemical asymmetric synthesis—a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries;

vi) Diastereomer separations—a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;

vii) First- and second-order asymmetric transformations—a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer;

viii) Kinetic resolutions—this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;

ix) Enantiospecific synthesis from non-racemic precursors—a synthetic technique whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;

x) Chiral liquid chromatography—a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase (including via chiral HPLC). The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions; xi) Chiral gas chromatography—a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;

xii) Extraction with chiral solvents—a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent;

xiii) Transport across chiral membranes—a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non- racemic chiral nature of the membrane that allows only one enantiomer of the racemate to pass through.

xiv) Simulated moving bed chromatography, is used in one embodiment. A wide variety of chiral stationary phases are commercially available.

General Scheme 1

Linker Degron Degron Linker Degron Linker Targeting Ligand Step 1 Step 2

General! Scheme 2

Linker Degron [Targeting Ligand Targeting Ligand Linker Step 1

sgron Linker [Targeting Ligand

As shown in General Scheme 1 compounds for use in the present invention can be prepared by chemically combining a Degron and a Linker followed by subsequent addition of a Targeting Ligand. Similarly, in General Scheme 2 compounds for use in the present invention are prepared by chemically combing a Targeting Ligand and Linker first, followed by subsequent addition of a Degron. As illustrated in the above and following schemes, compounds for use in the present invention can readily be synthesized by one skilled in the art in a variety of methods and chemical reactions. General Scheme 3

LG- Linker- -PG Degron Degron Linker -PG Degron Linker Step 1 Step 2

LG-†Targeting Ligand Degron Linker [Targeting Ligand Step 3

General Scheme 3:In Step 1, a nucleophilic Degron displaces a leaving group on the Linker to make a Degron Linker fragment. In Step 2, the protecting group is removed by methods known in the art to free a nucleophilic site on the Linker. In Step 3, the nucleophilic Degron Linker fragment displaces a leaving group on the Targeting Ligand to form a compound for use in the present invention. In an alternative embodiment Step 1 and/or Step 2 is accomplished by a coupling reaction instead of a nucleophilic attack. General Scheme 4

LG- Linker- Targeting Ligand Targeting Ligand-4 Linker PG Siep 1 Step 2

LG~4Degron [Targeting Ligand- Linker 5gron Linker Targeting Ligand Step 3

General Scheme 4 : In Step 1, a nucleophilic Targeting Ligand displaces a leaving group on the Linker to make a Targeting Ligand Linker fragment. In Step 2, the protecting group is removed by methods known in the art to free a nucleophilic site on the Linker. In Step 3, the nucleophilic Targeting Ligand Linker fragment displaces a leaving group on the Degron to form a compound for use in the present invention. In an alternative embodiment Step 1 and/or Step 2 is accomplished by a coupling reaction instead of a nucleophilic attack.

VIII. SYNTHESIS OF REPRESENTATIVE COMPOUNDS In the following Schemes, R is the point of attachment to the linker. A solution of 3-nitrophthalic anhydride ( 1 equiv.) in acetone is subjected to catalytic reduction under hydrogen (60 psi) in the presence of Raney nickel and anhydrous . After 2-3 hours, the solution is warmed to 50 °C, treated with Norite, filtered, and concentrated in vacuo at room temperature. The residue is treated with ethyl acetate, chilled and collected to give 3-aminophthalic anhydride. 15 N sulfuric acid is cooled to 0 °C, 3-aminophthalic anhydride ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the resulting crude 3-hydroxyphthalic acid sublimes at about 160-180 °C (0.2 mm.) to give 3-hydroxyphthalic anhydride. (J. Am. Chem. Soc, 1955, 77 (19), pp 5092-5095)

4-Hydroxythalidomide

A mixture of 3-hydroxyphthalic anhydride ( 1 equiv.), 3-aminopiperidine-2,6-dione HC1 salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- hydroxythalidomide.

4-Alkoxythalidomide

Alkyl halide ( 1 equiv.) is dissolved in MeCN. 4-hydroxythalidomide (1.1 equiv.) and cesium carbonate (2.75 equiv.) are added and the mixture is heated at 80 °C overnight. The mixture is cooled to room temperature and diluted water and extracted with chloroform and EtOAc. The combined organic layers are dried over sodium sulfate, filtered and concentrated. The crude is purified by column chromatography on silica gel to give 4-alkoxythalidomide. {Science 2015, 348 (6241), 1376-1381)

General procedure to prepare pomalidomide derivatives:

4-Fluorothalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 3-aminopiperidine-2,6-dione HC1 salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated

NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- fluorothali domide. Pomalidomide analogs

A mixture of amine ( 1 equiv.), 4-fluorothalidomide ( 1 equiv.) and DIEA (2 eq.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give pomalidomide analogs. (Chem Biol. 2015 Jun 18;22(6):755-63.)

General procedure to prepare lenalidomide derivatives:

3-(7-Nitro-l-oxoisoindolin-2-yl)piperidine-2,6-dione

To a solution of a-aminoglutarimide (1.0 equiv.) and methyl 2-(bromomethyl)-3- nitrobenzoate (1.3 equiv) in DMF is added triethylamine (3.0 equiv), and the mixture is heated to 75 °C and stirred for 20 h . Then it is cooled to RT and poured into LiCl (5% in H2O). It is extracted with EtOAc, and the combined organic phases are washed with LiCl (5% in H2O) and saturated aqueous NaCl, dried (Na2S0 4), filtered, and concentrated in vacuo. The crude is purified by column chromatography on silica gel to give 3-(7-nitro-l-oxoisoindolin-2-yl)piperidine-2,6-dione. (Org. Lett., 2013, 15 (17), pp 4312-4315) Lenalidomide

A mixture of 3-(7-nitro-l-oxoisoindolin-2-yl)piperidine-2,6-dione ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give lenalidomide. Lenalidomide anal

Alkyl halide ( 1 equiv.) is dissolved in DMF. Lenalidomide (1.1 equiv.) and potassium carbonate (2 equiv.) are added and the mixture is stirred at rt overnight. The mixture is diluted water and extracted with EtOAc. The combined organic layer is dried over sodium sulfate, filtered and concentrated. The crude is purified by column chromatography on silica gel to give lenalidomide analogs.

General procedure to prepare Boc-protected glutarimide:

To a stirred solution of 4-fluorothalidomide (1.0 equiv.) in DMF is added potassium carbonate (2.0 equiv.). The resulting solution is cooled to 0-5 °C and Boc anhydride (3.0 equiv.) is added slowly as a solution in 1,4-dioxane (also cooled). The resulting mixture is stirred at 0 °C for 1 h and allowed to warm to room temperature overnight. Water is then added and the aqueous layer is extracted 2 times with DCM and concentrated. The residue is purified by column chromatography on silica gel to obtain N-Boc 4-fluorothalidomide. (WO 2014/147531 A2)

Thal Pom Len analogs with variation on the phthalimide moiety General procedure to prepare O-aryl thalidomide: Example: O-Pyrazole thalidomide

l-(3-Hydroxy-lH-pyrazol-l-yl)ethanone

A solution of 3-hydroxy-lH-pyrazole ( 1 equiv.) in pyridine is heated to 95 °C. A mixture consisting of acetic anhydride (1.05 equiv.) and pyridine is added over 20 min. After stirring at 95

°C for 2 h, all volatiles are removed in vacuo and Et20 is added to the residue. The slurry is stirred overnight at rt. The solid was filtered off and washed with Et20 to give the 1-(3 -hydroxy- 1H- pyrazol-l-yl)ethanone. (ChemMedChem 2015, 10, 1184 - 1199) t rt-butyl 3-(4-((l-acetyl-lH-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l- carboxylate l-(3-Hydroxy-lH-pyrazol-l-yl)ethanone ( 1 equiv.) is dissolved in DMF. NaH (1.5 equiv.) is added slowly and stirred at rt for 30 min. 4-Fluorothalidomide ( 1 equiv.) is added and the solution is stirred at rt or 90 °C until the consumption of the starting materials. The reaction is quenched with methanol and extracted with EtOAc. The organic layer is washed with brine, dried

(Na2S0 4), and concentrated. The residue is purified by column chromatography on silica gel to give t rt-butyl 3-(4-((l-acetyl-lH-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6- dioxopiperidine-l-carboxylate. (PCT Int. Appl., 2015087234, 18 Jun 2015) tert- y 3-(4-((lH-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l- carboxylate

t rt-butyl 3-(4-((l-acetyl-lH-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6- dioxopiperidine-l-carboxylate ( 1 equiv.) is dissolved in a mixture of MeOH/THF (2:3). 10% NaOH is added, and the solution is stirred at rt for 5 h . Then all the volatiles are removed in vacuo. The residue is diluted with EtOAc and water. The aqueous phase was extracted with EtOAc. The combined organic layers are washed with water and brine and dried (Na2S0 4) . Evaporation of all the volatiles gives tert-butyl 3-(4-((lH-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6- dioxopiperidine-l-carboxylate. (PCT Int. Appl., 2015087234, 18 Jun 2015) tert-Butyl 3-(4-((l-alkyl-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l- carboxylate t rt-butyl 3-(4-((1H-pyrazol-3-yl)oxy)-1,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine- 1- carboxylate ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halide (1.2 equiv.). The mixture is stirred at rt overnight. The reaction is poured into water and extracted with EtOAc. The organic layer is washed with water and brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography on silica gel to give t rt-butyl 3-(4-((l-alkyl- pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l-carboxylate.

4-((l-Alkyl-pyrazol-3-yl)oxy)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione

A solution of t rt-butyl 3-(4-((l-alkyl-pyrazol-3-yl)oxy)-l,3-dioxoisoindolin-2-yl)-2,6- dioxopiperidine-1 -carboxylate ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give 4-((l-alkyl- pyrazol-3-yl)oxy)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione.

General procedure to prepare N-aryl pomalidomide:

Example: N-Pyrazole pomalidomide Pd2(dba) , XantPhos Cs C0 , dioxane

tert-Butyl 3-(4-((l-(terf-butoxycarbonyl)-lH-pyrazol-3-yl)amino)-l,3- dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l-carboxylate

4-Chlorothalidomide ( 1 equiv.) and 3-amino-l-Boc-pyrazole (2 equiv.) are dissolved in dioxane. XantPhos (0.1 equiv.) and CS2CO3 (2.5 equiv.) are added and the mixture is degassed with nitrogen. Pd2(dba)3 (0.05 equiv.) is added and the mixture is sealed and heated at 80 °C overnight. The reaction is poured into water and extracted with EtOAc. The organic layer is washed with water and brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography on silica gel to give t rt-butyl 3-(4-((l -(t rt-butoxycarbonyl)-lH-pyrazol-3- yl)amino)-l,3-dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l-carboxylate.

4-((lH-Pyrazol-3-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione

A solution of t rt-butyl 3-(4-((l -(t r t-butoxycarbonyl)-lH-pyrazol-3-yl)amino)-l,3- dioxoisoindolin-2-yl)-2,6-dioxopiperidine-l-carboxylate ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give 4-((lH-pyrazol-3-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione. 4-((l-Alkyl-pyrazol-3-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione

4-((lH-pyrazol-3-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione ( 1 equiv.) is dissolved in DMF and K2CO3 (1.2 equiv.) is added, followed by alkyl halide ( 1 equiv.). The mixture is stirred at rt overnight. The reaction is poured into water and extracted with EtOAc. The organic layer is washed with water and brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography on silica gel to give 4-((l-alkyl-pyrazol-3-yl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione.

General procedure to prepare N-aryl lenalidomide:

Example: N-Pyridyl lenalidomide

N-Pyridyl-Boc-lenalidomide ref: OL-2003-147S

Under argon, substituted 2-chloropyridine ( 1 equiv.) and lenalidomide (1.1 equiv.) are loaded into oven-dried vials, followed by the addition of palladium catalyst ( 1 mol%), NaOtBu (1.1 equiv.) and dry dioxane. The reaction is run at 70 °C. After consumption of reactants or no further conversion, the reaction is quenched with aqueous H 4CI solution and extracted with

CH2CI2. The organic layer is dried over Na2S04 and concentrated. The residue is purified by column chromatography on silica gel to give N-pyridyl-Boc-lenalidomide. (Org. Lett. 2003, 5, 1479-1482)

N-Pyridyl-lenalidomide

A solution of N-pyridyl-Boc-lenalidomide ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give N-pyridyl-lenalidomide.

General procedure to prepare O-aryl lenalidomide:

Example: O-Pyridyl-lenalidomide Substituted 2-chloropyridine ( 1 equiv.), 4-hydroxylenalidomide ( 1 equiv.), and K3PO4 (2 equiv.) are introduced under argon in a Schlenk tube, equipped with a magnetic stirring bar. The [{Pd(n -allyl)^-Cl)}2]/ferrocenyl ligand (ratio 1:2, 0.2 mol% Pd) catalyst and toluene are added, and the Schlenk tube is purged several times with argon. The reaction is heated at 115 °C for 20 h . The solvent is removed and the residue is purified by column chromatography on silica gel to give O-pyridyl-Boc-lenalidomide. (Adv. Synth. Catal. 2011, 353, 3403 - 3414)

O-Pyridyl-lenalidomide

A solution of O-pyridyl-Boc-lenalidomide ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give O-pyridyl-lenalidomide. 5-Methylthalidomide derivatives:

4-Methyl-3-nitrophthalic anhydride

Oxidation of ruthenium dioxide to ruthenium tetraoxide is carried out with aqueous sodium hypochlorite (household bleach, 5.25 % concentration). Into a 3-L, three-necked flask fitted with mechanical stirrer and air condenser are added carbon tetrachloride, RuC L O, and bleach. The reaction is stirred until all the insoluble black ruthenium dioxide is converted into soluble yellow ruthenium tetraoxide. As the two-phase mixture is being stirred, l-nitro-2-methylnaphthalene ( 1 equiv.) is added. The reaction is allowed to stir until the black color of the ruthenium dioxide begins to persist. More bleach is added and stirring is continued. The reaction is then allowed to stir for about 20 h . The aqueous phase is separated from the organic layer, concentrated, and extracted with diethyl ether. The ether layer is dried (Na2S0 4) and concentrated. The residue is dissolved in ether and filtered, which leaves behind a large amount of extracted salts. The product in ether solution is again taken to complete dryness and dissolved in acetone. The acetone solution is concentrated and water is added to retain any remaining undesired salts. The 4-methyl-3- nitrophthalic acid product is allowed to precipitate from the acetone-water solution. Excess acetic anhydride is added to 4-methyl-3-nitrophthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely dissolved. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 4-methyl-3-nitrophthalic anhydride. (J. Agric. FoodChem. 1991, 39, 554-559)

5-Methyl-4-nitrothalidomide

A mixture of 4-methyl-3-nitrophthalic anhydride ( 1 equiv.), 3-aminopiperidine-2,6-dione

HCl salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 5- methyl-4-nitrothalidomide.

5-Methylpomalidomide

A mixture of 5-methyl-4-nitrothalidomide ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give 5-methylpomalidomide.

Example: N-Linked 5-methylpomalidomide derivatives

5-methylpomalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S0 4), and concentrated. The residue is purified by column chromatography to give N-linked 5-methylpomalidomide derivatives.

Example: O-Linked 5-methylthalidomide derivatives

15 N sulfuric acid is cooled to 0 °C, 5-methylpomalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the residue is purified by column chromatography to give 5-methyl-4-hydroxythalidomide. (J. Am. Chem. So , 1955, 77 (19), pp 5092-5095)

5-methyl-4-hydroxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give O-linked 5-methylthalidomide derivatives.

6-Methylthalidomide derivatives: 3-Fluoro-5-methylphthalic acid

1,4-Dioxane is added to a mixture of Pd(OAc) 2 (10 mol%), 2-fluoro-4-methylbenzoic acid

( 1 equiv.), Ag2C0 3 (2 equiv.) and NaOAc (2 equiv.) in a Schlenk tube containing a Hi-Vac valve. The Schlenk tube is degassed under high vacuum and filled with carbon monoxide at room temperature. The reaction mixture is heated at 130 °C with vigorous stirring. After 18-48 h, the reaction is cooled to room temperature, acidified with 2 N HCl and thoroughly extracted with ethyl acetate. The ethyl acetate fractions are combined, the solvent is removed in a rotary evaporator and the product is purified on a silica gel column to provide 3-fluoro-5-methylphthalic acid. (J. Am. Chem. Soc, 2008, 130 (43), pp 14082-14083)

3-Fluoro-5-methylphthalic anhydride Excess acetic anhydride is added to 3-fluoro-5-methylphthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely consumed. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 3-fluoro-5-methylphthalic anhydride. (J. Agric. Food Chem. 1991, 39, 554-559) 4-fluoro-6-methylthalidomide A mixture of 3-fluoro-5-methylphthalic anhydride ( 1 equiv.), 3-aminopiperidine-2,6-dione

HC1 salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- fluoro-6-methylthalidomide.

Example: N-Linked 6-methylpomalidomide derivatives

A mixture of 4-fluoro-6-methylthalidomide ( 1 equiv.), alkyl amine ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide N-linked 6- methylpomalidomide derivatives. (Chem Biol. 2015 Jun 18;22(6):755-63.)

Example: O-Linked 6-methylthalidomide derivatives

To a cooled solution of alcohol (1.2 equiv.) in DMF is added NaH (1.3 equiv.) and stirred at 0 °C for 30 min. 4-Fluoro-6-methylthalidomide ( 1 equiv.) was added and the mixture is heated at 80 °C overnight. The reaction is cooled and quenched with methanol. The mixture is poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide O-linked 6- methylthalidomide derivatives.

7-Methylthalidomide derivatives:

3-Fluoro-6-methylphthalic acid

1,4-Dioxane is added to a mixture of Pd(OAc) 2 (10 mol%), 2-fluoro-5-methylbenzoic acid

( 1 equiv.), Ag2CC (2 equiv.) and NaOAc (2 equiv.) in a Schlenk tube containing a Hi-Vac valve. The Schlenk tube is degassed under high vacuum and filled with carbon monoxide at room temperature. The reaction mixture is heated at 130 °C with vigorous stirring. After 18-48 h, the reaction is cooled to room temperature, acidified with 2 N HC1 and thoroughly extracted with ethyl acetate. The ethyl acetate fractions are combined, the solvent is removed in a rotary evaporator and the product is purified on a silica gel column to provide 3-fluoro-6-methylphthalic acid. (J. Am. Chem. Soc, 2008, 130 (43), pp 14082-14083)

3-Fluoro-6-methylphthalic anhydride Excess acetic anhydride is added to 3-fluoro-6-methylphthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely consumed. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 3-fluoro-6-methylphthalic anhydride. (J. Agric. FoodChem. 1991, 39, 554-559)

4-Fluoro-7-methylthalidomide

A mixture of 3-fluoro-6-methylphthalic anhydride ( 1 equiv.), 3-aminopiperidine-2,6-dione

HC1 salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- fluoro-7-methylthalidomide.

Example: N-linked 7-methylpomalidomide derivatives

A mixture of 4-fluoro-7-methylthalidomide ( 1 equiv.), alkyl amine ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide N-linked 7- methylpomalidomide derivatives. (Chem Biol. 2015 Jun 18;22(6):755-63.)

Example: O-Linked 7-methylthalidomide derivatives

To a cooled solution of alcohol (1.2 equiv.) in DMF is added NaH (1.3 equiv.) and stirred at 0 °C for 30 min. 4-Fluoro-7-methylthalidomide ( 1 equiv.) was added and the mixture is heated at 80 °C overnight. The reaction is cooled and quenched with methanol. The mixture is poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide O-linked 7- methylthalidomide derivatives.

5-Trifluoromethylthalidomide derivatives:

2-Fluoro-3,4-dimethyl-l-(trifluoromethyl)benzene An oven-dried vial is equipped with Ru(phen) 3 2 (1-2 mol%), dry K2HPO4 (3 equiv.), 1- fluoro-2,3-dimethylbenzene ( 1 equiv.) and MeCN (0.125 M), and degassed by alternating vacuum evacuation and argon backfill at -78 °C. The triflyl chloride (1-4 equiv.) is then added and the solution is stirred at room temperature adjacent to a 26-W compact fluorescent light bulb. After 24 h, the reaction mixture is purified by column chromatography on silica gel to furnish 2-fluoro-3,4- dimethyl-l-(trifluoromethyl)benzene and l-fluoro-2,3-dimethyl-4-(trifluoromethyl)benzene. The former one is taken to proceed to the next steps. {Nature 2011, 480, 224)

3-Fluoro-4-trifluoromethylphthalic acid

2-Fluoro-3,4-dimethyl-l-(trifluoromethyl)benzene ( 1 equiv.) is dissolved in acetic acid glacial (0.045 M). To this solution, sulfuric acid about 96% is added (1/5 volume of AcOH) and the mixture is cooled to approximately 15 °C in an ice bath, under stirring. Chromium(VI) trioxide (4.6 equiv.) is added stepwise within approximately 30 min. After the addition, the ice bath is removed and the mixture is allowed to warm to approximately room temperature and then is heated to about 35 °C for about 20 h . Further, the mixture is diluted approximately 1.5 fold with water and methanol is added cautiously in order to destroy the excess Cr0 3. The aqueous mixture is extracted with ethyl acetate (3 x) and the combined organic fractions are washed with water (2 x) and dried over Na2S04. After filtration, the solvent is evaporated completely under vacuum and the crude is recrystallized from toluene to provide 3-fluoro-4-trifluoromethylphthalic acid. (PCT Int. Appl., 2012061344, 10 May 2012)

3-fluoro-4-trifluoromethylphthalic anhydride

Excess acetic anhydride is added to 3-fluoro-4-trifluoromethylphthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely consumed. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 3-fluoro-4- trifluoromethylphthalic anhydride. (J. Agric. Food Chem. 1991, 39, 554-559)

4-Fluoro-5-trimethylfluorothalidomide

A mixture of 3-fluoro-4-trifluoromethylphthalic anhydride ( 1 equiv.), 3-aminopiperidine- 2,6-dione HCl salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4-fluoro-5-trimethylfluorothalidomide.

Example -Linked 5-trimethylfluoropomalidomide derivatives

A mixture of 4-fluoro-5-trimethylfluorothalidomide ( 1 equiv.), alkyl amine ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide N-linked 5- trimethylfluoropomalidomide derivatives. (Chem Biol. 2015 Jun 18;22(6):755-63.)

Example: O-Linked 5-trimethylfluorothalidomide derivatives To a cooled solution of alcohol (1.2 equiv.) in DMF is added NaH (1.3 equiv.) and stirred at 0 °C for 30 min. 4-fluoro-5-trimethylfluorothalidomide ( 1 equiv.) was added and the mixture is heated at 80 °C overnight. The reaction is cooled and quenched with methanol. The mixture is poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide O-linked 5- trimethylfluorothalidomide derivatives.

6-Trifluoromethylthalidomide derivatives:

In particular, a mixture of sulfuric acid about 96% (42 equiv.) and fuming nitric acid (14 equiv.) is given under stirring to l,2-dimethyl-4-(trifluoromethyl)benzene ( 1 equiv.) and heated to approximately 60 °C for about 3 h . After cooling to room temperature, the mixture is poured over crushed ice. The milky solution is then extracted with CH2CI2 (2 x). The combined organic fractions are washed with 3% Na2C03 solution (2 x) and then water (2 x). The organic solution is dried over N a2SC"4, filtered and evaporated in vacuo. The crude is purified by column chromatography on silica gel to give l,2-dimethyl-3-nitro-5-(trifluoromethyl)benzene. (PCT Int. Appl., 2012061344, 10 May 2012)

3-Nitro-5-trifluoromethylphthalic acid

l,2-Dimethyl-3-nitro-5-(trifluoromethyl)benzene ( 1 equiv.) is dissolved in acetic acid glacial (0.045 M). To this solution, sulfuric acid about 96% is added (1/5 volume of AcOH) and the mixture is cooled to approximately 15 °C in an ice bath, under stirring. Chromium(VI) tri oxide (4.6 equiv.) is added stepwise within approximately 30 min. After the addition, the ice bath is removed and the mixture is allowed to warm to approximately room temperature and then is heated to about 35 °C for about 20 h . Further, the mixture is diluted approximately 1.5 fold with water and methanol is added cautiously in order to destroy the excess CrC . The aqueous mixture is extracted with ethyl acetate (3 x) and the combined organic fractions are washed with water (2 x) and dried over N a2S0 4. After filtration, the solvent is evaporated completely under vacuum and the crude is recrystallized from toluene to provide 3-nitro-5-trifluoromethylphthalic acid. (PCT Int. Appl., 2012061344, 10 May 2012)

3-Nitro-5-trifluoromethylphthalic anhydride

Excess acetic anhydride is added to 3-nitro-5-trifluoromethylphthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely consumed. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 3-nitro-5-trifluoromethylphthalic anhydride. (J. Agric. FoodChem. 1991, 39, 554-559) 4-Nitro-6-trimethylfluorothalidomide

A mixture of 3-nitro-5-trifluoromethylphthalic anhydride ( 1 equiv.), 3-aminopiperidine- 2,6-dione HC1 salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4-nitro-6-trimethylfluorothalidomide.

4-Amino-6-trimethylfluorothalidomide

A mixture of 4-nitro-6-trimethylfluorothalidomide ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give 4-amino-6-trimethylfluorothalidomide.

Example: N-Linked 6-trimethylfluoropomalidomide derivatives

4-Amino-6-trimethylfluorothalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc.

The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give N-linked 6-trimethylfluoropomalidomide derivatives. Example -Linked 6-trifluoromethylthalidomide derivatives

15 N Sulfuric acid is cooled to 0 °C, 4-amino-6-trimethylfluorothalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the residue is purified by column chromatography to give 4-hydroxy-6-trimethylfluorothalidomide. (J. Am. Chem. Soc, 1955, 77 (19), pp 5092-5095) 4-Hydroxy-6-trimethylfluorothalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc.

The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give O-linked 6-trifluoromethylthalidomide derivatives.

7-Trifluoromethylthalidomide derivatives:

l-Fluoro-2,3-dimethyl-4-(trifluoromethyl)benzene An oven-dried vial is equipped with Ru(phen) 3 2 (1-2 mol%), dry K2HPO4 (3 equiv.), 1- fluoro-2,3-dimethylbenzene ( 1 equiv.) and MeCN (0.125 M), and degassed by alternating vacuum evacuation and argon backfill at -78 °C. The triflyl chloride (1-4 equiv.) is then added and the solution is stirred at room temperature adjacent to a 26-W compact fluorescent light bulb. After 24 h, the reaction mixture is purified by column chromatography on silica gel to furnish 2-fluoro-3,4- dimethyl-l-(trifluoromethyl)benzene and l-fluoro-2,3-dimethyl-4-(trifluoromethyl)benzene. The latter one is taken to proceed to the next steps. {Nature 2011, 480, 224)

3-Fluoro-6-trifluoromethylphthalic acid

l-Fluoro-2,3-dimethyl-4-(trifluoromethyl)benzene ( 1 equiv.) is dissolved in acetic acid glacial (0.045 M). To this solution, sulfuric acid about 96% is added (1/5 volume of AcOH) and the mixture is cooled to approximately 15 °C in an ice bath, under stirring. Chromium(VI) trioxide (4.6 equiv.) is added stepwise within approximately 30 min. After the addition, the ice bath is removed and the mixture is allowed to warm to approximately room temperature and then is heated to about 35 °C for about 20 h . Further, the mixture is diluted approximately 1.5 fold with water and methanol is added cautiously in order to destroy the excess Cr0 3. The aqueous mixture is extracted with ethyl acetate (3 x) and the combined organic fractions are washed with water (2 x) and dried over Na2S04. After filtration, the solvent is evaporated completely under vacuum and the crude is recrystallized from toluene to provide 3-fluoro-6-trifluoromethylphthalic acid. (PCT Int. Appl., 2012061344, 10 May 2012)

3-Fluoro-6-trifluoromethylphthalic anhydride Excess acetic anhydride is added to 3-fluoro-6-trifluoromethylphthalic acid. The mixture is heated at gentle reflux until the phthalic acid is completely consumed. Reflux is continued for an additional 30 min. The reaction mixture is concentrated in vacuo, and the excess acetic anhydride is allowed to evaporate under a gentle stream of dry air to give 3-fluoro-6- trifluoromethylphthalic anhydride. (J. Agric. Food Chem. 1991, 39, 554-559)

4-Fluoro-7-trimethylfluorothalidomide

A mixture of 3-fluoro-6-trifluoromethylphthalic anhydride ( 1 equiv.), 3-aminopiperidine- 2,6-dione HCl salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4-fluoro-7-trimethylfluorothalidomide.

Example: -Linked 7-trimethylfluoropomalidomide derivatives

A mixture of 4-fluoro-7-trimethylfluorothalidomide ( 1 equiv.), alkyl amine ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide N-linked 7- trimethylfluoropomalidomide derivatives. (Chem Biol. 2015 Jun 18;22(6):755-63.)

Example: -Linked 7-trimethylfluorothalidomide derivatives

To a cooled solution of alcohol (1.2 equiv.) in DMF is added NaH (1.3 equiv.) and stirred at 0 °C for 30 min. 4-fluoro-7-trimethylfluorothalidomide ( 1 equiv.) was added and the mixture is heated at 80 °C overnight. The reaction is cooled and quenched with methanol. The mixture is poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide O-linked 7- trimethylfluorothalidomide derivatives.

5,6,7-Hydroxythalidomide/pomalidomide: 5,6,7-hydroxypomalidomide and intermediates were reported in Celgene patent (WO- 2014018866-A1

5,6,7-Alkoxythalidomide/pomalidomide Starting materials are found in Celgene patent (WO-2014018866-A1) N-Linked 5-alkoxypomalidomide N-Linked 7-alkoxypomalidomide

O-Linked 7-alkoxythalidomide

Example: N-Linked 5-alkoxypomalidomide derivatives 5-Alkoxy-4-nitrothalidomide

5-Hydroxy-4-nitrothalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give 5-alkoxy-4-nitrothalidomide.

4-Amino-5-alkoxythalidomide

A mixture of 5-alkoxy-4-nitrothalidomide ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give 4-amino-5-alkoxythalidomide.

N-Linked 5-alkoxypomalidomide derivatives

4-Amino-5-alkoxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give N-linked 5-alkoxypomalidomide derivatives.

Example: O-Linked 5-alkoxythalidomide derivatives 4-Hydroxy-5-methoxythalidomide

15 N Sulfuric acid is cooled to 0 °C, 4-amino-5-methoxythalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the residue is purified by column chromatography to give 4-hydroxy-5-methoxythalidomide.

4-Alkoxy-5-methoxythalidomide

4-Hydroxy-5-methoxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give 4-alkoxy-5-methoxythalidomide.

4-Alkoxy-5-hydroxythalidomide

To a solution of 4-alkoxy-5-methoxythalidomide ( 1 equiv.) in DCM is added BBn (10 equiv.). The mixture is stirred at rt for 12 h . The reaction is quenched by adding DCM and water. The organic layer is separated, dried over sodium sulfate, filtered, and concentrated. The residue is purified by column chromatography to give 4-alkoxy-5-hydroxythalidomide.

O-linked 5-alkoxythalidomide derivatives

4-Alkoxy-5-hydroxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give O-linked 5-alkoxythalidomide derivatives.

Thal/Pom/Len analogs with variation on the glutarimide moiety a-fluoro, methyl, deutero-substituted thalidomide/pomalidomide

neral scheme:

Example: N-linked a-D pomalidomide derivatives A mixture of a-D N-Boc-4-nitrothalidomide (known compound reported in BMCL, 2003, 13, 3415) ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give a-D N-Boc-pomalidomide.

N-linked a-D pomalidomide derivatives

a-D N-Boc-pomalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S0 4), and concentrated. The residue is purified by column chromatography to give a-D N-Boc-pomalidomide derivatives. A solution of a-D N-Boc-pomalidomide derivatives ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give N-linked a-D pomalidomide derivatives.

Example: O-linked a-D thalidomide derivatives 15 N Sulfuric acid is cooled to 0 °C, a-D N-Boc-pomalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the residue is purified by column chromatography to give a-D N-Boc-4-hydroxythalidomide.

O-linked a-D thalidomide derivatives

a-D N-Boc-4-hydroxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give a-D N-Boc-4-hydroxythalidomide derivatives. A solution of a-D N-Boc-4-hydroxythalidomide derivatives ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give O-linked a-D thalidomide derivatives. Example: N-linked a-F omalidomide derivatives

a-F N-Boc-pomalidomide (known compound reported in BMCL, 2003, 13, 3415) ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give a-F N-Boc-pomalidomide derivatives. A solution of a-F N-Boc-pomalidomide derivatives ( 1 equiv.) in TFA/DCM ( 1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give N-linked a-F pomalidomide derivatives. Example: O-linked a-F thalidomide derivatives

a-F N-Boc-4-hydroxythalidomide

15 N sulfuric acid is cooled to 0 °C, a-F N-Boc-pomalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated and the residue is purified by column chromatography to give a-F N-Boc-4-hydroxythalidomide.

O-linked a-F thalidomide derivatives

a-F N-Boc-4-hydroxythalidomide ( 1 equiv.) is dissolved in DMF and K2CO3 (2 equiv.) is added, followed by alkyl halides (1.2 equiv.). The mixture is stirred at rt until the consumption of the starting material. The mixture is diluted with water and extracted with EtOAc. The organic layer is washed with brine, dried (Na2S04), and concentrated. The residue is purified by column chromatography to give a-F N-Boc-4-hydroxythalidomide derivatives. A solution of a-F N-Boc-4-hydroxythalidomide derivatives ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give O-linked a-F thalidomide derivatives.

Example: N-linked a-methylpomalidomide derivatives

To a stirred solution of 4-fluorothalidomide (1.0 equiv.) in DMF is added potassium carbonate (2.0 equiv.). The resulting solution is cooled to 0-5 °C and Boc anhydride (3.0 equiv.) is added slowly as a solution in 1,4-dioxane (also cooled). The resulting mixture is stirred at 0 °C for 1 h and allowed to warm to room temperature overnight. Water is then added and the aqueous layer is extracted 2 times with DCM and concentrated. The residue is purified by column chromatography on silica gel to obtain N-Boc 4-fluorothalidomide.

N-Boc 4-fluoro-a-methylthalidomide

To a solution of N-Boc 4-fluorothalidomide ( 1 equiv.) in DMF is added sodium hydride (2 equiv.) slowly at 0 °C. The mixture is stirred at 0 °C for 30 min and methyl iodide (1.1 equiv.) is added dropwise. The mixture is stirred at rt until the consumption of the starting material and then quenched with methanol. The mixture is diluted with saturated NaHCCb, extracted with EtOAc, dried (Na2S04), and concentrated. The residue is purified by column chromatography on silica gel to obtain N-Boc 4-fluoro-a-methylthalidomide.

N-linked a-methylpomalidomide derivatives

A mixture of N-Boc 4-fluoro-a-methylthalidomide ( 1 equiv.), alkyl amine ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to provide N-Boc a- methylpomalidomide derivatives (ref: Chem Biol. 2015 Jun 18;22(6):755-63.) A solution of N-Boc a-methylpomalidomide derivatives ( 1 equiv.) in TFA/DCM ( 1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give N-linked α-methylpomalidomide derivatives.

5'-Hydroxyl, alkoxy, amino, sulfonamide substituted glutarimide General scheme:

Example: 5'-Hydroxyl substituted pomalidomide derivatives

5'-ferf-Butyldimethylsilyloxy-4-fluorothalidomide

A mixture of 3-amino-5-((ter t-butyldimethylsilyl)oxy)piperidine-2,6-dione (known compound reported in Chem. Pharm. Bull. 2006, 54, 1709) ( 1 equiv.), 3-aminopiperidine-2,6- dione HCl salt ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S0 4) and concentrated to provide 5'-tert-butyldimethylsilyloxy-4-fluorothalidomide. 5'-ferf-butyldimethylsilyloxy-pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 5'-t r t-butyldimethylsilyloxy-4-fluorothalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 5'-ter t-butyldimethylsilyloxy-pomalidomide derivatives. {ChemBiol. 2015 Jun 18;22(6):755-63.)

5'-hydroxyl pomalidomide derivatives

To a solution of 5'-t r t-butyldimethylsilyloxy-pomalidomide derivatives ( 1 equiv.) in dry THF is added TBAF (1.1 equiv.) at 0 °C, and the reaction mixture is stirred at 0 °C for 40 min, and then at room temperature for 2 h . CHCh is added, and the mixture is washed with 0.5 M HCl and brine, dried over N a2SC"4, filtered and concentrated. The crude product is purified by flash column chromatography on silica gel to provide 5'-hydroxyl pomalidomide derivatives. (Chem. Pharm. Bull. 2006, 54, 1709)

Example: 5'-alkoxy substituted pomalidomide derivatives To a solution of 5'-hydroxyl pomalidomide derivatives ( 1 equiv.) and alkyl halide (2 eq.) in Et20 (0.05 M) under a nitrogen atmosphere is added Ag20 (2 equiv.) at room temperature. The resulting suspension is stirred at dark at room temperature for 16 h then filtered through a pad of

Celite using Et20 as eluent. The residue is concentrated in vacuo and purified by column chromatography on silica gel to provide 5'-alkoxy pomalidomide derivatives. (Org. Lett. 2013, 75, 5878)

Example: 5'-Amino substituted pomalidomide derivatives

5'-Azido pom lidomide derivatives

5'-Hydroxy pomalidomide derivatives ( 1 equiv.) is dissolved in anhydrous 1,4-dioxane under nitrogen. 1.4 M n-butyllithium hexane solution (5 equiv.) diluted with 1,4-dioxane is added, and stirred for 10 minutes at room temperature. A solution of p-toluene sulfonyl chloride in 1,4- dioxane is added and stirred for 1 hour. After reaction completion, it is quenched with aqueous ammonia chloride, and extracted with EtOAc. The organic layer is dried (Na2S04) and concentrated to provide 5'-tosyloxy-pomalidomide derivatives, which are used in next reaction without further purification. (Jpn. Kokai Tokkyo Koho, 2009215195, 24 Sep 2009) To a stirred solution of 5'-tosyloxy-pomalidomide derivatives ( 1 equiv.) in anhydrous DMF at 70

°C under a N2 atmosphere is added NaN3 (5 equiv.). The mixture is then allowed to stir at 70 °C for 6 h, and then quenched with cool H20 and extracted with EtOAc (2 x). The combined organic extracts are washed with cool H2O and brine, dried (Na2S04), and concentrated and purified by flash column chromatography on silica gel to provide 5'-azido pomalidomide derivatives. (Synthesis 2013, 45, 3383-3386) 5'-amino pomalidomide derivatives

A mixture of azide ( 1 equiv.) and Pd/C 10% w/w in MeOH is stirred under hydrogen for 8 h . The mixture is then filtered and concentrated to give the crude amine, which is purified by flash column chromatography on silica gel to give 5'-amino pomalidomide derivatives.

Example: 5'-sulfonamide substituted pomalidomide

To a solution of 5'-amino pomalidomide derivatives ( 1 equiv.), triethylamine (2 equiv.) in chloroform is added sulfonyl chloride (1.1 equiv.). The reaction is stirred till the consumption of the starting material. The mixture is then extracted with EtOAc, washed with water and brine, dried (Na2S04), and concentrated. The residue is purified by flash column chromatography on silica gel to give 5'-sulfonamide pomalidomide derivatives.

5',5'-Difluoro substituted glutarimide Example: 5',5'-difluoro substituted pomalidomide derivatives

N-Boc-4,4-difluoropyroglutamic acid

To a stirred solution of 4,4-difluoropyroglutamic acid (known compound reported in Org. Lett. 1999, 1, 2105) ( 1.0 equiv.) in DMF is added Boc anhydride (2.0 equiv.), followed by DMAP (0.1 equiv.). The resulting mixture is stirred at room temperature overnight. Water is then added and the aqueous layer is extracted 2 times with DCM and concentrated. The residue is purified by column chromatography on silica gel to obtain N-Boc-4,4-difluoropyroglutamic acid.

N-Boc-4,4-difluoropyroglutamide

To a solution of N-Boc-4,4-difluoropyroglutamic acid ( 1 equiv.) in dry CH3CN is added EDCI HCl (1.2 equiv.) and HOBT (1.2 equiv.) at 0 °C. The mixture is stirred at room temperature for 6 h, then cooled to 0 °C, and 28% aqueous ammonia (excess) is carefully added. Stirring is continued at 0 °C for 30 min and then at room temperature for 1 h . Insoluble material is removed by filtration, then the filtrate was concentrated and purified by flash column chromatography on silica gel to provide N-Boc-4,4-difluoropyroglutamide. (Chem. Pharm. Bull. 2006, 54, 1709)

3-N-Boc-amino-5,5-difluoroglutarimide

A solution of N-Boc-4,4-difluoropyroglutamide ( 1 equiv.) in dry THF is stirred at 0 °C, and 1.0 M aqueous LiOH (3 equiv.) is carefully added. The reaction mixture is stirred at 0 °C for 3 h, then acidified by addition of 2.0 M aqueous HC1, and extracted 3 times with CHCh. The organic layer is washed with brine, dried over Na2S04, filtered and concentrated to afford 5-amino- 4-(N-Boc-amino)-2,2-difluoro-5-oxopentanoic acid, which is used directly for the next step without further purification.

To a solution of 5-amino-4-(N-Boc-amino)-2,2-difluoro-5-oxopentanoic acid ( 1 equiv.) in dry DCM is added CDI (1.15 equiv.), DMAP (0.1 equiv.), and DIPEA (2 equiv.) at 0 °C. The mixture is stirred at room temperature for 5 d, then washed with 0.5 M aqueous HC1 and brine, dried over Na2S0 4, filtered and concentrated. The residue was purified by flash column chromatography on silica gel to provide 3-N-Boc-amino-5,5-difluoroglutarimide. (Chem. Pharm. Bull. 2006, 54, 1709) 3-amino-5,5-difluoroglutarimide

A solution of 3-N-Boc-amino-5,5-difluoroglutarimide ( 1 equiv.) in TFA/DCM (1:1) is stirred at rt overnight. Solvent is evaporated and the residue is purified by column chromatography on silica gel to give 3-amino-5,5-difluoroglutarimide.

5',5'-Difluoro-4-fluorothalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 3-amino-5,5-difluoroglutarimide ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated

NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 5',5'- difluoro-4-fluorothalidomide.

5',5'-Difluoro pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 5',5'-difluoro-4-fluorothalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 5',5'-difluoro pomalidomide derivatives. {Chem Biol. 2015, 22, 755-763)

Oxetano glutarimide 2'-Oxetanothalidomide/lenalidomide were reported by Erick M . Carreira {Org. Lett. 2013, 75, 4312)

OL-2013-4312

Example: 6'-oxetanopomalidomide Ethyl 3-(3-((ferf-butylsulfinyl)amino)oxetan-3-yl)propiolate

ref: L-2 - 6

Known compound and procedure reported in Org. Lett. 2010, 72, 1116.

2-Methyl-N-(3-(3-oxopropyl)oxetan-3-yl)propane-2-sulfinamide

To a solution of ethyl 3-(3 -((t rt-butylsulfinyl)amino)oxetan-3-yl)propiolate ( 1 equiv.) in

EtOAc is added Pt0 2/C (0.1 wt equiv.). The reaction vessel is purged three times with hydrogen and heated under hydrogen until the consumption of starting material. Upon cooling to ambient temperature, the reaction mixture is filtered through Celite® and concentrated to give ethyl 3-(3- ((tert-butylsulfinyl)amino)oxetan-3-yl)propanoate, which is used directly without further purifications. The ester obtained above is dissolved in THF, and cooled to -78 °C. DIBAL (1.0 equiv.) is added carefully while monitoring the conversion of the starting material. When considerable amount of aldehyde is formed, the reaction is quenched with saturated ammonium chloride, extracted with EtOAc, washed with brine, dried over sodium sulfate, and concentrated. The aldehyde is used directly or purified by column chromatography. N-(3-(3-amino-3-cyanopropyl)oxetan-3-yl)-2-methylpropane-2-sulfinamide

To a mixture of magnesium sulfate (0.5 equiv.), ammonium chloride (0.5 equiv.), and sodium cyanide ( 1 equiv.) is added 7 M ammonia in methanol (4 equiv.). The mixture is stirred and cooled to 0 °C. After 10 min, the resulting suspension is added to aldehyde ( 1 equiv.). The mixture is stirring for 1 h at 0 °C, then increased to RT and continued to react for 4 h . The solvent is then removed under reduced pressure while maintaining the internal temperature <30 °C until nearly all of the methanol and ammonia are removed. The resulting slurry of inorganic salts and the product is diluted with MTBE, stirred at RT for 30 min, and filtered. The inorganic wet cake is washed with MTBE, and the solvent is removed under reduced pressure to provide N-(3-(3- amino-3-cyanopropyl)oxetan-3-yl)-2-methylpropane-2-sulfinamide. (ACS Med. Chem. Lett. 2014, 5, 1152-1 155).

2-Amino-4-(3-aminooxetan-3-yl)butanoic acid

N-(3-(3-Amino-3-cyanopropyl)oxetan-3-yl)-2-methylpropane-2-sulfinamide ( 1 equiv.) is dissolve in 12 M HC1 (excess) and stirred for 48 hours at 100 °C in a closed vial. Solvent is then removed in vacuo to provide 2-amino-4-(3-aminooxetan-3-yl)butanoic acid. (Angew. Chem. Int. Ed. 2014, 53, 557)

-amino-2-oxa-5-azaspiro [3.5] nonan-6-one

A mixture of 2-amino-4-(3-aminooxetan-3-yl)butanoic acid ( 1 equiv.), alumina (3 wt. equiv.) in toluene is heated under reflux for 1.5 h . The water produced during the reaction is collected in a Dean-Stark trap. The reaction mixture is allowed to cool and the alumina is filtered off and washed with 10% MeOH/C C . The filtration is combined and the solvent is removed under vacuum to provide 7-amino-2-oxa-5-azaspiro[3.5]nonan-6-one. (Org. Biomol. Chem. 2015, 13, 7624-7627)

4-fluoro-6'-oxetano-thalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 7-amino-2-oxa-5-azaspiro[3.5]nonan- 6-one ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- fluoro-6 ' -oxetano-thali domide.

6'-oxetano-pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 4-fluoro-6'-oxetano-thalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 6'-oxetano- pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

6'-Sulfonyl glutarimide ref: PCT int. Appl., 2 SS37, 0 Feb 2000 Example: 6'-sulfonyl pomalidomide derivatives

Ethyl 2-nitro-4-(phenoxysulfonyl)butanoate

A mixture of ethyl 2-nitroacetate (2 equiv.), potassium carbonate ( 1 equiv.) and 18-crown- 6 (0.5 equiv.) in toluene is heated to 90 °C and phenyl vinylsulfonate ( 1 equiv.) in toluene is added dropwise over one hour. The mixture is heated for 16 hours, cooled to ambient temperature and diluted with ethyl acetate. The mixture is washed with saturated brine. The organic layer is separated and the aqueous layer is extracted with ethyl acetate. The combined organic layers are dried (Na2S04), filtered and concentrated in vacuo. The residue is purified by column chromatography on silica gel to give ethyl 2-nitro-4-(phenoxysulfonyl)butanoate. (PCT Int. Appl., 2000006537, 10 Feb 2000)

2-nitro-4-(phenoxysulfonyl)butanoic acid

A mixture of 2-nitro-4-(phenoxysulfonyl)butanoate ( 1 equiv.), 1 M NaOH (excess) in methanol is stirred at rt for 2 h . The reaction is neutralized with 1 M HCl and concentrated to provide 2-nitro-4-(phenoxysulfonyl)butanoic acid. phenyl 4-amino-3-nitro-4-oxobutane-l-sulfonate

To a solution of 2-nitro-4-(phenoxysulfonyl)butanoic acid ( 1 equiv.) in dry CH3CN is added EDCI HCl (1.2 equiv.) and HOBT (1.2 equiv.) at 0 °C. The mixture is stirred at room temperature for 6 h, then cooled to 0 °C, and 28% aqueous ammonia (excess) is carefully added. Stirring is continued at 0 °C for 30 min and then at room temperature for 1 h . Insoluble material is removed by filtration, then the filtrate was concentrated and purified by flash column chromatography on silica gel to provide phenyl 4-amino-3-nitro-4-oxobutane-l -sulfonate. (Chem. Pharm. Bull. 2006, 54, 1709)

4-nitro-l,2-thiazinan-3-one 1,1-dioxide

To a 0 °C solution of 1.0 M THF solution of potassium t rt-butoxide (2 equiv.) in THF is added a solution of 4-amino-3-nitro-4-oxobutane-l -sulfonate ( 1 equiv.) in THF dropwise over 30 minutes. After stirring at 0 °C for 2 h, the cooling bath is removed and stirring continued for 30 minutes. The mixture is diluted with water and extracted two times with ethyl ether. The aqueous portion is acidified to pH 2 with 10% aqueous sodium bisulfate and extracted four times with 20 mL each of ethyl acetate. The combined ethyl acetate layers are dried (Na2S04), filtered and concentrated in vacuo. The residue is purified by column chromatography on silica gel to give 4- nitro-l,2-thiazinan-3-one 1,1-dioxide. (PCT Int. Appl., 2000006537, 10 Feb 2000)

4-Amino-l,2-thiazinan-3-one 1,1-dioxide

A mixture of 4-nitro-l,2-thiazinan-3-one 1,1-dioxide ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give 4-amino-l,2-thiazinan-3-one 1,1- dioxide.

4-Fluoro-6'-sulfonyl-thalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 4-amino-l,2-thiazinan-3-one 1,1- dioxide ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4- fluoro-6'-sulfonyl-thalidomide.

6'-Sulfonyl pomalidomide derivatives A mixture of alkyl amine ( 1 equiv.), 4-fluoro-6'-sulfonyl-thalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 6'-sulfonyl pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

'-Sulfonyl glutarimide

Example: 2'-sulfonyl pomalidomide derivatives

Phenyl nitromethanesulfonate To a dry THF solution containing phenyl methanesulfonate ( 1 equiv.) at -10 °C under a nitrogen atmosphere is added dropwise 1.6 M n-BuLi in hexane (2.4 equiv.). The mixture is stirred for 10 min, and then cooled to -30 °C and isoamylnitrate (2.2 equiv.) is then added dropwise. After stirring for 4 h at the same temperature, the reaction is quenched with water, and the volatiles are evaporated in vacuo. 1 M NaOH (2 equiv.) is added to the reaction mixture and the mixture is washed with ethyl ether. The aqueous layer is acidified with acetic acid (pH=5), and extracted with

CH2CI2. The organic layer is washed with brine and dried over anhydrous Na2S04, filtered, and concentrated. The residue is purified by column chromatography on silica gel to provide phenyl nitromethanesulfonate. (Bioorg. Med. Chem. 2000, 8, 2167-2173)

Phenyl 4-amino-l-nitro-4-oxobutane-l-sulfonate

A mixture of phenyl nitromethanesulfonate ( 1 equiv.), potassium carbonate ( 1 equiv.) and 18-crown-6 (0.5 equiv.) in toluene is stirred and acrylamide ( 1 equiv.) is added dropwise. The mixture is stirred for 16 hours, and diluted with ethyl acetate. The mixture is washed with saturated brine. The organic layer is separated and the aqueous layer is extracted with ethyl acetate. The combined organic layers are dried (Na2S04), filtered and concentrated in vacuo. The residue is purified by column chromatography on silica gel to give phenyl 4-amino-l-nitro-4-oxobutane-l- sulfonate.

6-Nitro-l ,2-thiazin -3-one 1,1-dioxide

To a 0 °C solution of 1.0 M THF solution of potassium t rt-butoxide (2 equiv.) in THF is added a solution of phenyl 4-amino-l-nitro-4-oxobutane-l -sulfonate ( 1 equiv.) in THF dropwise over 30 minutes. After stirring at 0 °C for 2 h, the cooling bath is removed and stirring continued for 30 minutes. The mixture is diluted with water and extracted two times with ethyl ether. The aqueous portion is acidified to pH 2 with 10% aqueous sodium bisulfate and extracted four times with 20 mL each of ethyl acetate. The combined ethyl acetate layers are dried (Na2S04), filtered and concentrated in vacuo. The residue is purified by column chromatography on silica gel to give 6-nitro-l,2-thiazinan-3-one 1,1-dioxide. (PCT Int. Appl., 2000006537, 10 Feb 2000)

6-amino-l,2-thiazinan-3-one 1,1-dioxide

A mixture of 6-nitro-l,2-thiazinan-3-one 1,1-dioxide ( 1 equiv.) and palladium on activated carbon (10% wt) in THF is hydrogenated until the disappearance of the starting material. The mixture is filtered through Celite and concentrated to give 6-amino-l,2-thiazinan-3-one 1,1- dioxide.

4-Fluoro-2'-sulfonyl-thalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 6-amino-l,2-thiazinan-3-one 1,1- dioxide ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S0 4) and concentrated to provide 4- fluoro-2 '-sulfony1-thalidomide.

2'-Sulfonyl pomalidomide derivatives A mixture of alkyl amine ( 1 equiv.), 4-fluoro-2'-sulfonyl-thalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 2'-sulfonyl pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

Oxetano sulfonamide glutarimide derivatives

Example: 6'-oxetano-2'-sulfonyl pomalidomide derivatives Sodium l-amino-3-(3-((ieri-butylsulfinyl)amino)oxetan-3-yl)propane-l-sulfonate

ref: ACS-20 4- 52

A solution of sodium bisulfite ( 1 equiv.) in water is slowly added to the solution of 2- methyl-N-(3-(3-oxopropyl)oxetan-3-yl)propane-2-sulfinamide (preparation was mentioned above) ( 1 equiv.) in methanol. The mixture is stirred for 60 min at RT, and then the solvent is removed to provide sodium 3-(3-((t rt-butylsulfinyl)amino)oxetan-3-yl)-l-hydroxypropane-l- sulfonate, which is used for the next step without further purification. An aqueous solution of ammonia (30%, 1 equiv.) is added to a solution of sodium 3-(3-

((tert-butylsulfinyl)amino)oxetan-3-yl)-l-hydroxypropane-l -sulfonate ( 1 equiv.) in MeOH:H20 1:1, and the mixture is stirred at room temperature. After 1h, the reaction is concentrated to provide sodium l-amino-3 -(3 -((t rt-butylsulfinyl)amino)oxetan-3-yl)propane-l -sulfonate without further purification. (AC Med. Chem. Lett. 2014, 5, 1152) l-Amino-3-(3-aminooxetan-3-yl)propane-l-sulfonic acid

Sodium 1-amino-3 -(3 -((t rt-butylsulfinyl)amino)oxetan-3 -yl)propane- 1-sulfonate (1 equiv.) is dissolve in 12 M HC1 (excess) and stirred at rt until the consumption of the starting material. Solvent is then removed in vacuo to provide l-amino-3 -(3 -aminooxetan-3-yl)propane-l- sulfonic acid. 7-amino-2-oxa-6-thia-5-azaspiro[3.5]nonane 6,6-dioxide

l-Amino-3 -(3 -aminooxetan-3-yl)propane-l -sulfonic acid ( 1 equiv.) is dissolved in POCh (diluted) and stirred at 100 °C until the consumption of the starting material. Solvent is removed in vacuo and the residue is dissolved in DCM and washed carefully with saturated sodium carbonate, then brine. The organic layer is dried (Na2S04), concentrated, and purified by column chromatography on silica gel to give 7-amino-2-oxa-6-thia-5-azaspiro[3.5]nonane 6,6-dioxide. 4-fluoro-6'-oxetano-2'-sulfonyl-thalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 7-amino-2-oxa-6-thia-5- azaspiro[3.5]nonane 6,6-dioxide ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in

DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4-fluoro-6'-oxetano-2'-sulfonyl-thalidomide.

6'-Oxetano-2'-sulfonyl pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 4-fluoro-6'-oxetano-2'-sulfonyl-thalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 6'-oxetano-2'-sulfonyl pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

Example: 2'-oxetano-6'-sulfonyl pomalidomide derivatives

Phenyl 3-nitropropane-l-sulfonate

ref: PCT Int. AppL, 2008050830, 18 Va y 2006

A mixture of phenyl ethenesulfonate ( 1 equiv.) and nitromethane ( 1 equiv.) is heated to 80 °C and, at 80 °C, ethyldiisopropylamine (2 equiv.) is added dropwise. The mixture is stirred at 80 °C till the consumption of starting materials. Cooling to RT is followed by addition of a 2 M aqueous HCl solution and extraction 3 times with EtOAc. Drying over N a2S04 and removal of the solvent in vacuo are followed by coevaporation 3 times with toluene to provide phenyl 3- nitropropane-1 -sulfonate. (PCT Int. AppL, 2006050830, 18 May 2006)

phenyl 3-nitro-3-(oxetan-3-ylidene)propane-l-sulfonate

To phenyl 3-nitropropane-l-sulfonate (1.0 equiv.) is added oxetan-3-one (1.3 equiv.) and

Et3N (0.2 equiv.), and the mixture is stirred at RT for 75 min. Then it is dissolved in CH2CI2 and the solution is cooled to -78 °C. Et3N (3.0 equiv.) is added followed by MsCl (2.5 equiv.), and the mixture is stirred at -78 °C for 30 min, when it is allowed to warm to -25 °C over ca. 45 min. After stirring at RT for 20 min, it is quenched with HCl (0. 1 M in H2O). The resulting mixture is diluted with CH2CI2, and the phases are separated. The aqueous phase is extracted with CH2CI2, and the combined organic layers are dried (Na2S0 4), filtered, and concentrated in vacuo. Purification of the residue by column chromatography on silica gel provides phenyl 3-nitro-3- (oxetan-3-ylidene)propane-l -sulfonate. (Org. Lett. 2013, 75, 4312).

Phenyl 3-(3-((4-methoxybenzyl)amino)oxetan-3-yl)-3-nitropropane-l-sulfonate

To a cooled (0 °C) solution of phenyl 3-nitro-3-(oxetan-3-ylidene)propane-l -sulfonate (1.0 equiv.) in THF is added 4-methoxybenzylamine (1.0 equiv.), and the mixture is stirred at 0 °C for 30 min. Then it is allowed to warm to RT and stirring is continued for another 10 min. At this point the mixture is concentrated in vacuo and the residue is purified by by column chromatography on silica gel to provide phenyl 3-(3-((4-methoxybenzyl)amino)oxetan-3-yl)-3-nitropropane-l- sulfonate. (Org. Lett. 2013, 75, 4312).

Phenyl 3-(hydroxyimino)-3-(3-((4-methoxybenzyl)amino)oxetan-3-yl)propane-l-sulfonate

To a solution of phenyl 3-(3-((4-methoxybenzyl)amino)oxetan-3-yl)-3-nitropropane-l- sulfonate (1.0 equiv.) in THF is added at RT tetrabutylammonium iodide (0.05 equiv.), benzyl bromide (1.2 equiv.), and potassium hydroxide (1.2 equiv.). The mixture is stirred atRT for 3.5 h .

Then it is diluted with Et20 and washed with saturated aqueous NaCl. Then it is dried (Na2S0 4), filtered, and concentrated in vacuo. The residue is purified by column chromatography on silica gel to provide phenyl 3-(hydroxyimino)-3-(3-((4-methoxybenzyl)amino)oxetan-3-yl)propane-l- sulfonate as a mixture of oxime isomers. (Org. Lett. 2013, 75, 4312).

9-(Hydroxyimino)-5-(4-methoxybenzyl)-2-oxa-6-thia-5-azaspiro[3.5]nonane 6,6-dioxide A solution of phenyl 3-(hydroxyimino)-3-(3-((4-methoxybenzyl)amino)oxetan-3- yl)propane-l -sulfonate (mixture of oxime isomers; 1.0 equiv.) in xylenes is heated to 140 °C and stirred at that temperature for 23 h . Then it is cooled to RT and concentrated in vacuo. The residue is purified by column chromatography on silica gel to provide 9-(hydroxyimino)-5-(4- methoxybenzyl)-2-oxa-6-thia-5-azaspiro[3.5]nonane 6,6-dioxide. (Org. Lett. 2013, 15, 4312).

9-Amino-5-(4-methoxybenzyl)-2-oxa-6-thia-5-azaspiro [3.5] nonane 6,6-dioxide

Raney-Ni (50% slurry in H2O) is washed with EtOH (3 x), then is added a solution of 9- (hydroxyimino)-5-(4-methoxybenzyl)-2-oxa-6-thia-5-azaspiro[3.5]nonane 6,6-dioxide (1.0 equiv.) in EtOH, and the mixture is diluted with EtOH. A hydrogen atmosphere (balloon) is built up, and the mixture is vigorously stirred at RT for 9 h . Then the atmosphere is changed to N 2 and the mixture is filtered over Celite and washed with MeOH. The filtrate is then concentrated in vacuo. The crude product is dissolved in CH2CI2 and filtered again over Celite. The filtrate is concentrated in vacuo to afford 9-amino-5-(4-methoxybenzyl)-2-oxa-6-thia-5- azaspiro[3.5]nonane 6,6-dioxide. (Org. Lett. 2013, 15, 4312).

4-Fluoro-2'-oxetano-6'-sulfonyl-thalidomide

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 9-amino-5-(4-methoxybenzyl)-2-oxa- 6-thia-5-azaspiro[3.5]nonane 6,6-dioxide ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried

(Na2S0 4) and concentrated to provide 4-fluoro-2'-oxetano-6'-sulfonyl-thalidomide.

2'-Oxetano-6'-sulfonyl pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 4-fluoro-2'-oxetano-6'-sulfonyl-thalidomide ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. (Chem Biol. 2015, 22, 755-763) To a solution of the resulting residue (1.0 equiv) in CH3CN, cooled to 0 °C, is added a solution of CAN (3.0 equiv) in H2O, and the yellow clear solution is stirred at 0 °C for 2 h . Then it is diluted with EtOAc and quenched with half saturated aqueous NaCl and diluted with EtOAc and H2O. The phases are separated, and the aqueous phase is extracted with EtOAc (2 χ ) and

CH2CI2 (2 x). The combined organic phases are dried (Na2S0 4), filtered, and concentrated in vacuo. The residue is purified by column chromatography on silica gel to provide 2'-oxetano-6'- sulfonyl pomalidomide derivatives. (Org. Lett. 2013, 75, 4312)

7-Membered Imides A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 3-aminoazepan-2-one ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 4-fluoro-2-(2-oxoazepan- 3-yl)isoindoline- 1,3-dione . 2-(2,7-Dioxoazepan-3-yl)-4-fluoroisoindoline-l,3-dione

A solution of 4-fluoro-2-(2-oxoazepan-3-yl)isoindoline-l,3-dione ( 1 equiv.), manganic acetylacetonate (0.01 wt equiv.), and 68.8% t-butyl hydroperoxide (1.5 equiv.) is stirred at rt for 4 d . The mixture is concentrated and purified by column chromatography on silica gel to provide 2- (2,7-dioxoazepan-3-yl)-4-fluoroisoindoline-l,3-dione. (J. Org. Chem. 1970, 35, 2121)

7-membered pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 2-(2,7-dioxoazepan-3-yl)-4-fluoroisoindoline-l,3- dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give 7-membered pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

[3.1.0] System Example: [3.1.0] pomalidomide derivatives

ref: BC-20 4 S

A solution of t rt-butyl ethyl malonate ( 1 equiv.) in dry MeCN is added to N- chlorosuccinimide (1.2 equiv.) and magnesium perchlorate (0.3 equiv.). The reaction mixture is stirred for 4 hours. After the solvent is removed on a rotary evaporator, the mixture is diluted with

EtOAc and washed with brine. The organic layers are dried with Na2S04, and concentrated in vacuo. The residue is purified by column chromatography on silica gel to afford l -(t rt-butyl) 3- ethyl 2-chloromalonate. (Org. Biomol. Chem. 2014, 72, 1510). l-(ferf-Butyl) 1,2-diethyl cyclopropane-l,l 2-tricarboxylate

ref: J C - 991 -1329

To a stirred slurry of NaH (60% in mineral oil, 1 equiv.) in anhydrous Et20 is added 1- (t r t-butyl) 3-ethyl 2-chloromalonate ( 1 equiv.) and ethyl acrylate ( 1 equiv.) during which the temperature is maintained between 25 and 30 °C. The mixture is stirred at room temperature for

24 h, and then unreacted NaH is decomposed with methanol. H20 is added, and the organic layer is separated, dried over Na2S0 4, filtered, and concentrated. The residue is purified by column chromatography on silica gel to afford l -(t r t-butyl) 1,2-diethyl cyclopropane- 1,1,2- tricarboxylate. (J. Med. Chem. 1981, 24, 481) l-(ferf-Butoxycarbonyl)cyclopropane-l,2-dicarboxylic acid

A mixture of l -(t r t-butyl) 1,2-diethyl cyclopropane-l,l,2-tricarboxylate ( 1 equiv.) and

NaOH (2 equiv.) in 1:1 EtOH-H 20 is stirred at rt for 6 h and then is evaporated to one-half volume.

The aqueous solution is extracted with Et20 , chilled in ice, and then made acidic with 1 N HC1. Crystalline product is collected by filtration and is recrystallized to give the \-{tert- butoxycarbonyl)cyclopropane- 1,2-dicarboxylic acid.

3-(4-Methoxybenzyl)-2,4-dioxo-3-azabicyclo[3.1.0]hexane-l-carboxylic acid

ref: JMC-1 991 -1329

A solution of l -(t r t-butoxycarbonyl)cyclopropane-l,2-dicarboxylic acid ( 1 equiv.) in xylene (isomeric mixture) is boiled under a water trap for 5 h and cooled. To this solution of the intermediary anhydride formed during the reaction is added a solution of PMBNH 2 ( 1 equiv.) in xylene. The mixture is boiled for a further 15 h and evaporated to dryness. (J. Med. Chem. 1991, 34, 1333). The residue is dissolved in 1:1 TFA/DCM and stirred at rt for 2 h . Solvents are evaporated to provide 3-(4-methoxybenzyl)-2,4-dioxo-3-azabicyclo[3.1.0]hexane-l-carboxylic acid. tert-Butyl (3-(4-methoxybenzyl)-2,4-dioxo-3-azabicyclo[3.1.0]hexan-l-yl)carbamate

ref: JOC-2005-61 8 To a solution of 3-(4-methoxybenzyl)-2,4-dioxo-3-azabicyclo[3.1.0]hexane-l-carboxylic acid ( 1 equiv.) in toluene is added triethylamine (1.2 equiv.). Diphenylphosphoryl azide (1.1 equiv.) is added dropwise and the mixture is heated to reflux while stirring for 30 min. The mixture is then cooled to rt and quenched with saturated aqueous H 4CI. After separation of the two phases, the aqueous solution is extracted with Et20 (3 x) and the combined organic layers are washed with brine, dried with anhydrous Na2S0 4 and concentrated in vacuo. The crude product is purified by flash chromatography on silica gel to afford isocyanate. (J. Org. Chem. 2005, 70, 6 118) To a solution of the isocyanate (1.0 equiv. of 0.2 M in benzene) is added the t-butyl alcohol (1.5 equiv). Titanium tetratbutoxide (0.10 equiv.) is then added, and the mixture is stirred at 120 °C until completion. After quenching with saturated aqueous H 4CI and separating the two phases, the aqueous layer is extracted three times with CH2C 12, and the combined organic layers are dried with anhydrous Na2S0 4, and concentrated in vacuo. The residue is purified by flash chromatography on silica gel to afford t rt-butyl (3-(4-methoxybenzyl)-2,4-dioxo-3- azabicyclo[3.1.0]hexan-l-yl)carbamate. (J. Org. Chem. 2005, 70, 6 118) l-Amino-3-(4-methoxybenzyl)-3-azabicyclo[3.1.0]hexane-2,4-dione A solution of tert-b y (3-(4-methoxybenzyl)-2,4-dioxo-3-azabicyclo[3.1.0]hexan-l- yl)carbamate ( 1 equiv.) in 1:1 TFA/DCM is stirred at rt for 2 h . Solvents are evaporated to provide l-amino-3-(4-methoxybenzyl)-3-azabicyclo[3.1.0]hexane-2,4-dione.

2-(2,4-dioxo-3-azabicyclo[3.1.0]hexan-l-yl)-4-fluoroisoindoline-l,3-dione

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), l-amino-3-(4-methoxybenzyl)-3- azabicyclo[3.1.0]hexane-2,4-dione ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in

DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S0 4) and concentrated to provide 4-fluoro adduct. To a solution of the resulting residue (1.0 equiv) in CH3CN, cooled to 0 °C, is added a solution of CAN (3.0 equiv) in H2O, and the yellow clear solution is stirred at 0 °C for 2 h . Then it is diluted with EtOAc and quenched with half saturated aqueous NaCl and diluted with EtOAc and H2O. The phases are separated, and the aqueous phase is extracted with EtOAc (2 χ ) and

CH2CI2 (2 x). The combined organic phases are dried (Na2S0 4), filtered, and concentrated in vacuo. The residue is purified by column chromatography on silica gel to provide 2-(2,4-dioxo-3- azabicyclo[3.1.0]hexan-l-yl)-4-fluoroisoindoline-l,3-dione. (Org. Lett. 2013, 15, 4312)

[3.1.0] pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 2-(2,4-dioxo-3-azabicyclo[3.1.0]hexan-l-yl)-4- fluoroisoindoline-l,3-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give [3 .1.0] pomalidomide derivatives. {Chem Biol. 2015, 22, 755- 763)

[4.1.0] System

ref: PCT t. AppL, 20 0007032, 2 1 Jan 201 ref: JOC-2GQ5-61

Example: [4.1.0] pomalidomide derivatives

Methyl 2-oxo-3-azabicyclo [4.1.0]heptane- 1-carboxylate

ref: PCT Int. AppL, 20 0007032, 2 1 Jan 201 0 Known compound and procedure reported in PCT Int. Appl., 2010007032, 2 1 Jan 2010. tert-Butyl (2-oxo-3-azabicyclo[4.1.0]heptan-l-yl)carbamate

ref: JOC-20D5-611 8

A mixture of methyl 2-oxo-3-azabicyclo[4. 1.0]heptane-l-carboxylate ( 1 equiv.) and LiOH (2 equiv.) in 1:1 MeOH-H20 is stirred at rt for 6 h and then is evaporated to one-half volume. The aqueous solution is extracted with Et20 , chilled in ice, and then made acidic with 1 N HC1. Crystalline product is collected by filtration to give the 2-oxo-3-azabicyclo[4.1.0]heptane-l- carboxylic acid.

To a solution of 2-oxo-3-azabicyclo[4.1.0]heptane-l-carboxylic acid ( 1 equiv.) in toluene is added triethylamine (1.2 equiv.). Diphenylphosphoryl azide (1.1 equiv.) is added dropwise and the mixture is heated to reflux while stirring for 30 min. The mixture is then cooled to rt and quenched with saturated aqueous H 4CI. After separation of the two phases, the aqueous solution is extracted with Et20 (3 x) and the combined organic layers are washed with brine, dried with anhydrous Na2S04 and concentrated in vacuo. The crude product is purified by flash chromatography on silica gel to afford isocyanate. (J. Org. Chem. 2005, 70, 6 118) To a solution of the isocyanate (1.0 equiv. of 0.2 M in benzene) is added the t-butyl alcohol (1.5 equiv). Titanium tetratbutoxide (0.10 equiv.) is then added, and the mixture is stirred at 120 °C until completion. After quenching with saturated aqueous H 4CI and separating the two phases, the aqueous layer is extracted three times with CH2CI2, and the combined organic layers are dried with anhydrous Na2S04, and concentrated in vacuo. The residue is purified by flash chromatography on silica gel to afford t r t-butyl (2-oxo-3-azabicyclo[4.1.0]heptan-l- yl)carbamate. (J. Org. Chem. 2005, 70, 6 118) tert-Butyl (2,4-dioxo-3-azabicyclo[4.1.0]heptan-l-yl)carbamate ref: JOC-1 70-2 2 1

A solution of t r t-butyl (2-oxo-3-azabicyclo[4.1.0]heptan-l-yl)carbamate ( 1 equiv.), manganic acetyl acetonate (0.01 wt equiv.), and 68.8% t-butyl hydroperoxide (1.5 equiv.) is stirred at rt for 4 d . The mixture is concentrated and purified by column chromatography on silica gel to provide t rt-butyl (2,4-dioxo-3-azabicyclo[4.1.0]heptan-l-yl)carbamate. (J. Org. Chem. 1970, 35, 2121) l-Amino-3-azabicyclo[4.1.0]heptane-2,4-dione

A solution of t rt-butyl (2,4-dioxo-3-azabicyclo[4.1.0]heptan-l-yl)carbamate ( 1 equiv.) in

1:1 TFA/DCM is stirred at rt for 2 h . Solvents are evaporated to provide l-amino-3- azabicyclo[4. 1.0]heptane-2,4-dione.

2-(2,4-Dioxo-3-azabicyclo[4.1.0]heptan-l-yl)-4-fluoroisoindoline-l,3-dione

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), l-amino-3-azabicyclo[4.1.0]heptane- 2,4-dione ( 1 equiv.), and potassium acetate (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 2- (2,4-dioxo-3-azabicyclo[4.1.0]heptan-l-yl)-4-fluoroisoindoline-l,3-dione.

[4.1.0] pomalidomide derivatives A mixture of alkyl amine ( 1 equiv.), 2-(2,4-dioxo-3-azabicyclo[4.1.0]heptan-l-yl)-4- fluoroisoindoline-l,3-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The crude residue is purified by column chromatography on silica gel to give [4. 1.0] pomalidomide derivatives. (Chem Biol. 2015, 22, 755- 763)

[3.1.1] System

N-Acryloyl-2-fluoro-N-(4-methoxybenzyl)acrylamide ref: J C- 99 - 329

A solution of 2-fluoroacryloyl chloride ( 1 equiv.) is cooled to 0 °C and added dropwise to a precooled solution of N-(4-methoxybenzyl)acrylamide ( 1 equiv.) and triethylamine (2 equiv.) in methylene chloride. The mixture is stirred for 1.5 h at room temperature and evaporated to dryness. The residue is extracted with ether, the filtrate is evaporated. The crude residue is purified by column chromatography on silica gel to give N-acryloyl-2-fluoro-N-(4- methoxybenzyl)acrylamide. J . Med. Chem. 1991, 34, 1333). l-Fluoro-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione

ref: JMC-1 991 -1329

A solution of N-acryloyl-2-fluoro-N-(4-methoxybenzyl)acrylamide ( 1 equiv.) and 2,6-di- t rt-butyl-p-cresionl (13.75 equiv.) in 1,2-dichlorbenzene is stirred at 170 °C for 1.5 h . After concentration by evaporation, the residue is purified by column chromatography on silica gel to give l-fluoro-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione. l-amino-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione

ref: Tetrahedron, 201 3, 10357-1 0360 Under nitrogen atmosphere, a mixture of l-fluoro-3-(4-methoxybenzyl)-3- azabicyclo[3.1.1]heptane-2,4-dione ( 1 equiv.), sodium azide (1.2 equiv.), and Al(OTf)3 (0.3 equiv.) in dry CH2CI2 is stirred under reflux for 12 h . The mixture is poured into water and extracted with CH2CI2 (3 x). The combined organic layers are dried over Na2S0 4 and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel to give 1- azido-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione. The azide obtained above is dissolved in methanol and added palladium on activated carbon (10% wt). The mixture is hydrogenated until the consumption of the starting material. After filtration and concentration, the residue is purified by column chromatography on silica gel to give l-amino-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione. l-(4-fluoro-l,3-dioxoisoindolin-2-yl)-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4- dione

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), l-amino-3-(4-methoxybenzyl)-3- azabicyclo[3.1.1]heptane-2,4-dione (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated

NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide l-(4-fluoro- l,3-dioxoisoindolin-2-yl)-3-(4-methoxybenzyl)-3-azabicyclo[3.1.1]heptane-2,4-dione.

[3.1.1] Pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), l-(4-fluoro-l,3-dioxoisoindolin-2-yl)-3-(4- methoxybenzyl)-3-azabicyclo[3 .1.l]heptane-2,4-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. {Chem Biol. 2015, 22, 755-763) To a solution of the resulting residue (1.0 equiv) in CH3CN, cooled to 0 °C, is added a solution of CAN (3.0 equiv) in H2O, and the yellow clear solution is stirred at 0 °C for 2 h . Then it is diluted with EtOAc and quenched with half saturated aqueous NaCl and diluted with EtOAc and H2O. The phases are separated, and the aqueous phase is extracted with EtOAc (2 χ ) and

CH2CI2 (2 x). The combined organic phases are dried (Na2S0 4), filtered, and concentrated in vacuo. The residue is purified by column chromatography on silica gel to provide [3.1.1] pomalidomide derivatives. {Org. Lett. 2013, 75, 4312)

[3.2.1] System

Example: [3.2.1] pomalidomide derivatives

(lR,4S)-2-Oxobicyclo[2.2.1]heptane-l-carboxylic acid 4. Jones oxidation

ref: JOC-1 993-3668

Known compound and procedure reported in J. Org. Chem. 1993, 58, 3668. tert-Butyl ((lR,4S)-2-oxobicyclo[2.2.1]heptan-l-yl)carbamate

To a solution of (lR,4S)-2-oxobicyclo[2.2.1]heptane-l-carboxylic acid ( 1 equiv.) in toluene is added triethylamine (1.2 equiv.). Diphenylphosphoryl azide (1.1 equiv.) is added dropwise and the mixture is heated to reflux while stirring for 30 min. The mixture is then cooled to rt and quenched with saturated aqueous H 4CI. After separation of the two phases, the aqueous solution is extracted with Et20 (3 x) and the combined organic layers are washed with brine, dried with anhydrous Na2S0 4 and concentrated in vacuo. The crude product is purified by flash chromatography on silica gel to afford isocyanate. (J. Org. Chem. 2005, 70, 6 118) To a solution of the isocyanate (1.0 equiv. of 0.2 M in benzene) is added the t-butyl alcohol (1.5 equiv). Titanium tetratbutoxide (0.10 equiv.) is then added, and the mixture is stirred at 120 °C until completion. After quenching with saturated aqueous H 4CI and separating the two phases, the aqueous layer is extracted three times with CH2C 12, and the combined organic layers are dried with anhydrous Na2S0 4, and concentrated in vacuo. The residue is purified by flash chromatography on silica gel to afford t rt-butyl ((lR,4S)-2-oxobicyclo[2.2.1]heptan-l- yl)carbamate. (J. Org. Chem. 2005, 70, 6 118) tert-Butyl (2,4-dioxo-3-azabicyclo[3.2.1]octan-l-yl)carbamate ref: TA-2 0-3 3

t rt-Butyl ((lR,4S)-2-oxobicyclo[2.2.1]heptan-l-yl)carbamate ( 1 equiv.) is mixed with selenium dioxide (1.1 equiv.), dissolved in acetic acid and refluxed for 18 h . The reaction mixture is filtered, concentrated to smaller volume and 30% hydrogen peroxide (excess) is added. After cooling to 0 °C the reaction is filtered, dried and crystallized from toluene/hexane to provide anhydride. The resulting anhydride ( 1 equiv.) is dissolved in 25% aqueous ammonia (excess) and evaporated to dryness. The residue is heated at 180 °C for 0.5 h, dissolved in chloroform, decolorized with silica gel, filtered and the solvent is evaporated. The residue is purified by flash chromatography on silica gel to afford t rt-butyl (2,4-dioxo-3-azabicyclo[3.2.1]octan-l- yl)carbamate. {Tetrahedron:Asymmetry 2000, 11, 3113-3 122)

l-Amino-3-azabicyclo [3.2. 1]octane-2,4-dione

A solution of tert-butyl (2,4-dioxo-3-azabicyclo[3.2.1]octan-l-yl)carbamate ( 1 equiv.) in 1:1 TFA/DCM is stirred at rt for 2 h . Solvents are evaporated to provide l-amino-3- azabicyclo[3.2.1]octane-2,4-dione.

2-(2,4-Dioxo-3-azabicyclo[3.2.1]octan-l-yl)-4-fluoroisoindoline-l,3-dione

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), l-amino-3-azabicyclo[3.2.1]octane- 2,4-dione (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 2-(2,4-dioxo-3- azabicyclo[3 .2. 1Joctan- 1-yl)-4-fluoroisoindoline- 1,3-dione.

[3.2.1] Pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 2-(2,4-dioxo-3-azabicyclo[3.2.1]octan-l-yl)-4- fluoroisoindoline-l,3-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel to give [3.2.1] pomalidomide derivatives. {ChemBiol. 2015, 22, 755- 763)

4'-Spiroglutarimide

Example: 4'-spiro pomalidomide derivatives 4-Bromo-6-azaspiro[2.5]octane-5,7-dione

Bromine ( 1 equiv.) is added to a solution of 6-azaspiro[2.5]octane-5,7-dione ( 1 equiv.) in

1, 1,2-trichloroethane at room temperature. The mixture is allowed to stir for 2 hours at 110 °C and then for 1 hour at room temperature. The reaction mixture is concentrated in vacuo and the resulting residue is purified by flash column chromatography on silica gel to provide 4-bromo-6- azaspiro[2.5]octane-5,7-dione. (PCT Int. Appl., 2013033901, 14 Mar 2013)

4-amino-6-azaspiro [2.5] octane-5,7-dione

4-Bromo-6-azaspiro[2.5]octane-5,7-dione ( 1 equiv.) is dissolved in DMF and NaN 3 (3 equiv.) is added. The resulting mixture is heated at 50 °C for 3 h . Solvent is removed in vacuo and the crude azide is dried under vacuum. (U.S. Pat. Appl. Publ., 20040006062, 08 Jan 2004) The crude azide is dissolved in methanol and palladium on activated carbon (10% wt) is added. The mixture is hydrogenated at rt until the consumption of starting material. The mixture is filtered, and concentrated. The residue is purified by flash column chromatography on silica gel to provide 4-amino-6-azaspiro[2.5]octane-5,7-dione.

2-(5,7-Dioxo-6-azaspiro[2.5]octan-4-yl)-4-fluoroisoindoline-l,3-dione A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 4-amino-6-azaspiro[2.5]octane-5,7- dione (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 2-(5,7-dioxo-6-azaspiro[2.5]octan-4- yl)-4-fluoroisoindoline- 1,3-dione.

4'-spiro pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 2-(5,7-dioxo-6-azaspiro[2.5]octan-4-yl)-4- fluoroisoindoline-l,3-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel to give 4'-spiro pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763)

5'-Spiroglutarimide Example: 5'-spiro pomalidomide derivatives

Di-terf-butyl 4-methylene-5-oxopyrrolidine-l,2-dicarboxylate

ref: TL-2004-3241

Known compound and procedure reported in TetrahedronLett. 2004, 45, 3241. methyl l-(3-(ter t-butoxy)-2-((t rt-butoxycarbonyl)amino)-3-oxopropyl)cyclopropane-l- carboxylate ref: J. Chem. Soc, Perkin Trans. 1, 1997, 351 9

Known compound and procedures reported in J . Chem. Soc, Perkin Trans. 1, 1997, 3519.

2-Amino-3-(l-carbamoylcyclopropyl)propanoic acid

To a stirred solution of methyl l-(3-(ter t-butoxy)-2-((t rt-butoxycarbonyl)amino)-3- oxopropyl)cyclopropane-l-carboxylate ( 1 equiv.) in methanol at 0 °C in a 2-5 mL microwave vial is added magnesium nitride (5 equiv.) in a single portion. The vial is sealed immediately and allowed to warm to room temperature in a water bath. After approximately 1 hour the reaction is heated at 80 °C for 24 h . The reaction is allowed to cool to room temperature and diluted with chloroform and water. The aqueous layer is neutralized with 3 N HC1 and the organic layer is separated. The aqueous layer is further extracted with chloroform (2 x) and the organic layers are combined, passed through a hydrophobic frit and concentrated in vacuo to afford crude carboxamide. (Org. Lett. 2008, 10, 3623) The resulting carboxamide ( 1 equiv.) is dissolved in 1:1 TFA/DCM and stirred at rt for 2 h . Solvents are evaporated and the residue is purified by column chromatography on silica gel to provide 2-amino-3-(l-carbamoylcyclopropyl)propanoic acid.

7-Amino-5-azaspiro [2.5] octane-4,6-dione

A mixture of 2-amino-3-(l-carbamoylcyclopropyl)propanoic acid ( 1 equiv.), alumina (3 wt. equiv.) in toluene is heated under reflux for 1.5 h . The water produced during the reaction is collected in a Dean-Stark trap. The reaction mixture is allowed to cool and the alumina is filtered off and washed with 10% MeOH/C C . The filtration is combined and the solvent is removed under vacuum to provide 7-amino-5-azaspiro[2.5]octane-4,6-dione. (Org. Biomol. Chem. 2015, 13, 7624-7627)

2-(4,6-Dioxo-5-azaspiro[2.5]octan -7-yl)-4-fluoroisoindoline-l,3-dione

A mixture of 3-fluorophthalic anhydride ( 1 equiv.), 7-amino-5-azaspiro[2.5]octane-4,6- dione (2.5 equiv.) in acetic acid is stirred at 120 °C overnight. The dark mixture is cooled and filtered. The filter cake is dissolved in DCM and washed with saturated NaHCCb and brine. The organic layer is dried (Na2S04) and concentrated to provide 2-(4,6-dioxo-5-azaspiro[2.5]octan-7- yl)-4-fluoroisoindoline- 1,3-dione.

5'-Spiro pomalidomide derivatives

A mixture of alkyl amine ( 1 equiv.), 2-(4,6-dioxo-5-azaspiro[2.5]octan-7-yl)-4- fluoroisoindoline-l,3-dione ( 1 equiv.) and DIEA (2 equiv.) in dry DMF is stirred at 90 °C for 12 h . The mixture is cooled to room temperature, poured into water and extracted with ethyl acetate. The combined organic phases are washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel to give 5'-spiro pomalidomide derivatives. (Chem Biol. 2015, 22, 755-763) Intermediate functionalization in preparation for linker installation General procedure to convert 4-fluoro-thalidomide to 4-bromo-thalidomide, which then be used to convert to various functional groups for linker installation.

JMC- 90- 645 Y = OH, e y yi, propargyioxy, COOH, CH OH, CHO, CH Br, CH General procedure to convert lenalidomide to 4-bromo-lenalidomide, which then be used to convert to various functional groups for linker installation.

JMC-1 990- 45 Y = OH, thy y , propargyioxy, COOH, CH OH, CHO, CH Br, C 3 Intermediate functionalization Exa 5'-spiro-pomalidomide

N-Boc 4-fluoro-5'-spiro-thalidomide

To a stirred solution of 4-fluoro-5'-spiro-thalidomide (1.0 equiv.) in DMF is added potassium carbonate (2.0 equiv.). The resulting solution is cooled to 0-5 °C and Boc anhydride (3.0 equiv.) is added slowly as a solution in 1,4-dioxane (also cooled). The resulting mixture is stirred at 0 °C for 1 h and allowed to warm to room temperature overnight. Water is then added and the aqueous layer is extracted 2 times with DCM and concentrated. The residue is purified by column chromatography on silica gel to obtain N-Boc 4-fluoro-5'-spiro-thalidomide. (WO 2014/147531 A2) N-Boc 4-benzylamino-5'-spiro-thalidomide

A mixture containing N-Boc 4-fluoro-5'-spiro-thalidomide ( 1 equiv.) and benzylamine (2 equiv.) in CH3CN is stirred at room temperature until the consumption of the starting material. After concentration of the reaction mixture under reduced pressure, the residue is diluted with water and extracted with CHCh. The organic layer is dried and concentrated to dryness under reduced pressure. The residue is purified by flash column chromatography on silica gel to provide

N-Boc 4-benzylamino-5'-spiro-thalidomide. {J. Med. Chem. 1990, 33, 1645)

N-Boc 4-amino-5-spiro-thalidomide

A mixture containing N-Boc 4-benzylamino-5'-spiro-thalidomide ( 1 equiv.) and 5% Pd/C (0.1 wt equiv.) in dioxane is hydrogenated at 50-55 °C until the required volume of H 2 has been taken up. The reaction mixture is filtered to remove the catalyst. The filtrate is concentrated under reduced pressure to provide N-Boc 4-amino-5'-spiro-thalidomide. (J. Med. Chem. 1990, 33, 1645)

Intermediate functionalization Example 2 : N-Boc 4-bromo-5'-spiro-thalidomide

To a suspension of N-Boc 4-amino-5'-spiro-thalidomide ( 1 equiv.) in 20% HBr is added N aNC (1.2 equiv.) under ice cooling. After stirring for 5 min, the reaction mixture is added to a solution of CuBr (1.5 equiv.) in 20% HBr. The reaction mixture is stirred at room temperature for 1 h . After neutralization with 20% NaOH, the solution is extracted with CHCh. The organic layer is dried and concentrated to dryness. The residue is purified by flash column chromatography on silica gel to provide N-Boc 4-bromo-5'-spiro-thalidomide. (J. Med. Chem. 1990, 33, 1645) Intermediate functionalization Example 3 : N-Boc 4-hydroxyl-5'-spiro-thalidomide

15 N sulfuric acid is cooled to 0 °C, N-Boc 4-bromo-5'-spiro-thalidomide ( 1 equiv.) is added gradually with good stirring. After one hour, a solution of sodium nitrite ( 1 equiv.) in water is added slowly while maintaining the temperature below 5 °C. After another 30 minutes, the mixture is warmed to 80 °C and maintained at this temperature until nitrogen evolution ceases. The now dark orange solution is diluted with water and extracted with ether. The organic layer is dried with sodium sulfate, filtered and shaken for two hours with finely powdered barium chloride to remove traces of sulfuric acid. The solution is filtered and concentrated. The residue is purified by flash column chromatography on silica gel to provide N-Boc 4-hydroxyl-5'-spiro-thalidomide. (J. Am. Chem. Soc, 1955, 77 (19), pp 5092-5095)

Intermediate functionalization Example 4: N-Boc 4-ethynyl-5'-spiro-thalidomide

A reaction vessel is charged with bis(triphenylphosphine)palladium(II) chloride (2 mol%), copper(I) iodide (4 mol%) and N-Boc 4-bromo-5'-spiro-thalidomide ( 1 equiv.). The reaction atmosphere is cycled between nitrogen and vaccum 3 times then triethylamine (1.55 equiv.) and trimethylsilylacetylene (1.25 equiv.) are added and the reaction is mixed for 24 hours. When the starting materials are consumed, the reaction is diluted with ethyl acetate and filtered through a plug of celite. The filtrate is concentrated and the residue is purified by silica gel chromatography to provide N-Boc 4-trimethylsilylethynyl-5'-spiro-thalidomide. {Org. Lett. 2014, 6, 6302) A reaction vessel is charged with N-Boc 4-trimethylsilylethynyl-5'-spiro-thalidomide ( 1 equiv.), potassium carbonate (4 equiv.) and MeOH. The reaction is mixed at ambient temperature for 8 hours then concentrated. The residue is diluted with water and ethyl acetate. The aqueous layer is extracted with ethyl acetate and the combined organic layer is dried over sodium sulfate, filtered and concentrated. The crude residue is purified by silica gel chromatography to provide N-Boc 4-ethynyl-5 '-spiro-thalidomide.

Intermediate functionalization Example 5: N-Boc 4-propargyloxy-5'-spiro-thalidomide

A reaction vessel is charged with N-Boc 4-hydroxyl-5' -spiro-thalidomide ( 1 equiv.) and acetone (0.25 M). To this solution is added sequentially potassium carbonate (4 equiv.) and propargyl bromide (1.2 equiv.). The reaction is refluxed overnight, cooled to ambient temperature, filtered through a medium frit, and concentrated. The crude residue is purified by silica gel chromatography to provide N-Boc 4-propargyloxy-5' -spiro-thalidomide. (J. Med. Chem. 2013, 56, 2828)

Intermediate functionalization Example 6: N-Boc 4-carboxy-5'-spiro-thalidomide

A flame-dried reaction vessel is charged with N-Boc 4-bromo-5' -spiro-thalidomide ( 1 equiv.) and the atmosphere is cycled between nitrogen and vacuum three times. Ether is added and the solution is cooled to -78 °C. t rt-Butyllithium (2 equiv.) is added dropwise and the reaction is mixed for 15 min then carbon dioxide gas is bubbled through the solution for 15 min. The reaction is warmed to ambient temperature allowing excess carbon dioxide gas to slowly evolve from solution. The reaction is quenched with 1 M aq. NaOH solution and washed with ether (2 x). The pH of the aqueous layer is adjusted to 3 and extracted with ethyl acetate (3 x). The combined organic layer is dried over sodium sulfate and concentrated to dryness with toluene (3 x) to provide N-Boc 4-carboxy-5 '-spiro-thalidomide.

Intermediate functionalization Example 7: N-Boc 4-hydroxymethyl-5'-spiro-thalidomide A reaction vessel is charged with N-Boc 4-carboxy-5'-spiro-thalidomide ( 1 equiv.), THF and cool to 0 °C. Triethylamine (1.1 equiv.) and isobutylchloroformate (1.1 equiv.) are added and the reaction mixed ambient temperature for 1 hour. The reaction is filtered through a medium frit and cooled to 0 °C. To the solution of mixed anhydride is added a solution of sodium borohydride (2 equiv.) in MeOH. Upon complete reduction to the corresponding benzylic alcohol, the reaction is concentrated then treated with ethyl acetate and 10% aq. HC1. The phases are separated and aqueous solution is extracted with ethyl acetate (3 x). The combined organic layer is washed with 5% sodium bicarbonate solution, dried over sodium sulfate, and concentrated. The residue is purified by silica gel chromatography to provide N-Boc 4-hydroxymethyl-5'-spiro-thalidomide.

Intermediate functionalization Example 8: N-Boc 4-formyl-5'-spiro-thalidomide

A reaction vessel is charged with N-Boc 4-hydroxymethyl-5'-spiro-thalidomide ( 1 equiv.) and manganese dioxide (10 equiv.) and DCM. The reaction is heated at reflux overnight then cooled to ambient temperature and filtered. The filtrate is concentrated and purified by silica gel chromatography to provide N-Boc 4-formyl-5'-spiro-thalidomide.

Intermediate functionalization Example 9: N-Boc 4-bromomethyl-5'-spiro-thalidomide

A reaction vessel is charged with N-Boc 4-hydroxymethyl-5'-spiro-thalidomide ( 1 equiv.) and DCM. The solution is cooled to 0 °C and N-bromosuccinimide (1.25 equiv.) and triphenylphosphine (1.25 equiv.) are then added. The reaction is mixed for 3 hours then concentrated. The crude residue is purified by silica gel chromatography to provide N-Boc 4- bromomethyl-5'-spiro-thalidomide. (J. Med. Chem. 2015, 58, 1215)

Intermediate functionalization Example 10: N-Boc 4-azidomethyl-5'-spiro-thalidomide

Sodium azide (3 equiv.) is added to a solution of N-Boc 4-bromomethyl-5'-spiro- thalidomide ( 1 equiv.) in water and acetone (1:3, 0.25 M). The reaction is heated at 60 °C for 6 hours. The reaction is cooled to ambient temperature and the solvent removed by rotary evaporation. The aqueous layer is extracted with DCM (3 x) and the combined organic layer is dried over sodium sulfate and filtered. The filtrate is concentrated and the crude residue is purified by silica gel chromatography to provide N-Boc 4-azidomethyl-5'-spiro-thalidomide. (Angew. Chem. Int. Ed. 2014, 53, 10155)

Linker Installation

Linker Installation Example 1: r -Butyl 7-(4-((8-hydroxyoctyl)oxy)-l,3-dioxoisoindolin-2- yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate

A reaction vessel is charged with N-Boc 4-hydroxyl-5'-spiro-thalidomide ( 1 equiv.) and

DMF (0.3 M) then cooled to 0 °C. Sodium hydride (60% dispersion in mineral oil, 1.1 equiv.) is added and the reaction is warmed to ambient temperature and mixed for 1 hour. The reaction is cooled to 0 °C then 8-bromooctan-l-ol (1.1 equiv.) is added and the reaction is mixed at ambient temperature overnight. DMF is removed by rotary evaporation and the residue is deposited onto silica gel and purified by silica gel chromatography to provide t rt-butyl 7-(4-((8- hydroxyoctyl)oxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate. Linker Installation Example 2 : r -Butyl 7-(4-(2-(2-(2-hydroxyethoxy)ethoxy)ethoxy)-l,3- dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate

A reaction vessel is charged with N-Boc 4-hydroxyl-5'-spiro-thalidomide ( 1 equiv.) and

DMF (0.3 M) then cooled to 0 °C. Sodium hydride (60% dispersion in mineral oil, 1.1 equiv.) is added and the reaction is warmed to ambient temperature and mixed for 1 hour. The reaction is cooled to 0 °C then 2-(2-(2-bromoethoxy)ethoxy)ethan-l-ol (1.1 equiv.) is added and the reaction is mixed at ambient temperature overnight. DMF is removed by rotary evaporation and the residue is deposited onto silica gel and purified by silica gel chromatography to provide t rt-butyl 7-(4-(2- (2-(2-hydroxyethoxy)ethoxy)ethoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane- 5-carboxylate.

Linker Installation Example 3 : tert- y 7-(4-((l-(3-hydroxypropyl)-lH-l,2,3-triazol-4- yl)methoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate

A reaction vessel is charged with the polymer supported catalyst (Amberlyst A-21, 1.23 mmol/g; Cul, 13 mol%). The azide (0.5 M in DCM, 1 equiv.) is added dropwise followed by a solution of N-Boc 4-propargyloxy-5'-spiro-thalidomide (0.5 M in DCM, 1 equiv.). The suspension is mixed for 12 hours at ambient temperature. The reaction solution is filtered through a frit and the polymer cake is washed is washed with DCM (2 x). The combined filtrate is concentrated and the residue purified by silica gel chromatography to provide tert-butyl 7-(4-((l-(3-hydroxypropyl)- lH-l,2,3-triazol-4-yl)methoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5- carboxylate. (Org. Lett. 2006, 8, 1689) Linker Installation Example 4: r -Butyl 7-(4-(2-((3,5-dihydroxy-3- methylpentyl)oxy)ethoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5- carboxylate

tert- y 7-(4-(2-hydroxyethoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane- 5-carboxylate

A reaction vessel is charged with N-Boc 4-hydroxyl-5'-spiro-thalidomide ( 1 equiv.), potassium carbonate (2 equiv.) and DMF (0.5 M). 2-(2-Chloroethoxy)tetrahydro-2H-pyran (1.1 equiv.) is added and the reaction is heated at 110 °C for 12 hours. The reaction is then cooled to ambient temperature and concentrated. The residue is taken up in water and ethyl acetate and the layers separated. The aqueous layer is extracted with ethyl acetate (2 x). The combined organic layer is washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue is used directly in the following reaction.

A reaction vessel is charged with crude residue ( 1 equiv.), MeOH and DCM (1:1, 0.2 M). ^-Toluenesulfonic acid (0. 1 equiv.) is added and the reaction mixed at ambient temperature. Upon completion of the hydrolysis reaction, the volatiles are removed by rotary evaporation and the residue purified by silica gel chromatography to provide tert-butyl 7-(4-(2-hydroxyethoxy)-l,3- dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate. tert-butyl 7-(l,3-dioxo-4-(2-(2-oxopropoxy)ethoxy)isoindolin-2-yl)-4,6-dioxo-5- azaspiro [2.5] octane-5-carboxylate A reaction vessel is charged with t rt-butyl 7-(4-(2-hydroxyethoxy)-l,3-dioxoisoindolin-

2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate ( 1 equiv.), potassium carbonate (1.2 equiv.) and acetone (0.1 M). Chloroacetone (1.2 equiv.) is then added and the reaction heated at reflux overnight. The reaction is cooled then concentrated and the crude residue partitioned between water and ethyl acetate. The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x). The combined organic layers are dried over sodium sulfate, filtered and concentrated. The crude residue is purified by column chromatography to provide t rt-butyl 7-(l,3-dioxo-4-(2- (2-oxopropoxy)ethoxy)isoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate. (J. Med. Chem. 2007, 50(18), 4304) tert- y 7-(4-(2-((3,5-dihydroxy-3-methylpentyl)oxy)ethoxy)-l,3-dioxoisoindolin-2-yl)-4,6- dioxo-5-azaspiro [2.5] octane-5-car boxylate

1. vinyl MgBr - - -

A reaction vessel is charged with t rt-butyl 7-(l,3-dioxo-4-(2-(2- oxopropoxy)ethoxy)isoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate ( 1 equiv.), and THF (0.2 M), purged with nitrogen and cooled to -78 °C. Vinylmagnesium bromide (4 equiv.) is added dropwise and the reaction is warmed to 0 °C over 1 hour. The reaction is quenched with aq. 1% HCl solution and extracted with ethyl acetate (3 x). The combined organic layer is washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue is purified by silica gel chromatography to provide t rt-butyl 7-(4-(2-((2-hydroxy-2-methylbut-3-en-l- yl)oxy)ethoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate.

Cyclohexene (4.2 equiv.) is added to a solution of BH3 THF ( 1 M in THF, 2 equiv.) at 0 °C under argon. After stirring for 1 hour at 0 °C, a solution of t rt-butyl 7-(4-(2-((2-hydroxy-2- methylbut-3-en-l-yl)oxy)ethoxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5- carboxylate ( 1 equiv.) in THF (0.15 M) is added to the mixture at 0 °C. After stirring for 2 hours at 0 °C, 3 NNaOH (6 equiv.) and 30% H2O2 (33% volume of aq. NaOH solution addition) is added to the mixture. This solution is allowed to mix at ambient temperature for 30 min. The reaction is quenched with saturated aqueous H 4CI (8 volumes) at 0 °C, and the resulting mixture is extracted with ethyl acetate (3 x). The combined extracts are washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure. The crude residue is purified by silica gel chromatography to provide t rt-butyl 7-(4-(2-((3,5-dihydroxy-3-methylpentyl)oxy)ethoxy)-l,3- dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate. (Org. Lett. 2012, 14, 6374)

Linker Installation Example 5: tert -b y 7-(4-((6-chloro-4-hydroxy-4-methylhex-2-yn-l- yl)oxy)-l,3-dioxoisoindolin-2-yl)-4,6-dioxo-5-azaspiro[2.5]octane-5-carboxylate

A reaction vessel is charged with N-Boc 4-propargyloxy-5'-spiro-thalidomide ( 1 equiv.) and the atmosphere cycled between nitrogen and vacuum three times. Anhydrous THF (0.1 M) is added and the reaction cooled to -78 °C. Butyllithium (1.05 equiv.) is added and the reaction is mixed for 15 min. 5-Chloro-2-pentanone (1.1 equiv.) in THF (5 volumes) is then added and the reaction is warmed to ambient temperature and quenched with sat. aq. ammonium chloride solution. Ethyl acetate is added and the phases are separated. The aqueous layer is extracted with ethyl acetate (2 x). The combined organic layers are washed with brine, dried over sodium sulfate, filtered and concentrated. The crude residue is purified by silica gel chromatography to provide tert-butyl 7-(4-((6-chloro-4-hydroxy-4-methylhex-2-yn-l-yl)oxy)-l,3-dioxoisoindolin-2-yl)-4,6- dioxo-5-azaspiro [2 .5]octane-5-carboxy1ate. PREPARATIO ING LIGANDS

(S)-6-(4-Chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H-benzo[f|[l,2,4]triazolo[4,3- a][l,4]diazepine

tert-Butyl (R)-(l-((4-bromo-2-(4-chlorobenzoyl)phenyl)amino)-l-oxopropan-2-yl)carbamate (2-amino-5-bromophenyl)(4-chlorophenyl)methanone (1.0 equiv.) and Boc-(L)-Ala (1.0 equiv.) is suspended in DMF and cooled to 0 °C. DIEA (2.0 equiv.) is added followed by HATU (1.1 equiv.) and the reaction is stirred at reduced temperature for 30 minutes and then warmed to room temperature. When the reaction is judged to be complete it is quenched with aq. ammonium chloride and extracted with ethyl acetate. The combined organic layers are dried over sodium sulfate, concentrated and purified by silica gel chromatography to provide t rt-butyl (R )-(l-((4- bromo-2-(4-chlorobenzoyl)phenyl)amino)-l-oxopropan-2-yl)carbamate.

(S)-7-Bromo-5-(4-chlorophenyl)-3-methyl-l,3-dihydro-2H-benzo[e][l,4]diazepin-2-one

To a stirred solution of boc protected amine in CHCh at r. , is added hydrogen chloride gas slowly. After 20 minutes the addition is stopped and the reaction is stirred at r.t. until deprotection is complete. The reaction mixture is then washed with saturated bicarbonate solution (2x) and water (2x). The organic layer is concentrated under reduced pressure. The residue is dissolved in 2 :1methanol:water and the pH is adjusted to 8.5 by the addition of IN aqueous NaOH. The reaction is then stirred at r.t. until the cyclization is complete. MeOH is then removed under redued pressure and the solution is extracted with DCM (3x). The combined organic layer is dried over sodium sulfate, concentrated and purified by silica gel chromatography to provide (S)-7- bromo-5-(4-chlorophenyl)-3 -methyl- 1,3-dihydro-2H-benzo[e] [1,4]diazepin-2-one (US 2010

026171 1.).

(S)-8-Bromo-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine

A solution of diazapine (1.0 equiv.) in THF is cooled to -10°C and NaH (0.85 equiv.) is added in one portion. After an hour at reduced temperature di-4-morphilinylphosphinic chloride (1.07 equiv.) is added at -10°C and the reaction is allowed to warm to r.t. and stir for 2 hours. To this mixture is added a solution of acetic hydrazide (1.4 equiv.) in n-butanol and stirring is continued for 30 minutes. The solvent is then removed under reduced pressure and the residue dissolved in fresh dry n-butanol before refluxing for the desired time frame. Upon the completion of the reaction the volatiles are removed by rotary evaporation and the residue is partitioned between DCM and brine. The organic layer is dried, concentrated and purified by silica gel chromatography to provide (S)-8-bromo-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (US 2010 026171 1.).

(S)-6-(4-Chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H-benzo[f|[l,2,4]triazolo[4,3- a][l,4]diazepine To a vial containing (S)-8-bromo-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine ( 1 equiv.) is added Pd(PPh3)4 (20 mol %), 4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (1.5 equiv.), and potassium carbonate (2.5 equiv.). The vial is then evacuated and purged under N2. To the vial is added dioxane:water (2:1). The contents were once again evacuated and purged under N2 and the reaction mixture was heated to 80 °C until the SM is converted. The mixture is then cooled to room temperature and filtered over a pad of Celite®. The filter pad is rinsed with EtOAc (3x) and the filtrate is concentrate. The crude material is purified by flash chromatography (WO 2015156601). HO (S)-4-(l,4-Dimethyl-8-(l-methyl-lH-pyrazol-4-yl)-4H-benzo[f][l,2,4]triazolo[4,3- a] [l,4]diazepin-6-yl)phenol

Methyl (R)-5-bromo-2-(2-((teri-butoxycarbonyl)amino)propanamido)benzoate Methyl 2-amino-5-bromobenzoate (1.0 equiv.) and Boc-(L)-Ala (1.0 equiv.) is suspended in DMF and cooled to 0 °C. DIEA (2.0 equiv.) is added followed by HATU (1.1 equiv.) and the reaction is stirred at reduced temperature for 30 minutes and then warmed to room temperature. When the reaction is judged to be complete it is quenched with aq. ammonium chloride and extracted with ethyl acetate. The combined organic layers are dried over sodium sulfate, concentrated and purified by silica gel chromatography to provide methyl (R)-5-bromo-2-(2-((fer t- butoxycarbonyl)amino)propanamido)benzoate.

Methyl 5-bromo-2-(3-((R)-l-((teri-butoxycarbonyl)amino)ethyl)-5-methyl-4H-l,2,4-triazol- 4-yl)benzoate

Methyl (R)-5-bromo-2-(2-((teri-butoxycarbonyl)amino)propanamido)benzoate A solution of methyl (R)-5-bromo-2-(2-((ter t-butoxycarbonyl)amino)propanamido)benzoate (1.0 equiv.) in THF is cooled to -10°C and NaH (0.85 equiv.) is added in one portion. After an hour at reduced temperature di-4-morphilinylphosphinic chloride (1.07 equiv.) is added at -10°C and the reaction is allowed to warm to r.t. and stir for 2 hours. To this mixture is added a solution of acetic hydrazide (1.4 equiv.) in n-butanol and stirring is continued for 30 minutes. The solvent is then removed under reduced pressure and the residue dissolved in fresh dry n-butanol before refluxing for the desired time frame. Upon the completion of the reaction the volatiles are removed by rotary evaporation and the residue is partitioned between DCM and brine. The organic layer is dried, concentrated and purified by silica gel chromatography to provide methyl (R)-5-bromo-2- (2-((tert-butoxycarbonyl)amino)propanamido)benzoate (BMCL 2015, 25, 1842-48).

(S)-8-Bromo-l,4-dimethyl-4,5-dihydro-6H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-one

Methyl (R)-5-bromo-2-(2-((ter t-butoxycarbonyl)amino)propanamido)benzoate is brought up in DCM and cooled to 0°C. 4M HC1 in dioxane is added and the reaction is warmed to r. . When deprotection is complete the reaction is concentrated and then azeotroped from toluene (2x). The crude amine salt is then dissolved in THF and cooled to -40°C at which time iPrMgBr solution is added dropwise (2.0 equiv.) and the reaction is stirred at reduced temp until complete conversion (BMCL 2015, 25, 1842-48).

(S)-l,4-Dimethyl-8-(l-methyl-lH-pyrazol-4-yl)-4,5-dihydro-6H-benzo[f][l,2,4]triazolo[4,3- a] [l,4]diazepin-6-one

To a vial containing (S)-8-bromo-l,4-dimethyl-4,5-dihydro-6H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-one ( 1 equiv.) is added Pd2 (dba) 3 (10 mol %), tri- t rt-butylphosphonium tetrafluorob orate (20 mol %), l-methyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)4H-pyrazole (1.5 equiv.), and potassium phosphate tribasic, monohydrate (2.5 equiv.). The vial is then evacuated and purged under N2. To the vial is added 20: 1 ratio by volume of dioxane:water. The contents were once again evacuated and purged under N2 (g) and the reaction mixture was heated to 100 °C until the SM is converted. The mixture is then cooled to room temperature and filtered over a pad of Celite The filter pad is rinsed with EtOAc (3x) and the filtrate is concentrate. The crude material is purified by flash chromatography.

(S)-6-Chloro-l,4-dimethyl-8-(l-methyl-lH-pyrazol-4-yl)-4H-benzo[f|[l,2,4]triazolo[4,3- a][l ,4]diazepi

(S)- 1,4-Dimethyl-8-( 1-methyl- lH-pyrazol-4-yl)-4, 5-dihydro-6H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-6-one (1.0 equiv.) is dissolved in DCM and PCI5 (1.7 equiv.) is added in one-portion. After conversion of SM 2M sodium carbonate is added. The biphasic mixture is subsequently extracted with EtOAc (4x). The combined organic layers were dried over sodium sulfate and concentrated to dryness. The resultant residue is purified by flash chromatography.

(S)-4-(l,4-Dimethyl-8-(l-methyl-lH-pyrazol-4-yl)-4H-benzo[f][l,2,4]triazolo[4,3- a][l ,4]diaz in-6-yl)phenol

To a vial containing ((S)-6-chloro-l,4-dimethyl-8-(l-methyl-lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine ( 1 equiv.) is added Pd(PPh3 (20 mol %), 4-hydroxy- Phenyl boronic acid (1.5 equiv.), and sodium carbonate (2.5 equiv.). The vial is then evacuated and purged under N2. To the vial is added tol:DME:water ( 1:1:5). The contents were once again evacuated and purged under N2 and the reaction mixture was heated to 80 °C until the SM is converted. The mixture is then cooled to room temperature and filtered over a pad of Celite®. The

350 351 352 Oxetane Sulfone

ref: J. C e . Soc, Perkm Trans. 1, 997, 351 9 yclopropyl 2

IX. REPRESENTATIVE SYNTHESIS OF COMPOUNDS

Methods for HPLC purification:

All prep HPLC was conducted on Waters auto purification instrument equipped with a - Waters XBridge Prep CI8 5 um OBD 19*100 mm column operating at ambient temperature and a flow rate of 25.0 ml/min. Mobile phase: A = 0.1% TFAin water, B=0.1% TFA in Acetonitrile; Unless otherwise noted, Gradient Profile: Mobile phase initial composition of 99% A and 1% B, then to 1% A and 99% B over 6 minutes, holds at that percentage for 0.5 min. and returns to 99: 1 by 8 minutes.

Methods for flash chromatography purification: Use Isco Combiflash systems. Normal phase: silica gel cartridges; ethyl acetate/hexanes, methanol/dichoromethane or specifically stated. Reverse Phase (RP): C18 cartridges; acetonitrile/water w 0.1% TFA or specifically stated. Intermediate 1-3. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione heme 1.

1-1 1-2 1-3

A mixture of 3-fluorophthalic anhydride (1-1) (25 g, 150 mmol), 3-aminopiperidine-2,6- dione, HCl salt (1-2) (24.6 g, 150 mmol), and potassium acetate (36.8 g, 375 mmol) in acetic acid (200 mL) was stirred at 120 °C overnight. The dark mixture was cooled and filtered. The filter cake was dissolved in DCM and washed with saturated NaHC03 and brine. The organic layer was dried (Na2S0 4) and concentrated to provide 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3- dione as a pale yellow solid (1-3) (19.12 g, 46.1 %). LCMS (ES+): m/z 277 [M + H]+

Intermediate 2-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-5-fluoroisoindoline-l,3-dione Scheme 2.

A mixture of 5-fluoroisobenzofuran-l,3-dione (2-1) (25 g, 150 mmol), 3-aminopiperidine- 2,6-dione, HCl salt (1-2) (4.44 g, 27.0 mmol), and potassium acetate (5.76 g, 58.7 mmol) in acetic acid (3 1 mL) was stirred at 120 °C overnight. The dark mixture was cooled and filtered. The filter cake was dissolved in DCM and washed with saturated NaHC03 and brine. The organic layer was dried (Na2S0 4) and concentrated to provide 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-l,3- dione as a grey solid (2-2) (5.78 g, 89.0 %). LCMS (ES+): m/z 277 [M + H]+

Intermediate 3-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-5-ethynylisoindoline-l,3-dione cheme 3.

Suspended 5-bromo-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (3-1) (1.01 g, 2.99 mmol) in 8 mL DIPA and purged with nitrogen for 15 min. Added copper iodide (0.05, 28.4 mg, 0 .1495 mmol) and dichlorobis(tripheny (0.05, 104 mg, 0 .1495 mmol), followed by nitrogen purge for another 15 min. Then added ethynyltrimethylsilane (537 L, 3.88 mmol) reflux for overnight. Diisopropylamine was removed under reduced vacuum, subject to isco 0-15% MeOH/DCM to give 2-(2,6-dioxopiperidin-3-yl)-5-((trimethylsilyl)ethynyl)isoindoline-l,3-dione (680 mg, 1.91 mmol, 64.7 %) as an oil. LCMS (ES+): m/z 355 [M + H]+ Dissolved 2-(2,6-dioxopiperidin-3-yl)-5-((trimethylsilyl)ethynyl)isoindoline-l,3-dione (300 mg, 0.8464 mmol) in 1 mL dry THF at rt and added TBAF solution in THF (1M) slowly, and stir at rt, after 1 hour, quenched with 5 mL citric acid aq. solution (0.5M), wash organic phase with water and brine, dry over with sodium sulfate. Filtered off solid, remove solvent to afford 2-(2,6- dioxopiperidin-3-yl)-5-ethynylisoindoline-l,3-dione (3-2) (215 mg,90.3 %). m/z 283 [M + H]+

Intermediate 4-1. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(prop-2-yn-l- yloxy)isoindoline-l,3-dione Scheme 4.

1-3 4-1

Sodium hydride (73.8 mg, 1.85 mmol) was added to a solution of 2-(2,6-dioxopiperidin-3- yl)-4-fluoroisoindoline-l,3-dione (1-3) (.513 g, 1.85 mmol) in DMF (9 mL ) at 0°C. In another flask sodium hydride (95.8 mg, 2.40 mmol) was added to a solution of prop-2-yn-l-ol (134 mg, 2.40 mmol) in DMF (9 mL ) at 0°C. After stirring for one hour the solution containing prop-2-yn- l-ol (134 mg, 2.40 mmol) was cannulated into the first flask and the reaction was left to stir for 16 hours. Quench with citric acid and extraction with 4:1 chloroform/iPrOH followed by concentration onto celite. The product was purified by silica gel flash chromatography (Column, DCM/MeOH 0-15%) to give 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-yloxy)isoindoline-l,3- dione (4-1) (125 mg, 0.4002 mmol, 21.6 %) as a white solid. LCMS (ES+): m/z 313 [M + H]+

Intermediate 5-1. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(prop-2-yn-l- ylamino)isoindoline-l,3-dione Scheme 5.

1-3 S-1

2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (.425 g, 1.53 mmol) was dissolved in DMAC (3 mL) in a small vial under nitrogen prop-2-yn-l -amine (126 , 1.98 mmol) and Diisopropylethylamine (532 , 3.06 mmol) were addded by syringe and the reaction heated to 90 °C for 16 hours. After cooling the reaction was diluted with water, extracted with EtOAc. The organic layer was washed with brine, dried with sodium sulfate and concentrated directly onto celite. Purification by silica gel chromatography (0-15% DCM/methanol) gave 2-(2,6- dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (313 mg, 1.00 mmol, 65.7 %) as a yellow oil. LCMS (ES+): m/z 312 [M + H]+

Intermediate 6-1. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(methyl(prop-2-yn-l- yl)amino)isoindoline-l,3-dione Scheme 6. 1-3 6-1 Brought 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (400 mg, 1.44 mmol) up in DMAC (1.8 mL) and added DIEA (502 , 2.88 mmol), followed by N-methylprop- 2-yn-l-amine (133 , 1.58 mmol) at r.t. Started heating in a sealed vial at 95°C. After 7 hours seems to have stalled by lcms still some Ar-F ~ 25%. Added ~ 30 uL of propargyl amine and stirred O/N. Partitioned between EA and water. Washed Aq w water 2x followed by 5% LiCl solution dried org over Na2S0 4 and concentrated to ~ 600 mg crude. Purified by isco 2-12% MeOH/DCM to give 2-(2,6-dioxopiperidin-3-yl)-4-(methyl(prop-2-yn-l-yl)amino)isoindoline- 1,3-dione (6-1) (200 mg, 0.6147 mmol, 42.7 %) as a yellow solid. LCMS (ES+): m/z 326 [M + H]+

Intermediate 7-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-((l- ethynylcyclopropyl)amino)isoindoline-l,3-dione Scheme 7.

Brought 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (300 mg, 1.08 mmol) up in DMAC (2.2 mL) and added DIEA(752 , 4.3 mmol), followed by 1- ethynylcyclopropanamine hydrochloride (7-1) (253 mg, 2.2 mmol) at r.t. Started heating in a sealed vial at 95°C. Stirred O/N, Partitioned between EA and water. Washed Aq w water 2x followed by 5% LiCl solution dried org over Na2S0 4 and concentrated. Purified by isco 2-12% MeOH/DCM to give 2-(2,6-dioxopiperidin-3-yl)-4-((l-ethynylcyclopropyl)amino)isoindoline- 1,3-dione (7-2) (180 mg, 0.540 mmol, 50.0 %) as a yellow solid. LCMS (ES+): m/z 338 [M + H]+ Intermediate 8-2. Preparation of 4-(but-3-yn-l-ylamino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione Scheme 8.

1" 8 1 " 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (1.17 mmol) was dissolved in DMAC (3 mL) in a small vial under nitrogen but-3-yn-l -amine hydrochloride (8-1) (1.17 mmol) and Diisopropylethylamine (629 , 3.62 mmol) were addded by syringe and the reaction heated to 90 °C for 16 hours. After cooling the reaction was diluted with water, extracted with EtOAc. The organic layer was washed with brine, dried with sodium sulfate and concentrated directly onto celite. Purification by silica gel chromatography (0-15% DCM/methanol) gave 2- (2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (8-2) (0.313 g, 66%) as a yellow oil. LCMS (ES+): m/z 326 [M + H]+

Intermediate 9-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(pent-4-yn-l- ylamino)isoindoline-l,3-dione Scheme 9.

Brought 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (500 mg, 1.81 mmol) up in DMAC (1.8 mL) and added DIEA(631 , 3.62 mmol), followed by pent-4-yn-l- amine (9-1) (174 , 1.81 mmol) at r.t. Started heating in a sealed vial at 95°C. After 1 night partitioned between EA and water. Washed Aq w water 2x followed by 5% LiCl solution dried org over Na2S04 and concnetrated. Purified by isco 2-12% MeOH/DCM to give 2-(2,6- dioxopiperidin-3-yl)-4-(pent-4-yn-l-ylamino)isoindoline-l,3-dione (9-2) (200 mg, 0.5893 mmol, 32.5 %) as a yellow solid. LCMS (ES+): m/z 340 [M + H]+

Intermediate 10-2. Preparation of 3-(4-Bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione Scheme 10.

A mixture of methyl 3-bromo-2-(bromomethyl)benzoate (10-1) (6.0 g, 19.4 mmol) 3- aminopiperidine-2,6-dione.HCl (1-2) (3.8 g, 23.2 mmol) and DIEA (12.5 g, 97.0 mmol) in CH3CN (60 mL) was heated to 90°Covernight. The reaction was filtered, washed with Ether and dried on high vac to afford 3-(4-bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione (10-2) (5.0 g, 15.4 mmol, 79.8%) as a purple soild. LCM4S (ES+): m/z 323 [M + H]+

1H M R (500 MHz, DMSO-de): δ 11.02 (s, 1H), 7.88 (d, J=8 Hz ,1H), 7.78 (d, J=7.5Hz, 1H), 7.53(t, J=7.5Hz, 1H), 5.17-5.13 (m, 1H), 4.44 (d, J=18Hz, 1H), 4.28 (d, J=18, 1H), 2.95-2.88 (m, 1H), 2.62 (d, J=1.5, 1H), 2.51-2.46 (m, 1H), 2.04-2.01 (m, 1H).

Intermediate 11-1. Preparation of reparation of 4-(2-(2,6-Dioxopiperidin-3-yl)-l- oxoisoindolin-4-yl)-4-hydroxy-lX4-piperidin-2-ylium 2,2,2-trifluoroacetate heme 11.

11-1

To solution of 3-(4-bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione (1.0 g, 3.09 mmol), t r t-butyl 4-oxopiperidine-l-carboxylate (10-2) (1.53 g, 7.72 mmol) in dry Tetrahydrofuran (20 mL ) , Sodium hydride (123 mg, 3.09 mmol) was added at 0 °C and the reaction mixture was stirred at same temperature for Ih.Then the reaction mixture was evacuated and back filled with nitrogen to remove hydrogen gas.Then n-Butyl (6.16 mL, 15.4 mmol) was added at -78 °C and stirred for another 16h at rt.The reaction progress was monitored by TLC and LCMS. TLC and LCMS showed product formation along with starting material and debrominated product.The reaction mixture was quenched saturated aqueous ammonium chloride solution(20 mL) and extracted with ethylacetate(100.0mLx3). The organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford crude product.The crude product was purified by flash column chromatography on combi-flash instrument (using 12.Og Redisef) and eluted with 4% to 5% methanol in DCM to get t rt-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-4- hydroxypiperidine-l-carboxylate (190 mg, 428 µπιοΐ , 13.8 %) as brown solid. LC/MS (ES+): m/z 444.27 [M + H]+ To solution of tert-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-4- hydroxypiperidine-l-carboxylate (200 mg, 450 µιηοΐ) in DCM (2 mL ) , 25% TFA in DCM (2.0 mL ) was added at 0 °C and the reaction mixture was stirred at rt for 2h.After completion of starting material the volatile of the reaction mixture was evaporated at 40 °C.The crude residue was triturated with diethylether to get 3-(4-(4-hydroxypiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine- 2,6-dione with TFA salt (11-1) (180 mg, 393 µιηοΐ , 87.8 %) as light brown solid. LC/MS (ES+): m/z 344.21 [M + H]+

Intermediate 12-3. Preparation of 4-(2-(2,6-Dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)- l,2,5,6-tetrahydro-lX4-pyridin-2-ylium 2,2,2-trifluoroacetate Scheme 12.

-2 -2 -3

To a solution of 3-(4-bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione (10-2) ( 1 g, 3.09 mmol) , t r t-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-l(2H)- carboxylate (12-1) (1.14 g, 3.70 mmol) , cesium carbonate (3.01 g, 9.27 mmol) and Dioxane (8 mL ) and WATER (2 mL ) in (4: 1) , purged nitrogen for 10 min.Then added Pd(OAc)2 (69.3 mg,

309 µ οΐ) n-butyl diadamantyl phosphine (221 mg, 618 µιηοΐ) and sealed the cap. stirred the reaction at 100°C for 1 h.The reaction progress was monitored by TLC.After completion of reaction quenched the reaction with water (20 mL) and extracted with ethyl acetate (10 mLx3) .Combined the organic layers and dried over anhydrous sodium sulfate,filtered and distilled ,washing with pentane (10 mLx3) was given to obtain t rt-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l- oxoisoindolin-4-yl)-5,6-dihydropyridine-l(2H)-carboxylate (12-2) (500 mg, 1.17 mmol, 38.1 %) as off white solid. LC/MS (ES+): m/z 426.16 [M + H]+ To a solution of ter t-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-5,6- dihydropyridine-l(2H)-carboxylate (12-2) (300 mg, 705 µιηοΐ) in DCM (6 mL ) at 0°C under nitrogen atmosphere added 25% TFA in DCM (6 mL, 705 µιηοΐ) drop wise,stirred the reaction at RT for 3h.Reaction progress was monitored by TLC. After completion of reaction distilled the DCM , washing from ether (lOmL) was given to obtain 4-(2-(2,6-dioxopiperidin-3-yl)-l- oxoisoindolin-4-yl)-l,2,3,6-tetrahydropyridin-l-ium 2,2,2-trifluoroacetate (12-3) (200 mg, 612

µιηοΐ , 86.9 %) as light brown solid. LC/MS (ES+): m/z 326 [M + H]+

Intermediate 13-1. Preparation of 3-(l-Oxo-4-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6- dione 2,2,2-trifluoroacetate Scheme 13.

12-2 13-1 To a solution of ter t-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-5,6- dihydropyridine-l(2H)-carboxylate (12-2) (50 mg, 117 µιηοΐ) in METHANOL (2.5 mL ), DMF (0.5 mL ) added 10%Pd/C (0 µg, 0 µιηοΐ) and hydrogenated the reaction using balloon pressure for 1 h.The reaction was filtered through a celite bed and concentratated, washing with diethyl ether (5 mLx3) was given to obtain t r t-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)piperidine-l -carboxylate (15.0 mg, 35.0 µπιοΐ , 30.0 %) as white solid. LC/MS (ES+): m/z 428.12 [M + H]+ t rt-Butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)piperidine-l-carboxylate (15.0 mg, 35.0 µηιοΐ) was dissolved in DCM (l.O mL) and TFA (0.25 mL) was added. The reaction was stirred for two hours and concentrated to give crude 3-(l-oxo-4-(piperidin-4-yl)isoindolin-2- yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (13-1) as a brownish solid which was used as is. LC/MS (ES+): m/z 328 [M + H]+

Intermediate 14-3. Preparation of 3-(l-Oxo-4-(piperazin-l-ylmethyl)isoindolin-2- yl)piperidine-2,6-dione Scheme 14.

To a solution of 3-(4-bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione (10-2) (0.500 g, 1.54 mmol) , potassium ((4-(t rt-butoxycarbonyl)piperazin-l-yl)methyl)trifluoroborate (14-1) (563 mg, 1.84 mmol) and Cesium carbonate (1.50 g, 4.62 mmol) were taken up in Dioxane (8 mL ) and water (2 mL ) The reaction mixture was bubbled with argon through solvents for 10 minutes n-butyl diadamantyl phosphine ( 110 mg, 308 µιηοΐ) and palladium acetate (34.5 mg, 154 µιηοΐ) were added and the vial was purged with Ar and sealed. The reaction was stirred at 100 °C for 16h,The reaction progress was monitored by TLC and LCMS.er TLC showed consumption of SM, the reaction mixture was cooled to room temperature and quenched by adding water (25 mL). The mixture was extracted with Ethyl Acetate (50 mLx3 ). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated . The crude product was purified by flash column chromatography using 12.0 g redisef and eluted with Methanol in DCM(3%-5%) to obtain tert- butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)methyl)piperazine-l-carboxylate (14- 2) (400 mg, 903 µιηοΐ , 58.7 %) as a grey solid. LC/MS (ES+): m/z 443.05 [M + H]+ To a RB flask tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)methyl)piperazine-l-carboxylate (14-2) (0.350 g, 0.7909 mmol) in DCM (6 mL ) was added under nitrogen atmosphere at RT. To the reaction mixture 25% TFA in DCM (5 mL ) was added dropwise at 0°C. and stirred the reaction mixture for 2 hours at RT. After completion of the starting material the reaction mixture was concentrated by using rota vapour. The residue was triturated by diethyl ether (15mL) to get 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)methyl)piperazin-l-ium 2,2,2-trifluoroacetate (0.30 g, 0.8771 mmol, 100. 0%) as an off white solid. DCM (5 mL), potassium carbonate (60.4 mg, 438 µιηοΐ) was added to the flask, and the reaction mixture was stirred for 1hour at RT. Filtered the reaction mixture by using sintered funnel to get 3-(l-oxo-4-(piperazin-l-ylmethyl)isoindolin-2-yl)piperidine-2,6-dione trifluoroacetate (14-

3) (15.0 mg, 43.8 µιηοΐ , 50.0 %) as the desired product. LC/MS (ES+): m/z 343.24 [M + H]+

Intermediate 15-2. Preparation of of 4-(2-(2,6-Dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-5- oxo-lX4-piperazin-2-ylium 2,2,2-trifluoroacetate

15-1

To a solution of 3-(4-bromo-l-oxoisoindolin-2-yl)piperidine-2,6-dione (10-2) ( 1 g, 3.09 mmol) , t rt-butyl 3-oxopiperazine-l-carboxylate (15-1) (1.23 g, 6.18 mmol) and Cesium carbonate (3.01 g, 9.27 mmol) were added in Dioxane (15 mL ) . Purged the reaction mixture for

15 minutes. DMEDA (272 mg, 3.09 mmol) and Cul (292 mg, 1.54 mmol) were added and purged the reaction mixture for 5 minutes through argon. Stirred the RM at 100°C for about 16 hours. Progress of the reaction was monitored by TLC and LCMS.TLC showed consumption of SM, the reaction mixture was cooled to room temperature and quenched by adding water (50*4 mL). The mixture was extracted with Ethyl Acetate (50*6 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and dried . The crude product was purified by flash column chromatography using 12.0 g redisef and eluted with Methanol in DCM(2%-5%) to obtain tert- butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-3-oxopiperazine-l-carboxylate (170 mg, 384 µιηοΐ , 12.5 %) as an off-white solid. LC/MS (ES+): m/z 443[M + H]+ To a RB flask t rt-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-3- oxopiperazine-l-carboxylate (0.050 g, 113 µηιοΐ) in DCM (1.5 mL ) was added under nitrogen atmosphere at RT. 25% TFA IN DCM ( 1 mL ) was added dropwise at 0°C. Stirred the reaction mixture for 2 hours at RT. After completion of the starting material the reaction mixture was concentrated. The residue was triturated by diethyl ether (4mL) to get 3-(l-oxo-4-(2-oxopiperazin- l-yl)isoindolin-2-yl)piperidine-2,6-dione with TFA salt (15-2) (12.0 mg, 35.0 µιηοΐ , 31.0 %) as an off white solid. LC/MS (ES-): m/z 341 [M-H]-

Intermediate 16-2. Preparation of 4-(Azetidin-3-yl(methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate Scheme 16.

2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (0.5 g, 1.81 mmol) was dissolved in DMSO (5 mL) in a small vial under nitrogen t rt-butyl 3-aminoazetidine-l-carboxylate (16-1) (542 mg, 2.71 mmol) and Diisopropylethylamine (944 µΐ 5.43 mmol) were added by syringe and the reaction heated to 90 °C for 16 hours. Shot directly on reversed phase isco 50g column 5-100% ACN/water w tfa to give tert-butyl 3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)(methyl)amino)azetidine-l-carboxylate (300 mg, 0.678 mmol, 37.5 %) as a yellow solid. LC/MS (ES+): m/z 465 [M+Na]+ Brought t r t-butyl 3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)(methyl)amino)azetidine-l-carboxylate (300 mg, 0.6781 mmol) up in dcm (5.0 mL ) and added 2,2,2- (1.0 ml). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 4-(azetidin-3-yl(methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate (16-2) (365 mg, 97.0 %) as crude oil. LC/MS (ES+): m/z 344 [M + H]+ Intermediate 17-2. Preparation of 4-(4-Aminopiperidin-l-yl)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate Scheme 17.

2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (0.5 g, 1.81 mmol) was dissolved in DMSO (5 mL) in a small vial under nitrogen t rt-butyl piperidin-4-ylcarbamate (17- 1) (542 mg, 2.71 mmol) and Diisopropylethylamine (944 , 5.43 mmol) were added by syringe and the reaction heated to 90 °C for 16 hours. Shot directly on reversed phase isco 50g column 5- 100% ACN/water w tfa to give t rt-butyl (4-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)cyclohexyl)carbamate (300 mg, 0.66 mmol, 36.5 %) as a yellow solid. LC/MS (ES+): m/z 493 [M+Na]+ Brought t r t-butyl (4-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)cyclohexyl)carbamate (300 mg, 0.6781 mmol) up in dcm (5.0 mL ) and added 2,2,2- trifluoroacetic acid (1.0 ml). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 4 -(4 -aminopiperidin-1 -yl)-2-(2,6-dioxopipericlin-3-yl)isoincloline-1 ,3-clione 2,2,2-trifluoroacetate (17-2) (365 mg, 96.0 %) as crude oil. LC/MS (ES+): m/z 358 [M + H]+

Intermediate 18-2. Preparation of 4-(Azetidin-3-ylamino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate Scheme 18. 2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (.5 g, 1.81 mmol) was dissolved in MP (3 mL) in a small vial under nitrogen tert-butyl 3-aminoazetidine-l-carboxylate (18-1) (358 mg, 2.08 mmol) and Diisopropylethylamine (787 , 4.52 mmol) were added by syringe and the reaction heated to 90 °C for 16 hours. Shot directly on reversed phase isco 50g column 5-100% ACN/water w tfa to give t r t-butyl 3-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxoisoindolin-4-yl)amino)azetidine-l-carboxylate (450 mg, 1.05 mmol, 58.0 %) as a yellow solid. LC/MS (ES+): m/z 451[M+Na]+ Brought t r t-butyl 3-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)azetidine-l-carboxylate (150 mg, 0.3501 mmol) up in dcm (3.50 mL ) and added 2,2,2- trifluoroacetic acid (0.75 mL, 9.47 mmol). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 4-(azetidin-3-ylamino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione 2,2,2-trifluoroacetate (18-2) (130 mg, 0.2938 mmol, 84.4 %) as crude oil. LC/MS (ES+): m/z 330 [M + H]+

Intermediate 19-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(piperidin-4- ylamino)isoindoline-l,3-dione 2,2,2-trifluoroacetate Scheme 19.

-2

2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (0.5 g, 1.81 mmol) was dissolved in NMP (3 mL) in a small vial under nitrogen t r t-butyl 4-aminopiperidine-l- carboxylate (19-1) (0.4 g, 1.99 mmol) and Diisopropylethylamine (787 µΐ 4.52 mmol) were addded by syringe and the reaction heated to 90 °C for 16 hours. Shot the reaction directly on the reversed phase isco 50g column 5-100% ACN/water w tfa and lyophilized fractions to give tert- butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)amino)piperidine-l-carboxylate (0.43 g, 0.94 mmol, 52.0%) as a yellow solid. LC/MS (ES+): m/z 379 [M+Na]+ Brought t r t-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)amino)piperidine-l-carboxylate (300 mg, 0.657 mmol) up in dcm (5.0 mL ) and added 2,2,2- trifluoroacetic acid (1.5 mL). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 4-(azetidin-3-ylamino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione 2,2,2- trifluoroacetate (19-2) (267 mg, 0.568 mmol, 86.4 %) as crude oil. LC/MS (ES+): m/z 357 [M + H]+

Intermediate 20-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(methyl(piperidin-4- yl)amino)isoindoline-l,3-dione 2,2,2-trifluoroacetate heme 20.

2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (0.5 g, 1.81 mmol) was dissolved in NMP (3 mL) in a small vial under nitrogen t rt-butyl 4-(methylamino)piperidine-l- carboxylate (20-1) (1.99 mmol) and Diisopropylethylamine (787 , 4.52 mmol) were addded by syringe and the reaction heated to 90 °C for 16 hours. Shot reaction mixture directly on reversed phase isco 50g column 5-100% ACN/water w tfa to give t r t-butyl 4-((2-(2,6-dioxopiperidin-3- yl)-l,3-dioxoisoindolin-4-yl)(methyl)amino)piperidine-l-carboxylate (699 mg, 1.48 mmol, 82.1 %) as a yellow solid after lyophilization. LC/MS (ES+): m/z 493 [M+Na]+ Brought t r t-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)(methyl)amino)piperidine-l-carboxylate (300 mg, 0.638 mmol) up in dcm (5.0 mL ) and added 2,2,2-trifluoroacetic acid (1.5 mL). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 2-(2,6-dioxopiperidin-3-yl)-4-(methyl(piperidin-4- yl)amino)isoindoline-l,3-dione 2,2,2-trifluoroacetate (20-2) (300 mg, 0.619 mmol, 97.4 %) as crude oil. LC/MS (ES+): m/z 371 [M + H]+

Intermediate 21-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(4-(methylamino)piperidin- l-yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate Scheme 21. 21-2

t r t-Butyl methyl(piperidin-4-yl)carbamate (21-1) (426 mg, 1.99 mmol) was dissolved in DMAC (3 mL) in a small vial under nitrogen. 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3- dione (1-3) (1.81 mmol) and Diisopropylethylamine (787 , 4.52 mmol) were addded by syringe and the reaction heated to 90 °C for 16 hours. Shot reaction mixture directly on reversed phase isco 50g column 5-100% ACN/water w tfa and lyophilizaed fractions to give t rt-butyl (l-(2-(2,6- dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)piperidin-4-yl)(methyl)carbamate (0.80 g, 1.70 mmol, 94.0%) as a yellow solid. LC/MS (ES+): m/z 493 [M+Na]+ Brought t rt-butyl (l-(2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)piperidin-4- yl)(methyl)carbamate (500 mg, 1.06 mmol) up in dcm (5.3 mL ) and added 2,2,2-trifluoroacetic acid (1.5 mL). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 2-(2,6-dioxopiperidin-3-yl)-4-(4-(methylamino)piperidin-l-yl)isoindoline-l,3-dione 2,2,2- trifluoroacetate (21-2) (500 mg, 1.03 mmol, 97.4 %) as crude oil. LC/MS (ES+): m/z 371 [M + H]+

Intermediate 22-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-((2- (methylamino)ethyl)amino)isoindoline-l,3-dione 2,2,2-triflouroacetate heme 22.

Brought 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (500 mg, 1.81 mmol) , rt-butyl methyl(2-(methylamino)ethyl)carbamate (22-1) (1.99 mmol) and DIEA (787 4.52 mmol) up in NMP (3.62 mL ) and heated to 90°C overnight. Cooled the mixture to r.t. and loaded directly onto reversed phase isco 10- 100% ACN/water w TFA. Lyophilized the combined fractions to give t r t-butyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)(methyl)amino)ethyl)(methyl)carbamate (800 mg, 1.79 mmol, 99.5 %) as a yellow solid. LC/MS (ES+): m/z 467 [M+Na]+ Brought t r t-butyl (2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)(methyl)amino)ethyl)(methyl)carbamate (300 mg, 0.675 mmol) up in dcm (5.0 mL ) and added 2,2,2-trifluoroacetic acid (1.5 mL). Stirred for 30 minutes and added 1 mL toluene before concentrating to dryness giving 2-(2,6-dioxopiperidin-3-yl)-4-((2- (methylamino)ethyl)amino)isoindoline-l,3-dione 2,2,2-trifluoroacetate (22-2) (339 mg, 100 %) as crude oil. LC/MS (ES+): m/z 346 [M + H]+

Intermediate 23-3. Preparation of 3-(5-Amino-l-oxoisoindolin-2-yl)piperidine-2,6-dione Scheme 23.

Preparation of methyl 2-(bromomethyl)-4-nitrobenzoate (23-1) (2.5 g, 9.12 mmol) and 3- aminopiperidine-2,6-dione (1-2) (1.74 g, 13.6 mmol) were added to DMF (12.5 ml) at 25°C. Potassium carbonate (3.15 g, 22.8 mmol) was added to the reaction mixture at 25°C and the temperature was raised to 42°C. The reaction mixture was stirred for 6 hours at 42°C and cooled to 20°C to 25°C. De-ionized water (12.5 ml) was added to the reaction mixture at 20°C and stirred for 20 minutes. The solid obtained was filtered, washed with de-ionized water (2 x 25 ml) and dried under vacuum at 40°C to 45°C for 20 hours to obtain the title compound. Then the crude material was slurried with PE/EA(1/1) 20 mL to give 3-(5-nitro-l-oxoisoindolin-2-yl)piperidine- 2,6-dione (23-2) (1.50 g, 5.18 mmol, ) as gray-green solid. LC/MS (ES+): m/z 290 [M + H]+ A mixture of 3-(5-nitro-l-oxoisoindolin-2-yl)piperidine-2,6-dione (23-2) (1.3 g, 4.49 mmol) and Pd/C (10 mg) in Methyl Alcohol (60 mL ) was stirred under H2 at room temperature for 48 hour.And part of reactant dissolved in methanol. The mixture was filtered through a Celite pad,the filtrate was concentrated and solid was precipitate out.Filter to get the 3-(5-amino-l- oxoisoindolin-2-yl)piperidine-2,6-dione (23-3) (683 mg, 2.63 mmol, 58.8 %) LC/MS (ES+): m/z 260 [M + H]+

Intermediate 24-3. Preparation of 3-(6-Amino-l-oxoisoindolin-2-yl)piperidine-2,6-dione Scheme 24.

To a solution of methyl 2-(bromomethyl)-5-nitrobenzoate (24-1) (5 g, 18.2 mmol) in ACN (50 mL ) , 3-aminopiperidine-2,6-dione hydrochloride (1-2) (2.99 g, 18.2 mmol) was added to the reaction mixture. The reaction mixture was stirred for 15 minutes at RT. Then DIEA ( 11.7 g, 91.0 mmol) was added the reaction mixture was heated and stirred for 18 hours at 90°C.RM was monitored by TLC. Dark violet solid was filtered through sintered and washed it with water (300ml) and DCM (100ml) and dried under high vaccum to get 3-(6-nitro-l-oxoisoindolin-2- yl)piperidine-2,6-dione (24-2) (4.00 g, 13.8 mmol, 76.0 %) as dark gray solid. LC/MS (ES+): m/z 290 [M + H]+ To a solution of 3-(6-nitro-l-oxoisoindolin-2-yl)piperidine-2,6-dione (24-2) (2.0 g, 6.91 mmol) in 10% DMF in THF ( 30ml ),10% Pd/C (50% Moisture) (2g) was added . The reaction mixture was stirred at rt for 1 hour under Hydrogen atmosphere. The reaction mixture was monitored by TLC and LCMS. After completion the reaction mixture was filtered through celite bed and washed with methanol(300.0 mL). The filtrarte was concentrated to get 3-(6-amino-l- oxoisoindolin-2-yl)piperidine-2,6-dione (24-3) (1.50 g, 5.78 mmol, 83.7 %) as dark gray solid. LC/MS (ES+): m/z 260 [M + H]+

Intermediate 25-4. Preparation of 3-(5-Amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione cheme 25.

To a stirred mixture of 2-amino-6-nitrobenzoic acid (25-1) ( 1 g, 5.49 mmol) and imidazole (1.2eq.) in Acetonitrile (10 mL ) was added acetyl chloride (25-2) (469 , 6.58 mmol) and the reaction was stirred overnight. 3-aminopiperidine-2,6-dione hydrochloride (903 mg, 5.49 mmol) was then added followed by rest of the base to make it total lH-Imidazole (1.34 g, 19.7 mmol). Then phosphorous acid, triphenyl ester (1.72 mL, 6.58 mmol) was added and the reaction was refluxed for two days. After cooling to r.t.added 40 mL water, slowly form suspension, filter off, wash with water and EtOAc. Isolated solid 3-(2-methyl-5-nitro-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (1-2) (953 mg, 3.01 mmol, 55.0 %) as a solid. LC/MS (ES+): m/z 317 [M + H]+ Brought 3-(2-methyl-5-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (25-3) (690 mg, 2.18 mmol) up in 10 mL dry DMF, and added dihydroxypalladium (306 mg, 0.218 mmol) on carbon 50% wet, purged with nitrogen for 15 min, purged with hydrogen three cycles and subjected to hydrogen 1 atm for overnight; Filtered off Pd via celite pad, concentrated and subject to purification by isco MeOH/DCM to give 3-(5-amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (25-4) (498 mg, 1.73 mmol, 79.8 %) as an off-white solid. LC/MS (ES+): m/z 287 [M + H]+

Intermediate 26-3. Preparation of 3-(8-Amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione Scheme 26. To a stirred mixture of 2-amino-3-nitrobenzoic acid (26-1) (2 g, 10.9 mmol) and imidazole (1.2eq.) in Acetonitrile (20 mL) was added acetyl chloride (25-2) (927 µΐ 13.0 mmol) and stirred overnight. Added 3-aminopiperidine-2,6-dione hydrochloride (1-2) (1.79 g, 10.9 mmol) followed by the remaining base to make it a total lH-Imidazole (2.66 g, 39.2 mmol), then added Phosphorous acid, triphenyl ester (3.40 mL, 13.0 mmol) and heated to reflux for 1 night. Cooled reaction and added 60 mL water, light yellow solid precipitated, wash with water and EtOAc. Crude 3-(2-methyl-8-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (26-2) (2.07 g, 6.54 mmol, 60.1 %) was obtained after concentration of organic layer. LC/MS (ES+): m/z 317 [M + H]+ Brought crude 3-(2-methyl-8-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (26-2) (2 g, 6.32 mmol) up in N,N-dimethylformamide (35 mL, 6.32 mmol) under nitrogen flow, purged and added dihydroxypalladium (879 mg, 1.26 mmol) on carbon. Purged with nitrogen, then purged with hydrogen, stirred overnight uder a balloon of hydrogen. Filtered through Celite, concentrated and subjected to isco MeOH/DCM 2-30%. Isolated as off-white solid 3-(8-amino- 2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (26-3) (796 mg, 2.78 mmol, 44.2 %). LC/MS (ES+): m/z 287 [M + H]+

Intermediate 27-3. Preparation of 3-(6-Amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione Scheme 27. To a stirred mixture of 2-amino-5-nitrobenzoic acid (27-1) (1.5 g, 8.23 mmol) and imidazole (0.66 g, 9.87 mmol) in acetonitrile (60 mL), was added Acetyl chloride (25-2) (774 mg, 9.87 mmol) at room temperature. The mixture was stirred at room temperature overnight. To the mixture, was added 3-amino-piperidine-2,6-dione hydrogen chloride (1-2) (1.35 g, 8.23 mmol) , imidazole (1.23 g, 18.03 mmol) and Phosphorous acid, triphenyl ester (3.06 g, 9.87 mmol) , and heated to reflux overnight. Water (40 mL) and EA(30 mL) were added to the mixture. The mixture was stirred for lOmin and filtrated to give 3-(2-methyl-6-nitro-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (27-2) (1.22 g, 3.87 mmol, 46.9 %) as an off-white solid. LC/MS (ES+): / 317 [M + H]+ To a solution of 3-(2-methyl-6-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (27- 2) (1.49 g, 4.71 mmol) in DMF(40 mL) was added Palladium hydroxide (330 mg, 2.35 mmol) and Cyclohexene (4.69 mL, 47.0 mmol) . The mixture was stirred at 125 °C overnight. The mixture was filtrated and 2/3 DMF was concentrated: then poured into water(40 mL). The mixture was stirred for 20min and filtrated. The solid was collected and THF(20mL) was added, concentrated to give 3-(6-amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (27-3) (967 mg, 3.37 mmol, 72.1 %). LC/MS (ES+): m/z 287 [M + H]+

Intermediate 28-3. Preparation 3-(7-Amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine- 2,6-dione Scheme 28. To a stirred mixture of 2-amino-4-nitrobenzoic acid (28-1) (2.0 g, 10.9 mmol) and imidazole (0.88 g, 12.84 mmol) in acetonitrile (40 mL), was added Acetyl chloride (25-2) (1.02 g, 13.0 mmol) at room temperature. The mixture was stirred at room temperature overnight. To the mixture, was added 3-amino-piperidine-2,6-dione hydrogen chloride (1-2) (1.78 g, 10.9 mmol) , imidazole (1.60 g, 23.24 mmol) and Phosphorous acid, triphenyl ester (4.03 g, 13.0 mmol) , and heated to reflux overnight. Water (200 mL) was added to the mixture. The suspension was filtered and the solid was stirred for 20min in CH3CN(25 mL). The mixture was filtrated to give 3-(2- methyl-7-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (28-2) (1.70 g, 5.37 mmol, 49.4 %) as an off-white solid. LC/MS (ES+): m/z 317 [M + H]+ To a solution of 3-(2-methyl-7-nitro-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (28- 2) (1.4 g, 4.42 mmol) in DMF(17 mL) was added Palladium hydroxide (310 mg, 2.21 mmol) and Cyclohexene (4.4 mL, 44.0 mmol) . The mixture was stirred at 125 °C overnight. The mixture was poured into water and stirred for 15min. The mixture was filtrated and the solid was collected to give 3-(7-amino-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (28-3) (946 mg, 3.30 mmol, 75.0 %) as white solid. LC/MS (ES+): m/z 287 [M + H]+

Intermediate 29-2. Preparation of 3-(l-Oxo-4-(piperidin-4-ylamino)isoindolin-2- yl)piperidine-2,6-dione as its TFA salt. Brought tert-butyl 4-oxopiperidine-l-carboxylate (76.8 mg, 0.3856 mmol) and 3-(4- amino-l-oxoisoindolin-2-yl)piperidine-2,6-dione (29-1) (50 mg, 0.1928 mmol) up in DCE (1.92 mL )/ acetic acid (200 µΐ 3.31 mmol) and added 4A M S (small scoop, activated) and stirred at r.t. for 4 hours. Added sodium triacetoxyborohydride (40.8 mg, 0.1928 mmol) and stirred O/N at r.t. In AM quenched with sat sodium bicarb solution and extracted into dcm x2. Dried combined organic layers over sodium sulfate and concentrated. Purified by isco 12g column 0-10% MeOH/DCM to give tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)amino)piperidine-l-carboxylate (80.0 mg, 0.1807 mmol, 93.7 %) as an oil. LC/MS (ES-): m/z 441 [M-H]- Brought tert-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)amino)piperidine-l-carboxylate (80 mg, 0.1807 mmol) up in DCM ( 1 mL ) and added 2,2,2- trifluoroacetic acid (0.3 mL, 3.91 mmol) . After stirring at r.t. for 3 hours, concentrated from toluene 3x to give 3-(l-oxo-4-(piperidin-4-ylamino)isoindolin-2-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (29-2) (80.0 mg, 0 .1752 mmol, 97.0 %) as a yellow oil. LCMS (ES+): m/z 343 [M + H]+ ¾NMR (400 MHz, DMSO-de) δ 10.99 (s, 1H), 7.28 (t, J = 7.7 Hz, 1H), 6.95 (d, J = 7.4 Hz, 1H), 6.86 (d, J = 8.1 Hz, 1H), 5.19 - 5.05 (m, 1H), 4.30 - 4.06 (m, 2H), 3.80 - 3.60 (m, 2H), 3.33 (d, J = 12.5 Hz, 2H), 3.10 - 2.84 (m, 4H), 2.66 - 2.58 (m, 1H), 2.36 - 2.20 (m, 1H), 2.12 - 1.98 (m, 2H), 1.90 - 1.75 (m, 1H), 1.68 - 1.44 (m, 2H).

Intermediate 30-2. Preparation of 3-(4-Chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione Scheme 30.

Brought methyl 3-(bromomethyl)-2-chloroisonicotinate (30-1) (7.1 1g, 26.9 mmol) and amino-glutartimide (1-2) (4.42g, 26.9 mmol) up in DMF (50.0 mL) and added TEA (8.24 mL, 59. 1 mmol). Stirred at r.t. for 3 days. Diluted and precipitated from MBTE. Filtered and washed with MBTE. Isolated 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (30-2) (1.59 g, 21.1 %) as an off-white solid. LCMS (ES+): m/z 297 [M + H ]+

Intermediate 31-2. Preparation of r -Butyl (E)-4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo [3,4-c]pyridin-4-yl)vinyl)piperidine-l-carboxylate Scheme 31.

Brought 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (30- 2) (150 mg, 0.5363 mmol) , (E)- rt-butyl 4-(2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)vinyl)piperidine-l-carboxylate (31-1) (360 mg, 1.07 mmol) , cesium carbonate (521 mg, 1.60 mmol) , dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (38.3 mg, 0.08044 mmol) and XPhos G3 (68.0 mg, 0.08044 mmol) up in THF (2.68 mL ) / water (268 ) under Ar, sealed vial and began heating at 60°C. After 6 hours, cooled to r.t. and filtered off white ppt. Rinsed ppt w EtOAc. Isolated (E)- -butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH- pyrrolo[3,4-c]pyridin-4-yl)vinyl)piperidine-l-carboxylate (31-2) (150 mg, 0.3300 mmol, 61.7 %) as a white solid. LCMS (ES+): m/z= 455 [M + H ]+

Intermediate 32-1. Preparation of r -Butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)ethyl)piperidine-l-carboxylate Scheme 32. Brought (E)-tert-butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH- pyrrolo[3,4-c]pyridin-4-yl)vinyl)piperidine-l-carboxylate (31-2) (150 mg, 0.3300 mmol) up in MeOH (1.65 mL ) / DMF (1.65 mL ) and added wet 5% Pd/C (30 mg). Put reaction under a Hydrogen balloon and stirred O/N. Filtered over celite and concentrated, then loaded onto isco for purification 0-15% MeOH/DCM. Isolated rt-butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)ethyl)piperidine-l-carboxylate (32-1) (90.0 mg, 0.1971 mmol, 60.0 %) as an oil. LCMS (ES-): m/z= 455 [M - H]-

Intermediate 33-1. Preparation of 3-(l-Oxo-4-(2-(piperidin-4-yl)ethyl)-lH-pyrrolo[3,4- c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate Scheme 33.

Brought t rt-butyl 4-(2-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)ethyl)piperidine-l-carboxylate (32-1) (90.0 mg, 0.197 mmol) up in DCM ( 1 mL) and added trifluoroacetic acid (0.20 mL, 2.52 mmol) . Stirred at r.t. for 45 minutes. Diluted with toluene and concentrated. Azeotroped from toluene 2x to give 3-(l-oxo-4-(2-(piperidin-4- yl)ethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (33-1) (92.0 mg, 0.1955 mmol, 99.2 %). LCMS (ES+): m/z= 357 [M + H]+ Intermediate 34-2. Preparation of r -Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)methylene)piperidine-l-carboxylate Scheme 34.

Brought 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (30- 2) (120 mg, 0.4290 mmol) , r t-butyl 4-((4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)methylene)piperidine-l-carboxylate (34-1) (277 mg, 0.858 mmol) , cesium carbonate (417 mg, 1.28 mmol) , dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (30.6 mg, 0.06435 mmol) and XPhos G3 (54.4 mg, 0.06435 mmol) up in thf (2.86 mL ) / water (300 ul ) under Ar.

Stirred at 60°C for 5 hours. Concentrated off THF and concentrated onto celite from toluene. Purified by isco 0-15% MeOH/DCM to give r t-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)methylene)piperidine-l-carboxylate (34-2) (65.0 mg, 0.1475 mmol, 34.5 % ) as an off-white solid. LCMS (ES+): m/z= 441 [M + H ]+

Intermediate 35-1. Preparation of r -Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3- dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)methyl)piperidine-l-carboxylate Scheme 35.

Brought t r t-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)methylene)piperidine-l-carboxylate (34-2) (62.0 mg, 0.141 mmol) up in DMF (2 mL ) / MeOH ( 1 mL ) and added a pinch of wetted 10% Pd/C. Evacuated vial with vac and backfilled with H 2 from a balloon. Stirred at r.t. for 24 hours periodically repurged solution with H2 from balloon. Filtered reaction mixture through celite and rinsed with a large amount of MeOH/DCM. Concentrated crude mixture and purified by isco 0-20% MeOH/DCM to give tert- butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4-c]pyridin-4- yl)methyl)piperidine-l-carboxylate (35-1) (28.0 mg, 0.06327 mmol, 45.0 %) as a white solid. LCMS (ES-): m/z= 441 [M - H]-

Intermediate 36-1. Preparation of 3-(l-Oxo-4-(piperidin-4-ylmethyl)-lH-pyrrolo[3,4- c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate Scheme 36.

Brought t r t-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)methyl)piperidine-l-carboxylate (35-1) (25 mg, 0.05649 mmol) up in DCM ( 1 mL ) and added 2,2,2-trifluoroacetic acid (250 µΐ 3.26 mmol). The reaction was stirred at r.t. for 1 hour, concentrated, dissolved in 1 mL DMSO and was purified by reversed phase isco 10-100% ACN/water + 0 .1% TFA. After lyophilization 3-(l-oxo-4-(piperidin-4-ylmethyl)-lH-pyrrolo[3,4- c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (36-1) (25.0 mg, 0.05477 mmol, 97.2 %) was isolated as a white solid. LCMS (ES+): m/z= 441 [M + H]+

Intermediate 37-2. Preparation of Methyl 3-(bromomethyl)-2-chloroisonicotinate Scheme 37.

Brought methyl 2-chloro-3-methylisonicotinate (37-1) (4.73g, 25.4 mmol), 1- bromopyrrolidine-2,5-dione (5.87 g, 33.0 mmol) and Propanenitrile, 2,2 (374 mg, 2.28 mmol) up in CC14 (84.6 mL ) under Ar. Refluxed reaction over night. Cooled reaction and partitioned between sodium bicarbonate and DCM. Dried organic layer over sodium sulfate and concentrated. Isolated methyl 3-(bromomethyl)-2-chloroisonicotinate (37-2) (6.00 g, 22.6 mmol, 89.4 % ) as a crude oil. Used material crude in the next reaction. LCMS (ES+): m/z= 264 [M + H ]+

Intermediate 38-2. Preparation of 3-(4-Chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)-3- methylpiperidine-2,6-dione Scheme 38.

Brought 3-methyl-2,6-dioxopiperidin-3-aminium chloride (38-1) (521 mg, 2.92 mmol) and methyl 3-(bromomethyl)-2-chloroisonicotinate (37-2) (0.86 g, 3.25 mmol) up in DMF (16.2 mL ) and added N-ethyl-N-isopropylpropan-2-amine (1.69 mL, 9.75 mmol) before heating the reaction to 60°C. After 24 hours reaction showed some product and mostly Sn2 intermediate. Increased temperature to 80°C for 24 hours. The reaction was then cooled and loaded directly onto reversed phase isco 10-100% ACN/water with 01.1% TFA. 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-

2(3H)-yl)-3-methylpiperidine-2,6-dione (38-2) (200 mg, 0.6809 mmol, 20.9 %) was isolated after lyophilization of the fractions as an oil. LCMS (ES+): m/z= 294 [M + H ]+

Intermediate 39-1. Preparation of r -Butyl 4-((2-(3-methyl-2,6-dioxopiperidin 2,3-dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)methyl)piperazine-l-carboxylate Scheme 39.

Brought 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)-3-methylpiperidine-2,6- dione (38-2) (120 mg, 0.4085 mmol) , trifluoroborate salt (14-1) (219 mg, 0.817 mmol) , cesium carbonate (397 mg, 1.22 mmol) , dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (29.2 mg, 0.06127 mmol) and XPhos G3 (51.8 mg, 0.06127 mmol) up in thf (2.72 mL ) / water (300 ul ) under Ar. Stirred at 60°C for 5 hours. Concentrated off THF and concentrated onto celite from toluene. Purified by isco 0-15% MeOH/DCM. After concentrating pure fractions isolated t rt-butyl 4-((2-(3-methyl-2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4-c]pyridin- 4-yl)methyl)piperazine-l-carboxylate (39-1) (125 mg, 0.2732 mmol, 67.2 %) as a white solid. LCMS (ES+): m/z= 458 [M + H]+

Intermediate 40-1. Preparation of 3-Methyl-3-(l-oxo-4-(piperazin-l-ylmethyl)-lH- pyrrolo [3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate Scheme 40.

Brought t rt-butyl 4-((2-(3-methyl-2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH- pyrrolo[3,4-c]pyridin-4-yl)methyl)piperazine-l-carboxylate (39-1) (125 mg, 0.2732 mmol) up in DCM ( 1 mL ) and added 2,2,2-trifluoroacetic acid (0.3 mL, 3.91 mmol). Stirred reaction at r.t. for 1 hour. Diluted reaction with toluene and concentrated. Isolated crude 3-methyl-3-(l-oxo-4- (piperazin-l-ylmethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (40-1) (120 mg, 0.2545 mmol, 93.7 %) as an oil. LCMS (ES+): m/z= 358 [M + H]+

Intermediate 41-3. Preparation of 2-((l-(ferf-Butoxycarbonyl)piperidin-4-yl)oxy)pyridine- 3,4-dicarboxylic acid Scheme 41. Sodium hydride (246 mg, 6.19 mmol) was added in small portions to a solution of tert- butyl 4-hydroxypiperidine-l-carboxylate (41-1) (297 mg, 1.48 mmol) and 2-chloropyridine-3,4- dicarboxylic acid (41-2) (.25 g, 1.24 mmol) in DMF (6.19 mL ) at 0 °C. The bath was removed and the reaction heated to 50 °C for 5 hours. After cooling, the reaction was quenched with citric acid, extracted with DCM (2 x 5 mL), dried over sodium sulfate and concentrated. The product was purified by column chromatography (silica, 0-10% MeOH in DCM) to give 2-{{\-{tert- butoxycarbonyl)piperidin-4-yl)oxy)pyridine-3,4-dicarboxylic acid (41-3) (355 mg, 78.1 %) LC/MS (ES+): m/z 389.1 [M+Na]+

Intermediate 42-1. Preparation of r -Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3- dihydro-l H -pyrrolo[3,4-c]pyridin-4-yl)oxy)piperidine-l-carboxylate 42.

2-((l-(ter t-Butoxycarbonyl)piperidin-4-yl)oxy)pyridine-3,4-dicarboxylic acid (41-3) (0.3 18 g, 0.8679 mmol) and amino-glutarimide (1-2) (0.8679 mmol) were taken up in ACN (5 mL ) and di(lH-imidazol-l-yl)methanone (308 mg, 1.90 mmol) was added. The white suspension was stirred for 2 hrs. The reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give t rt-butyl 4-((2-(2,6-dioxopiperidin-3-yl)- l,3-dioxo-2,3-dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)oxy)piperidine-l-carboxylate (42-1) (212 mg, 53.4 %) as a white solid. LC/MS (ES+): m/z 459.2 [M + H]+

Intermediate 43-1. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(piperidin-4-yloxy)-l H - pyrrolo [3,4-c]pyridine- 1,3(2H )-dione Scheme 43. t rt-Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)oxy)piperidine-l-carboxylate (42-1) (.06 g, 0.1308 mmol) was dissolved in a mixture of DCM (1.30 mL ) and 2,2,2-trifluoroacetic acid (0.300 mL, 3.91 mmol), stirred for 4 hours and concentrated to give 2-(2,6-dioxopiperidin-3-yl)-4-(piperidin-4-yloxy)-lH-pyrrolo[3,4- c]pyridine-l,3(2H)-dione 2,2,2-trifluoroacetate (43-1) (60.0 mg, 97.2 %) as a brown gum. LC/MS (ES+): m/z 359.3 [M + H]+

Intermediate 44-1. Preparation of 4-Chloro-2-(2,6-dioxopiperidin-3-yl)-l H-pyrrolo[3,4- c]pyridine-l,3(2 H)-dione Scheme 44.

3-Aminopiperidine-2,6-dione hydrochloride (1-2) (897 mg, 5.45 mmol) and 2- chloropyridine-3,4-dicarboxylic acid (41-2) ( 1 g, 4.96 mmol ) were taken up in ACN (40 mL ) and di(lH-imidazol-l-yl)methanone (1.76 g, 10.9 mmol) was added. The reaction was stirred for 16 hours and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in dCM) to give 4-chloro-2-(2,6-dioxopiperidin-3-yl)-lH-pyrrolo[3,4-c]pyridine-l,3(2H)- dione (44-1) (1.33 g, 91.7 %) as a white solid. LC/MS (ES+): m/z 294.1 [M + H]+

Intermediate 45-1. Preparation of tert- y 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3- dihydro-l H-pyrrolo[3,4-c]pyridin-4-yl)amino)piperidine-l-carboxylate Scheme 45. 4-Chloro-2-(2,6-dioxopiperidin-3-yl)-lH-pyrrolo[3,4-c]pyridine-l,3(2H)-dione (44-1) (.133 g, 0.4529 mmol), t rt-butyl 4-aminopiperidine-l-carboxylate (19-1) (99.7 mg, 0.4981 mmol) and DIEA (157 , 0.9058 mmol) were taken up in M P (4.52 mL ) and heated to 120°C for 2 hours in microwave reactor. The reaction was purified directly on reverse phase silica gel column chromatography (0-100% ACN in water) to give t rt-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3- dioxo-2,3-dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)amino)piperidine-l-carboxylate (45-1) (100 mg, 48.3 %) as a brown oil. LC/MS (ES+): m/z 402.6 [M-C4H ]+

Intermediate 46. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(piperidin-4-ylamino)- pyrrolo [3,4-c]pyridine- 1,3(2H )-dione Scheme 46-1.

t rt-Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)amino)piperidine-l-carboxylate (45-1) (0.1 g, 0.2185 mmol) was stirred in a mixture of DCM (1.09 mL ) and 2,2,2-trifluoroacetic acid (0.5 mL, 6.52 mmol) for 3 hours and concentrated to give 2-(2,6-dioxopiperidin-3-yl)-4-(piperidin-4-ylamino)-lH-pyrrolo[3,4- c]pyridine-l,3(2H)-dione 2,2,2-trifluoroacetate (46-1) (100 mg, 98.0 %) as a brown gum.

Intermediate 48-3. Preparation of l-(2,6-Dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxylic acid Scheme 48. To a stirred solution of t rt-butyl 2-oxo-2H-pyran-5-carboxylate (48-1) (3.0 g, 15.2 mmol) in Methanol (20.0 mL ) were added Triethylamine (3.28 mL, 22.8 mmol) and 3-Amino-piperidine- 2,6-dione hydrochloride (1-2) (2.50 g, 15.2 mmol). The reaction mixture was stirred at room temperature for 1 hour. It was concentrated under reduced pressure and diluted with ethyl acetate, water. Layers were separated and organic layer was washed with brine solution. It was dried over sodium sulfate and concentrated. Crude material was purified by column chromatography using (0%-l% MeOH/DCM) to afford rt-butyl l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6- dihydropyridine-3-carboxylate ( 1 g, 3.26 mmol, 21.5 %) as off white solid. LC/MS (ES+): m/z 307 [M + H]+ To a stirred solution of t rt-butyl l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine- 3-carboxylate (48-2) (900.0 mg, 2.93 mmol) in DCM (20.0 mL ) was added TFA (7.0 mL ) at 0°C and the reaction mixture was stirred at room temperature for 3 hours. It was concentrated under reduced pressure and stripped with DCM twice. Resultant material was triturated with Methanol- ether to afford l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid (48-3) (530 mg, 2.1 1 mmol, 72.3 %) as brown solid. LC/MS (ES+): m/z 251 [M + H]+

Intermediate 49-2. Preparation of 3-(2-Oxopiperazin-l-yl)piperidine-2,6-dione hydrochloride Scheme 49. A 50 mL 2 neck RB flask was charged with (Reverse addition protocol) tert-butyl 3- oxopiperazine-l-carboxylate (15-1) (520 mg, 2.60 mmol) in Tetrahydrofuran (10 mL ) was added sodium hydride (103 mg, 2.60 mmol) at 0°C. The reaction mixture was heated at 60°C for 30 mints. This hot reaction mass was added drop wise to a preheated (at 60oC), THF solution of 3- bromopiperidine-2,6-dione (47-2) (250 mg, 1.30 mmol). The reaction mixture was allowed to stir at the same temperature for another 3 hours. The RM was diluted with cold water and extracted with EtOAc (3 x 10 mL), washed with brine (10 mL), dried (Na2S0 4) and evaporated. The crude was purified by silica gel column chromatography by using 0-1% MeOH in EtOAc as eluting to provide t rt-butyl 4-(2,6-dioxopiperidin-3-yl)-3-oxopiperazine-l-carboxylate (49-1) (80.0 mg,

256 µιηοΐ , 19.8 %) as a Off-white solid. LC/MS (ES+): m/z 312 [M + H]+ A 50 mL 2 neck RB flask was charged with (R)-t rt-butyl 4-(2,6-dioxopiperidin-3-yl)-3- oxopiperazine-l-carboxylate (49-1) (50 mg, 160 µ οΐ) in 1,4-Dioxane ( 1 mL ) was added hydrochloric acid in dioxane (40.0 µΐ 160 µ οΐ) then stirred for 16 hours. The RM was concentrated under reduced pressure and was washed with n-pentane, dried under reduced pressure to provide (R)-3-(2-oxopiperazin-l-yl)piperidine-2,6-dione hydrochloride (49-2) (30.0 mg, 121 µιηοΐ , 75.7 %) go to next step. LC/MS (ES+): m/z 219 [M + H]+

Intermediate 50-3. Preparation of 3-(4-(4-Fluoropiperidin-4-yl)-l-oxoisoindolin-2- yl)piperidine-2,6-dione Scheme 50. To a solution of tert-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-4- hydroxypiperidine-l-carboxylate (50-1) (1.3 g, 2.93 mmol) in DCM (26 mL ) , Diethylaminosulfur trifluoride (DAST) (573 , 4.39 mmol) was added at 0 °C. The reaction was stirred at rt for lh. The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was quenched with water(20.0 mL) and extracted with DCM (25.0 mLx2). The organic layers were dried over sodium sulfate, filtered and concentrated to afford crude 50-2.The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-10%) to give tert-butyl 4-(2- (2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-4-fluoropiperidine-l-carboxylate (450 mg, 1.01 mmol, crude) with contamination of t r t-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)-3,6-dihydropyridine-l(2H)-carboxylate. The crude product (50-2) (58.51% by LCMS) as such taken for the next step with out further purification. LC/MS (ES+): m/z 446 [M + H]+ To a solution of tert-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)-4- fluoropiperidine-l-carboxylate (50-2) (300 mg, 673 µιηοΐ) in DCM (3.0 mL ) , 25% TFA in DCM (3.0 mL ) was added at 0 °C.The reaction mixture was stirred at rt for 2h. The reaction progress was monitored by TLC and LCMS analysis. After consumption of starting material the solvent was evaporated to dryness and triturated with diethylether(20.0 mL) to afford 3-(4-(4- fluoropiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione with TFA salt (50-3) (250 mg, 544 µπιοΐ, crude). This compound as such taken for the next step without further purification. LC/MS (ES+): m/z 346 [M + H]+

Intermediate 51-3. Preparation of 3-(5-Oxo-2-(piperazin-l-ylmethyl)-5 ,7-dihyd pyrrolo[3,4-b]pyridin-6-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate Scheme 51. To a stirred solution of methyl 2-(bromomethyl)-6-chloronicotinate (51-1) (2.6 g, 4.18 mmol) in ACN (26 mL ),3-aminopiperidine-2,6-dione hydrochloride (1-2) (824 mg, 5.01 mmol) was added after that dropwise DIEA (3.23 g, 25.0 mmol) was added to the reaction mixture and reaction stirred for 3 hr at 85°C. The progress of reaction monitored by TLC. The reaction mixture was dilluted with 150 mL of water and extracted with DCM (200mlx3) and dried over Na2S04 and concentrated under vacuum, after that crude reaction mass dilluted with ethylacetate and triturated with ether (50ml x 3) and filtered the reaction mass and dried under vacuum to give 3-(2-chloro- 5-oxo-5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6-dione (51-2) (810 mg, 2.89 mmol, 69.8 %) as a solid. LCMS (ES-): m/z= 278 [M-H]- To a stirred solution of 3-(2-chloro-5-oxo-5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine- 2,6-dione (51-2) (0.5 g, 1.78 mmol) in Dioxane:Water (10ml),potassium ((4-(tert- butoxycarbonyl)piperazin-l-yl)methyl)trifluoroborate (14-1) (652 mg, 2.13 mmol), Cesium Carbonate (1.73 g, 5.34 mmol) was added to the reaction mixture, reaction mixture was flushed with N2 gas at least for 5 minute after that n-butyl diadamantyl phosphine (127 mg, 356 µ οΐ) and palladium acetate (3.99 mg, 17.8 µ οΐ) was added after that reaction mixture was again flushed with N2 gas at least for 5 minute then reaction subjected to heating for 2hr at 90°C. The progress of reaction monitored by TLC and LCMS. The reaction mixture was dilluted with 50 mL of water and extracted with ethyl acetate (30mlx3) and dried over Na2S04 and concentrated under vaccum.The product was purified by silica gel flash chromatography (12gmColumn, DCM/MeOH 0-10%) to give t rt-butyl 4-((6-(2,6-dioxopiperidin-3-yl)-5-oxo-6,7-dihydro-5H-pyrrolo[3,4- b]pyridin-2-yl)methyl)piperazine-l-carboxylate (300 mg, 676 µιηοΐ , 38.0 %). LCMS (ES+): m/z= 444 [M + H]+ To a stirred solution of t rt-butyl 4-((6-(2,6-dioxopiperidin-3-yl)-5-oxo-6,7-dihydro-5H- pyrrolo[3,4-b]pyridin-2-yl)methyl)piperazine-l-carboxylate (0.250g, 563 µ οΐ) in DCM (2.5 mL

),25% TFA EST DCM(2.5 mL ) was added dropowise at 0°C. The progress of reaction monitored by TLC and LCMS. The reaction mixture was directly concentrated and triturated with ether (20ml x 3)and dried under vacuum to give 3-(5-oxo-2-((4-(2,2,2-trifluoroacetyl)piperazin-l-yl)methyl)-

5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6-dione (51-3) (291 mg, 525 µιηοΐ , 93.5 %). LCMS (ES+): m/z= 344 [M + H]+ Intermediate 52-3. Preparation of 3-(7-Oxo-2-(piperazin-l-ylmethyl)-5,7-dihyd pyrrolo[3,4-b]pyridin-6-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate Scheme 52.

To a solution of methyl 3-(bromomethyl)-6-chloropicolinate (52-1) (2.4 g, 9.07 mmol) in ACN (30 mL ) under nitrogen atmosphere added DIEA (7.03 g, 54.4 mmol) , 3-aminopiperidine- 2,6-dione hydrochloride (1-2) (1.77 g, 10.8 mmol) .Then refluxed the reaction mixture at 90°C for 18h.The reaction progress was monitored by TLC and LCMS. After consumption of SM poured (50mL) diethyl ether and filtered directly through sintered funnel and washed with di ethyl ether (20mLx5) to obtain 3-(2-chloro-7-oxo-5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,5-dione

(52-2) ( 1 g, 3.57 mmol, 39.5 %) as blue colored fluffy solid. LCMS (ES+): m/z= 279 [M + H]+ To a solution of 3-(2-chloro-7-oxo-5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6- dione (52-2) (350 mg, 1.25 mmol) in Dioxane (4 mL ) , H20 ( 1 mL ) in (4:1) added cesium carbonate (1.21 g, 3.75 mmol) and purged nitrogen for 15 min.After purging added Pd(oAc)2 (28.0 mg, 125 µ οΐ) , n-butyl diadamantyl phosphine (89.6 mg, 250 µ οΐ) and purged nitrogen for 5 min.Heated and stirred reaction at 100°C for lh. After consumption of SM quenched the reaction with water (20mL) and extracted with ethyl acetate (20x3mL). Combined organic layers and dried over anhydrous sodium sulfate,Filtered and distilled .The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-10%) to give r t-butyl 4-((6-(2,6- dioxopiperidin-3-yl)-7-oxo-6,7-dihydro-5H-pyrrolo[3,4-b]pyridin-2-yl)methyl)piperazine-l- carboxylate (250 mg, 563 µιηοΐ , 45.1 %) as off white solid. LCMS (ES+): m/z= 444 [M + H]+ To a solution of tert-butyl 4-((6-(2,6-dioxopiperidin-3-yl)-7-oxo-6,7-dihydro-5H- pyrrolo[3,4-b]pyridin-2-yl)methyl)piperazine-l-carboxylate (150 mg, 338 µ οΐ) in DCM (2 mL ) at RT under nitrogen atmosphere added 25% TFA IN DCM (6mL, 338 µιηοΐ) .Stirred the RM at RT for 2h. After consumption of SM directly distilled RM under reduced pressure and washing with di ethyl ether(5x3mL) was given to obtain 3-(7-oxo-2-(piperazin-l-ylmethyl)-5H- pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6-dione (WITH TFA SALT) (52-3) (150 mg, 338 µιηοΐ , 100 %) as brown colored solid. LCMS (ES+): m/z= 344 [M + H]+

Intermediate 53-2. Preparation of (2-Amino-5-bromophenyl)(4-chlorophenyl)methanone Scheme 53.

Brought (2-aminophenyl)(4-chlorophenyl)methanone (53-1) (4.7 g, 20.2 mmol) up in DCM (75 mL ) and cooled reaction to 0°C. Added B S (3.59 g, 20.2 mmol) in one portion and let the reaction gradually warm to r.t. Stirred reaction O/N and was complete by lc/ms. Diluted the reaction with DCM and water and extracted. Washed organic layer with brine and dried over sodium sulfate. Isolated (2-amino-5-bromophenyl)(4-chlorophenyl)methanone (53-2) (5.4 g, 17.3 mmol, 86. 1 %) as a yellow crude solid. LCMS (ES+): m/z= 310 [M + H]+

Intermediate 54-2. Preparation of r -Butyl (l-((4-bromo-2-(4- chlorobenzoyl)phenyl)carbamoyl)cyclopropyl)carbamate Scheme 54.

Brought (2-amino-5-bromophenyl)(4-chlorophenyl)methanone (53-2) (3.78 g, 12.1 mmol), N-ethyl-N-isopropylpropan-2-amine (3.12 g, 24.2 mmol) and \-{{tert- butoxycarbonyl)amino)cyclopropanecarboxylic acid (54-1) (2.43 g, 12.1 mmol) up in DMF (5 mL) and cooled to 0°C. Added HATU (4.60 g, 12.1 mmol) and let gradually warm to r.t. O/N. stirred for 8 days. Diluted into EA and washed with water 2x followed by 5% Aq LiCl 2x. Dried over sodium sulfate concentrated purified by isco 0-40% EA/hx to give t rt-butyl (l-((4-bromo- 2-(4-chlorobenzoyl)phenyl)carbamoyl)cyclopropyl)carbamate (54-2) (3.42 g, 6.92 mmol, 57.2 %) as a solid. LCMS (ES+): m/z= 437 [M - C4H ]+

Intermediate 55-1. Preparation chlorobenzoyl)phenyl)cyclopropanecarboxamide Scheme 55.

Brought tert-butyl (l-((4-bromo-2-(4- chlorobenzoyl)phenyl)carbamoyl)cyclopropyl)carbamate (54-2) (3.52 g, 7.12 mmol) up in chloroform (16 mL ) and added 4M hydrogen chloride (8.90 mL, 35.6 mmol) in dioxane. Stirred at r.t. overnight. Concentrated solution and partititoned between DCM and sat. sodium bicarbonate solution. Washed Aqueous layer 2x with DCM. Combined organics and dried over sodium sulfate before concentrating to solid l-amino-N-(4-bromo-2-(4- chlorobenzoyl)phenyl)cyclopropanecarboxamide (55-1) (2.80 g, 7.1 1 mmol, 100 %) which was off-white and used crude. LCMS (ES+): m/z= 395 [M + H]+

Intermediate 56-1. Preparation of Bromo-5-(4-chlorophenyl)spiro[benzo[e][l,4]diazepine- 3 1 Brought 1-amino-N-(4-bromo-2-(4-chlorobenzoyl)phenyl)cyclopropanecarboxamide (55- 1) (2.80 g, 7.12 mmol) up in EtOH (71.2 mL ) and added acetic acid (10.6 mL, 178 mmol) before heating to 80°C overnight. Cooled reaction and concentrated to ~ 1/3 volume. Collected ppt. to give 7-bromo-5-(4-chlorophenyl)spiro[benzo[e][l,4]diazepine-3,l (56-1) (1.60 g, 4.25 mmol, 59.9 %) as an off-white solid. LCMS (ES+): m/z= 377 [M + H]+

Intermediate 57-1. Preparation of 7-Bromo-5-(4-chlorophenyl)spiro[benzo[e][l,4]diazepine- 3,l'-cyclopropane]-2(lH)-thione Scheme 57.

Brought 7-bromo-5-(4-chlorophenyl)spiro[benzo[e][l,4]diazepine-3,l'-cyclopropan]- 2(lH)-one (56-1) (1.65 g, 4.39 mmol) up in thf (29.2 mL ) and added 2,4-bis(4-methoxyphenyl)- 1,3,2,4-dithiadiphosphetane 2,4-disulfide (974 mg, 2.41 mmol) and refluxed overnight. Cooled to r.t. and concentrated from toluene 2x to give crude 7-bromo-5-(4- chlorophenyl)spiro[benzo[e][l,4]diazepine-3,l'-cyclopropane]-2(lH)-thione (57-1) (1.71 g, 4.36 mmol, 100 %) as a solid. LCMS (ES+): m/z= 393 [M + H]+

Intermediate 58-1. Preparation of 8-Bromo-6-(4-chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l Scheme 58. Brought 7-bromo-5-(4-chlorophenyl)spiro[benzo[e][l,4]diazepine-3, 1(57-1) (1.71 g, 4.39 mmol) up in THF (43.8 mL ) added hydrazine hydrate (2.12 mL, 43.8 mmol) and stirred for 1 hour. Concentrated reaction down and redissolved in toluene (43.8 mL) and added Triethyl acetate (10 mL ) . Heated reaction to 100 °C for 6 hours. Saw some product and some uncyclized intermediate concentrated to dryness and redissolved in only toluene. Heated O/N. Cooled to r.t. and began concentrating. Precipitate formed at approximately ½ the solvent volume. Filtered to give 8-bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l

(58-1) (1.5 g, 3.63 mmol, 82.6 %) as a light yellow solid. LCMS (ES+): m/z= 413 [M + H]+

Intermediate 59-2. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 59.

8-Bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,1 (58-1) (0.55 g, 1.32 mmol) l-methyl-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-lH- pyrazole (274 mg, 1.32 mmol)and potassium carbonate (547 mg, 3.96 mmol) were dissolved in dioxane (5.28 mL) and water (2.64 mL) (Ar was bubbled through solvents for 10 minutes pri or) tetrakis (86.2 mg, 0.07468 mmol) was added and the vial was purged with Ar and sealed. The reaction was stirred at 95°C for 2 hours, at which time LCMS showed reaction was complete. Azeotroped reaction onto celite from toluene and purified on isco 0 - 10% MeOH/DCM to give 6- (4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropane] (59-2) (280.0 mg, 0.698 mmol, 52.9%) as an off-white solid. + LCMS (ES+): m/z= 401 [M + H] Intermediate 60-2. Preparation of (S)-ferf-Butyl (l-((4-bromo-2-(4- chlorobenzoyl)phenyl)amino)-l-oxopropan-2-yl)carbamate Scheme 60.

Brought (S)-2-((t rt-butoxycarbonyl)amino)propanoic acid (60-1) (3.27 g, 17.3 mmol) up in DMF (lOOmL) N-ethyl-N-isopropylp (4.47 g,34.6 mmol) was added at 0°C followed by HATU (7.87 g, 20.7 mmol). Stirred this under Ar at reduced temp for 20-30 minutes before adding Aniline (53-2) (5.4 g,17.3 mmol) and letting the reaction warm to r.t. Stirred at r.t. for 5 nights. Diluted with EA and washed with water 2x followed by 5% Aq LiCl (2x) to remove DMF. purified by isco 5-40% EA/hx to give (S)-t r t-butyl (l-((4-bromo-2-(4-chlorobenzoyl)phenyl)amino)-l- oxopropan-2-yl)carbamate (60-2) (3.7 g, 7.68 mmol, 44.4%) as a solid. LCMS (ES+): m/z= 424

[M - C4H ]+

Intermediate 61-1. Preparation of (S)-7-Bromo-5-(4-chlorophenyl)-3-methyl-l,3-dihydro-

2H-benzo [e] [1,4] diazepin-2-one Scheme 61.

Brought (S)-ter t-butyl (l-((4-bromo-2-(4-chlorobenzoyl)phenyl)amino)-l-oxopropan-2- yl)carbamate (60-2) (6.36 g, 13.2 mmol) up in chloroform (25 mL) and added 4 M HC1 in dioxane (13.2 mL, 52.8 mmol). Stirred for 4-5 hours at r.t. no SM by uplc. Concentrated and brought up in EtOAc. Washed 3x with sat NaHC03 and dried org over sodium sulfate before concentrating to crude (S)-7-bromo-5-(4-chlorophenyl)-3-methyl-l,3-dihydro-2H-benzo[e][l,4]diazepin-2-one (61-1) (4.6 g, 12.0 mmol, 91.4%) as an off-white solid. LCMS (ES+): m/z= 383 [M + H]+ Intermediate 62-1. Preparation of (S)-7-Bromo-5-(4-chlorophenyl)-3-methyl-lH- benzo [e] [1,4] diazepin-2(3H)-one Scheme 62.

Brought (S)-2-amino-N-(4-bromo-2-(4-chlorobenzoyl)phenyl)propanamide (61-1) (1.93 g, 5.06 mmol) up in MeOH/water and added NaOH (1M, 1.0 mL) supposedly to pH 8.5 may be a bit more basic then that. Stirred at r.t. for ~ 4 hours. No SM by UPLC. concentrated off the MeOH, added ~ 40 mL water and washed with 3x DCM, 2x chloroform and 2x EA, much emulsion and seemed there may have been ppt around. Dried combined orgs over sodium sulfate and concentrated. Brought up in ~ 30-40 mL MeOH and ~ 10 mL water and heated to 50°C. Almost all of the yellowish ppt dissolved. The flask was then cooled gradually to r.t. and then in an ice bath. Filtered ppt, Crop 1 (l.Og). Then concentrated off some of the MeOH and added ~ 5-10 mL more water cooled in an ice bath and filtered crop 2 . Dried on the high vacuum over the weekend to give (S)-7-bromo-5-(4-chlorophenyl)-3-methyl-lH-benzo[e][l,4]diazepin-2(3H)-one (62-1) (1.3 g, 3.57 mmol, 71.0 %) as a off-white solid. LCMS (ES+): m/z= 363 [M + H]+

Intermediate 63-1. Preparation of (S)-8-Bromo-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 63. Brought (S)-7-bromo-5-(4-chlorophenyl)-3-methyl-lH-benzo[e][l,4]diazepin-2(3H)-one (62-1) (185 mg, 0.5087 mmol) up in THF (1.0 mL) under Ar and cooled to -78°C. Added potassium 2-methylpropan-2-olate (62.7 mg, 0.5595 mmol) in one portion and stirred for 10 mins at -78 then warmed to -IOC for 30 minutes followed by r.t. for 30 minutes. Recooled to -78°C and added diethoxyphosphinic chloride (155 mg, 0.6104 mmol) in one portion after 15 minutes warmed to -10 C for 45-60 minutes and then added acetohydrazide (52.7 mg, 0.7121 mmol). Let vessel warm to r.t. and stirred for 1 hour at which time added 2 mL of n-BuOH and heated reaction to 95°C for 2-3 hours. Then stirred at r.t. O/N, concentrated and purified by isco to give (S)-8- bromo-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine (63-1) ( 110.0 mg, 0.274 mmol, 53.9 %) as an amorphous solid. LCMS (ES+): m/z= 402 [M + H]+

Intermediate 64-1. Preparation of (S)-6-(4-Chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)- 4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 64.

(S)-8-Bromo-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine (63-1) (0.200 g, 0.4978 mmol) rt-butyl 4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole-l-carboxylate (146 mg, 0.4978 mmol) and potassium carbonate (137 mg, 0.9956 mmol) were dissolved in dioxane (1.99 mL) and water (995 (Ar was bubbled through solvents for 10 minutes prior). Tetrakis (57.5 mg, 0.04978 mmol) was added and the vial was purged with Ar and sealed. The reaction was stirred at 95°C for 2 hours, at which time LCMS showed reaction was complete. Azeotroped reaction onto celite from toluene and purified by isco 0 - 10% MeOH/DCM to give (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (165.0 mg, 0.426 mmol, 85.5 %) as a white solid. LCMS (ES+): m/z= 389 [M + H]+ Intermediate 65-2. Preparation of 4-Methoxyfuro[3,2-c] pyridine Scheme 65.

4-Chlorofuro[3,2-c]pyridine (65-1) (12.3 g, 80.0 mmol) in 80 mL DMF was added via cannula to a flask containing sodium methoxide (6.53 g, 121 mmol) and the reaction heated to 110 °C for 2 hrs. After cooling, the reaction was concentrated and the residue taken up in water and extracted with DCM (2 x 150 mL). The organic layers were combined, washed with brine, and dried over sodium sulfate. Celite was added to the filtered solution and the solvent removed. The crude reaction was purified by column chromatography (silica, 0-10% gradient EtOAc in hexane) to give 4-methoxyfuro[3,2-c]pyridine (65-2) (8.70 g, 58.3 mmol, 73.1 %) as a white solid. LC/MS (ES+): m/z 150.0 [M + H]+ Intermediate 66-1. Preparation of 4-Methoxyfuro[3,2-c]pyridine-2-carbaldehyde Scheme 66

S5~2 66 1

4-Methoxyfuro[3,2-c] (65-2) (2.6 g,17.4 mmol) was dissolved in THF (70 mL) and cooled to -78°C. n-Butyllithium (1.66 g,26.0 mmol) was added and the solution allowed to stir at -78°C for 15 minutes. The bath was removed and the solution was allowed to warm for 15 minutes before being cooled back down to -78°C. DMF (2.54 g,34.8 mmol) was added as a solution in THF (3 mL) and the reaction stirred for 15 minutes at -78°C and lhr at room temp. The solution was then neutralized with sat sodium bicarbonate and extracted with ether (2 x 50 mL). The combined organics are washed with brine and dried over sodium sulfate.The solid residue was recrystallized from hexane to give 4-methoxyfuro[3,2-c]pyridine-2-carbaldehyde (66-1) (2.77 g, 15.6 mmol, 89.9 %) as yellow needles. LC/MS (ES+): m/z 179.0 [M + H]+

Intermediate 67-1. Preparation of 4-Oxo-4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde Scheme 67. 4-Methoxyfuro[3,2-c] (66-1) (2 g,l 1.2 mmol) was heated to reflux in 1 M HC1 ( 11 mL) for three hours. The reaction was cooled and the solid collected and recrystallized from dioxane to give 4- oxo-4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde (67-1) (1.62 g, 9.93 mmol, 89.0 %) as an brown solid. LC/MS (ES+): m/z 164.0 [M + H]+

Intermediate 68-1. Preparation of 7-Bromo-4-oxo-4,5-dihydrofuro[3,2-c]pyridine-2- carbaldehyde Scheme 68.

N-Bromosuccinimide (2.17 g, 12.2 mmol) was added portionwise to a stirred suspension of 4-oxo- 4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde (67-1) (1.6 g, 9.80 mmol) in THF (24.5 mL ) The reaction mixture was stirred at room temperature for 6 hours. The solvent was evaporated and the residue was triturated with diethyl ether. The solid was filtered off, washed with diethyl ether and dried to give 7-bromo-4-oxo-4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde (68-1) (2.30 g, 9.50 mmol, 97.0 %) as a light brown solid. LC/MS (ES+): m/z 241.9 [M + H]+

Intermediate 69-1. Preparation of 7-Bromo-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridine- 2-carbaldehyde Scheme 69. Methyl iodide (1.17 mL, 19.0 mmol) was added to a stirred suspension of 7-bromo-4-oxo- 4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde (68-1) (2.3 g, 9.50 mmol) and caesium carbonate (9.25 g, 28.4 mmol) in THF (47.4 mL ) . The reaction mixture was stirred at room temperature overnight. More methyl iodide (1.17 mL, 19.0 mmol) was added and the reaction stirred for 8 additional hours. The solvent was removed and the residue taken up in 50 mL water and stirred for one hour. The solid was filtered and dried to give 7-bromo-5-methyl-4-oxo-4,5-dihydrofuro[3,2- c]pyridine-2-carbaldehyde (69-1) (2.22 g, 8.67 mmol, 91.3 %) as a yellow solid. LC/MS (ES+): m/z 256.0 [M + H]+

Intermediate 70-2. Preparation of tert-Butyl (R)-4-((7-bromo-5-methyl-4- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate Scheme 70.

7-Bromo-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridine-2-carbaldehyde (69-1) ( 1 g, 3.90 mmol) and (R)-t r t-butyl 3-methylpiperazine-l-carboxylate (70-1) (0.935 g, 4.67 mmol) were dissolved in DCE (26 mL) and acetic acid (0.266 mL, 4.67 mmol) was added. The reaction was stirred for 30 min and sodium triacetoxyborohydride (4.13 g, 19.5 mmol) was added portionwise. After stirring for 16 hours the reaction was quenched by the slow addition of sat. sodium bicarbonate (100 mL) and extracted with DCM (2 x 100 mL). The combined organics were washed with brine, dried over sodium sulfate and concentrated. The residue was purified by column chromotagraphy (silica, 0-10 % MeOH in DCM) to give (R)-t rt-butyl 4-((7-bromo-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate (70.2) (1.3 g, 76 %) as a white solid. LC/MS (ES+): m/z 440.8 [M + H]+

Intermediate 71-3. Preparation of 4-Bromo-3-(cyclopropylmethoxy)pyridine Scheme 71. Sodium hydride (288 mg, 7.23 mmol) was add to 4-bromopyridin-3-ol (71-1) (1.05 g, 6.03 mmol) in DMF (30 mL) at 0 °C and stirred for 1 hr. (bromomethyl)cyclopropane (71-2) (701 , 7.23 mmol) was added and the reaction stirred overnight at room temperature. Water was added (100 mL) and the reaction extracted with EtOAc (2 x 100 mL), washed with brine, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (silica, 0- 10% MeOH in DCM) to give 4-bromo-3-(cyclopropylmethoxy)pyridine (71-3) (.766 g, 56%) of a brown oil. LC/MS (ES+): m/z 228.0 [M + H]+

Intermediate 72-2. Preparation of 3-(Cyclopropylmethoxy)-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)pyridine Scheme 72

4-Bromo-3-(cyclopropylmethoxy)pyridine (71-3) (0.3 g, 1.31 mmol), bis(piacolato)diboron (72-1) (431 mg, 1.70 mmol) and Potassium acetate (385 mg, 3.93 mmol) were taken up in dioxane (4.4 mL) in a sealable tube equipped with a septa and degassed by bubbling nitrogen for 15 min. PdCl2(dppf) (47.9 mg, 65.5 µ οΐ) was added and the septa was replaced with a screw cap. The reaction was heated at 80 °C for 8 hours and allowed to cool, diluted with EtOAc, filtered and concentrated to give 3-(cyclopropylmethoxy)-4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine (72-2) (0.360 g 100 %) as a brown oil. 100 % yield assumed . The product was used in the next step without further purification. LC/MS (ES+): m/z 193.9 [M + H]+ Intermediate 73-1. Preparation of tert-Butyl (R)-4-((7-(3-(cyclopropylmethoxy)pyridin-4- yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazine-l- carboxylate Scheme 73.

(R)-ter t-Butyl 4-((7-bromo-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazine-l-carboxylate (70-2) (.147 g, 333 µηιοΐ), 3-(cyclopropylmethoxy)-4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine (72-2) ( 118 mg, 432 µηιοΐ), tripotassium orthophosphate (1.4 mL, 700 µηιοΐ , 0.5 M) and THF (.7 mL) were added to a screw top reaction tube equipped with a septa. Nitrogen was bubbled through the solution for 15 min. XPhos-Pd-G3

(8.45 mg, 9.99 µιηοΐ) was added, the septa was replaced by a screw cap and the reaction was heated at 80 °C for 12 hrs. The reaction was concentrated and the residue was purified by column chromatography (silica, 0-10% MeOH in DCM) to give (R)-tert-butyl 4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-

3-methylpiperazine-l-carboxylate (73-1) (90.0 mg, 176 µπιοΐ , 53.2 %) as a white solid. LC/MS (ES+): m/z 509.5 [M + H]+

Intermediate 74-1. Preparation of (R)-7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2- ((2-methylpiperazin-l-yl)methyl)furo[3,2-c]pyridin-4(5 H)-one Scheme 74. (R)-ter t-Butyl 4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2- c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate (73-1) (.067 g, 0 .13 17 mmol) was taken up in dioxane (526 µΐ ) and hydrogen chloride (.524 mL, 2.09 mmol, 4 M dioxane) was added. After 1 hour the solvent was removed to give (R)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5- methyl-2-((2-methylpiperazin-l-yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (74-1) (50.0 mg, 0.1 123 mmol, 85.3 %)as a white solid. LC/MS (ES+): m/z 408.8 [M + H]+

Intermediate 75-1. Preparation of 6-(4-Chlorophenyl)-8-(l-(3-chloropropyl)-lH-pyrazol-4- yl)-l-methylspiro [benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 75.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (O.lg, 0.2494 mmol) and (121 mg, 0.3741 mmol) up in DMF (12.4 mL ) at r.t. Added l-bromo-3-chloropropane (29.6 , 0.2992 mmol) and stirred at r.t. for 16 hours. Concentrated reaction from toluene 3x and purified directly by isco 0-10% MeOH/DCM to give 6-(4-chlorophenyl)-8-(l-(3-chloropropyl)-lH-pyrazol-4-yl)- l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (75-1) (0.090 g, 0.1885 mmol, 76 %) as an oil. LCMS (ES+): m/z 478 [M + H]+

Intermediate 76-1. Preparation of 8-(l-(4-Chlorobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 76. Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (O.lg, 0.2494 mmol) and cesium carbonate (121 mg, 0.3741 mmol) up in DMF (12.4 mL ) at r.t. Added l-bromo-4-chlorobutane (34.4 , 0.2992 mmol) and stirred at r.t. for 16 hours. Concentrated reaction from toluene 3x and purified directly by isco 0-10% MeOH/DCM to give 8-(l-(4-chlorobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (76-1) (75 mg, 0.1526 mmol, 61.4%) as an oil. LCMS (ES+): m/z 491.6 [M + H]+

Intermediate 77-1. Preparation of 8-(l-(5-Chloropentyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 77.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (O.lg, 0.2494 mmol) and cesium carbonate (121 mg, 0.3741 mmol) up in DMF (831 µΙ_, ) at r.t. Added l-bromo-5-chloropentane (39.2 0.2992 mmol) and stirred at r.t. for 16 hours before concentrating from toluene onto celite. Purified by isco 0-10% MeOH/DCM to give 8-(l-(5-chloropentyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (77-1) (85.0 mg, 0.2494 mmol, 67.4 % yield) as an oil. LCMS (ES+): m/z 507 [M + H]+ Intermediate 78-1. Preparation of 8-(l-(6-Chlorohexyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 78.

Brought -(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (108.0 mg, 0.2694 mmol) and cesium carbonate ( 87.7 mg, 0.2694 mmol) up in Acetonitrile (2.69 mL) and stirred at r.t for 24 hours. Concentrated solution and purified by isco 0-10% MeOH/DCM to give 8-(l-(6- chlorohexyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l' -cyclopropane] (78-1) ( 110 mg, 0.21 17 mmol, 79.1 %) as an oil. LCMS (ES+): m/z 521 [M + H]+

Intermediate 79-2/3. Preparation of r -Butyl 4-(6-chlorohexyl)piperazine-l-carboxylate Scheme 79.

Brought t r t-butyl piperazine-l-carboxylate (79-1) (1.13 g, 6.06 mmol) up in dioxane (20.2 mL) and added l-bromo-7-chloroheptane (1.48 mL, 9.08 mmol) followed by DIEA (1.57 mL, 9.08 mmol) stirred O/N at 70°C. Complete after one night of stirring, mix of Br and CI product.

Concentrated down and purified by isco 0-20% MeOH/DCM to give 1.5 g of the mixture of Alkyl halides (79-2 and 3) as an oil, used as the mixture. LCMS (ES+): m/z 319.4 (CI) and 353.4 (Br) [M + H]+

Intermediate 80-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(7-(piperazin-l- yl)heptyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] as its TFA salt Scheme 80.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (50mg, 0.1247 mmol) and cesium carbonate (60.9 mg, 0.1870 mmol) up in DMF (1.24 mL ) at r.t. Added tert-butyl 4-(7-bromo(and chloro)heptyl)piperazine-l-carboxylate (79-2/3) (55.0 mg, 0.1496 mmol) and stirred at r.t. for 16 hours. Complete by lcms. Concentrated onto celite and purified by isco 0-15% MeOH/DCM. Isolated tert-butyl 4-(7-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptyl)piperazine-l-carboxylate (50.0 mg, 0.07317 mmol, 58.6 %) as an oil. LCMS (ES+): m/z 683.6 [M + H]+ Brought tert-butyl 4-(7-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptyl)piperazine-l-carboxylate (49.9 mg, 0.07317 mmol) up in DCM (731 ) and added 2,2,2-trifluoroacetic acid (210 , 2.73 mmol). Stirred reaction for 3 hours and concentrated from toluene 3x to give 6-(4-chlorophenyl)- 1-methyl-8-(l -(7-(piperazin- 1-yl)heptyl)- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (80-1) (48.0 mg, 0.06884 mmol, 94.1 %) as a crude oil. LCMS (ES+): m/z 584 [M + H]+. Intermediate 81-1. Preparation of r -Butyl 4-(6-(4-(6-(4-chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazine-l-carboxylate Scheme 81.

To a solution of 8-(l-(6-chlorohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4, -cyclopropane] (78-1) (400 mg, 770 µηιοΐ) in DMF(15 mL) was added potassium iodide (253 mg, 1.53 mmol), potassium carbonate (21 1 mg, 1.53 mmol) and t r t-butyl piperazine-l-carboxylate (284 mg, 1.53 mmol) . The mixture was heated to 90 °C overnight. Water(100 mL) and EA(50 mL*2) were added. The Combined organic layers was dried over N a2SC"4 and concentrated, purified with pre- TLC(DCM:MeOH=10:l) to give tert-butyl 4-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperazine-l-carboxylate (81-1) (367 mg, 548 µιηοΐ , 71.2 %). LCMS (ES+): m/z=669 [M + H ]+

Intermediate 82-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(6-(piperazin-l- yl)hexyl)-lH-pyrazol-4-yl)spiro [benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '- cyclopropane] Scheme 82. A solution of t r t-butyl 4-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperazine-l-carboxylate (81-1) (350 mg, 522 µιηοΐ) in MeOH/HCl(3M) (10 mL) was stirred for 4h at rt. The crude (82-1) was concentrated and used directly. LCMS (ES+): w/z=568 [M + H ]+

Intermediate 83-1. Preparation of 8-(l-(5-Chloropentyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] heme 83.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (200 mg, 498

µιηοΐ), Cesium carbonate (194 mg, 597 µιηοΐ) up in DMF (5ml ) and added l-bromo-5- chloropentane ( 111 mg, 0.6 mmol). Stirred reaction at r.t. for 4 hours. Used crude material of 8- ( 1-(5-chloropentyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (83-1) directly after aqueous workup. LCMS (ES+): m/z= 505 [M + H ]+ Intermediate 84-1. Preparation of r -Butyl 4-(5-(4-(6-(4-chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)piperazine-l-carboxylate Scheme 84.

Brought 8-(l-(5-chloropentyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (83-1) (370 mg, 732

µηιοΐ), ter t-butyl piperazine-l-carboxylate (79-1) (271 mg, 1.46 mmol), Potassium carbonate (201 mg, 1.46 mmol) Potassium iodide (242 mg, 1.46 mmol) up in DMF (5ml). Stirred reaction at 80°C for 16 hours. The crude material was concentrated and purified by prep TLC

(DCM/MeOH=10/l)to afford tert-butyl 4-(5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentyl)piperazine-l-carboxylate (84-1) (450 mg, 686 µιηοΐ , 93.9 % ) as white solid. LCMS (ES+): m/z= 492 [M + H ]+

Intermediate 85-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(5-(piperazin-l- yl)pentyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 85. Brought tert-butyl 4-(5-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)pentyl)piperazine- 1-carboxylate (85-1)

(400mg, 610 µηιοΐ) up in 3M HC1 (10 mL) and stirred for 4 hours at r. . Concentrated the reaction to solid (85-1) and used directly in the next step. LCMS (ES+): m/z= 556 [M + H]+

Intermediate 86-1. Preparation of 8-(l-(4-Chlorobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 86.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (59-2) (400 mg, 997 µηιοΐ) in DMF(5 mL) was added cesium carbonate (485 mg, 1.49 mmol) and l-bromo-4- chlorobutane (204 mg, 1.19 mmol) . The mixture was stirred overnight at rt. Water(50 mL) and

EA(60 mL*2) were added. The Combined organic layers was dried over Na2S04 and concentrated, purified with pre-TLC(EA=100%) to give 8-(l-(4-chlorobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (86-1) (277 mg, 565 µιηοΐ , 56.6 %). LCMS (ES+): m/z= 491 [M + H]+

Intermediate 87-1. Preparation of r -Butyl 4-(4-(4-(6-(4-chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)piperazine-l-carboxylate Scheme 87. To a solution of 8-(l-(4-chlorobutyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (86-1) (280 mg, 569 µηιοΐ) in DMF(10 mL) was added potassium iodide (187 mg, 1.13 mmol), potassium carbonate (156 mg, 1.13 mmol) and ter t-butyl piperazine-l-carboxylate (79-1) (210 mg, 1.13 mmol) . The mixture was heated to 90 °C overnight. Water(100 mL) and EA(50 mL*2) were added. The

Combined organic layers was dried over N a2SC"4 and concentrated, purified with pre- TLC(DCM:MeOH=10:l) to give tert-butyl 4-(4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butyl)piperazine-l-carboxylate (87-1) (168 mg, 262 µιηοΐ , 46. 1 %) LCMS (ES+): m/z= 641 [M + H ]+

Intermediate 88-1. Preparation of 4-(4-(4-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione Scheme 88.

A solution of ter t-butyl 4-(4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butyl)piperazine-l-carboxylate (87-1) (205 mg, 319 µιηοΐ) in MeOH/HCl(3M) (6 mL) was stirred for 4h at rt. The mixture was concentrated to used as is in the next step. LCMS (ES+): m/z= 541 [M + H]+

Intermediate 89-1. Preparation of 6-(4-Chlorophenyl)-8-(l-(3-chloropropyl)-lH-pyrazol-4- yl)-l-methylspiro [benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1'-cyclopropane] Scheme 89

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (59-2) (400 mg, 997 µ οΐ) in DMF (3.0 mL ) was added Cesium carbonate (648 mg, 1.99 mmol) and l-bromo-3- chloropropane (3 13 mg, 1.99 mmol) . The reaction mixture was stirred at room temperature for 2 h . The solution was poured into water (30 mL), then extracted with ethyl acetate (30 mLX3). The combined organic layers were washed with birne, and then dred with Na2S04. The solvent was removed and the residue was purified by prep-TLC(100% ethyl acetate).to affored target compound 6-(4-chlorophenyl)-8-(l-(3-chloropropyl)-lH-pyrazol-4-yl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (89-1) (352 mg, 739 µιηοΐ , 74.1 %). LCMS (ES+): m/z= All [M + H]+

Intermediate 90-1. Preparation of r -Butyl 4-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)piperazine-l-carboxylate Scheme 90. Brought 6-(4-chlorophenyl)-8-( 1-(3 -chloropropyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (89-1) (375 mg, 775umol), t r t-butyl piperazine-l-carboxylate (79-1) (286 mg, 1.54 mmol) Potassium carbonate (212 mg, 1.54 mmol) Potassium iodide (255 mg, 1.54 mmol) up in DMF (5ml ) . Stirred reaction at 80. for 16 hours. Purified by prep TLC (DCM/MeOH=10/l) to afford r t-butyl 4-(3-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)propyl)piperazine-l-carboxylate (90-1) (450 mg, 717 µπιοΐ , 44.3 %) as white solid. LCMS (ES+): m/z= 492 [M + H ]+

Intermediate 91-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(3-(piperazin-l- yl)propyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 91.

Brought ter t-butyl 4-(3 -(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)propyl)piperazine- 1-carboxylate (90-1) (400 mg, ) in 3M Hydrochloric acid (10ml ) . Stirred reaction at r.t. for 4 hours and then purified by prep TLC (DCM/MeOH=10/l)to afford 6-(4-chlorophenyl)-l-methyl-8-(l-(3-(piperazin-l- yl)propyl)- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (91-1) (330 mg, 626 µιηοΐ , 98.5 %) as yellow solid. LCMS (ES+): m/z= 528 [M + H]+

Intermediate 92-1. Preparation of Synthesis of 8-(l-(2-Chloroethyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 92.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (59-2) (240 mg, 598 µ οΐ) in DMF (2.0 mL ) was added l-bromo-2-chloroethane (170 mg, 1.19 mmol) and Cesium carbonate (387 mg, 1.19 mmol). The reaction mixture was stirred at room temperature for 2 h . The solution was poured into water (20 mL) and then extracted with ethyl acetate (20 mLX3). The combined organic layes were washed with brine and then dried over Na2S04.The solvent was removed by reduce pressure and the residue was purified by prep-TLC (100% ethyl acetate to afford white target compound 8-(l-(2-chloroethyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (92-1) (178 mg, 384 µπιοΐ , 64.2 %). Another batch (300 mg) was carried with a same condition and afford product (240 mg). LCMS (ES+): m/z= 463 [M + H]+

Intermediate 93-1. Preparation of r -Butyl 4-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane]-8-yl)-lH- pyrazol-l-yl)ethyl)piperazine-l-carboxylate Scheme 93. To a solution of 8-(l-(2-chloroethyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (92-1) (180 mg, 388 µηιοΐ) in DMF (2.0 mL ) was added t rt-butyl piperazine-l-carboxylate (79-1) (144 mg, 776 µιηοΐ) and Potassium carbonate (107 mg, 776 µιηοΐ) , Potassium iodide (128 mg, 776 µιηοΐ) . The reaction mixture was heated to 80 oC and stirred for 16 h . After cooled to room temperature, the solution was poured into water (20 mL), then extracted with ethyl acetate (20 mLX3). The combined organic layers were washed with brine and dried over Na2S04. The solvent was removed and the residue was purified by prep-TLC (100% ethyl acetate) to afford target compound t rt-butyl 4-(2- (4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethyl)piperazine-l-carboxylate (93-1) (140 mg, 228 µπιοΐ , 59.0 %). Another batch (200 mg) was carried with a same condition and afford target compund (240 mg) as a white solid. LCMS (ES+): m/z= 613 [M + H]+

Intermediate 94-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(2-(piperazin-l- yl)ethyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 94. The reaction was carried by two batches with t rt-butyl 4-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethyl)piperazine-l-carboxylate (93-1) (440 mg, 717 µηιοΐ) (200 mg and 240 mg) in the solution of HCl/MeOH (5 mL, 3 M). The reaction mixture was stirred at room temperature for 16 h . The solvent was removed under reduce pressure and the residue was purified by reverse phase flash (MeCN/H2O/0.01% M NH4C03) to afford target compound 6-(4-chlorophenyl)-l-methyl-8-(l- (2-(piperazin- 1-yl)ethyl)- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (94-1) (95.0 mg, 185 µιηοΐ , 25.8 %) as a yellow solid. LCMS (ES+): m/z= 513 [M + H]+

Intermediate 95-1. Preparation of 8-(l-(3-Azidopropyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 95.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (0.053 g, 0.1322 mmol) up in DMF (1.3 mL) at 0 °C and added potassium 2-methylpropan-2-olate (16.3 mg, 0.1454 mmol) . After 20 minutes at reduced temp added l-bromo-3-chloropropane (14.3 0 .1454 mmol) stirred for 60 minutes at r.t. Saw a bit of remaining SM so added an additional 4 uL of l-bromo-3- chloropropane. After 30 minutes more no SM by lcms. Added sodium azide (17.1 mg, 0.2644 mmol) and heated O/N at 50°C. Cooled reaction and concentrated before purifying by isco 0-10% MEOH/DCM to give 8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (95-1) (47.0 mg, 97.1 umol, 73.5%) as an oil. LCMS (ES+): m/z= 484 [M + H]+ Intermediate 96-1. Preparation of 8-(l-(4-Azidobutyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)- l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 96.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (0.05 g, 0.1247 mmol) , cesium carbonate (60.9 mg, 0.1870 mmol) up in DMF (1.24 m L ) and added l-bromo-4- chlorobutane (17.2 , 0.1496 mmol) . Stirred reaction at r.t. for 4 hours. Added sodium azide (16.2 mg, 0.2494 mmol) and heated reaction to 50°C O/N, when complete concentrated onto celite from toluene and purified by isco (0-15% MeOH/DCM) Isolated 8-(l-(4-azidobutyl)-lH-pyrazol-

4-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (96-1) (53.0 mg, 0.1064 mmol, 85.4 % ) as an oil. LCMS (ES+): m/z= 498 [M + H ]+

Intermediate 97-1. Preparation of 8-(l-(5-Azidopentyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 97.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (0.1 g, 0.2494 mmol) and cesium carbonate (121 mg, 0.3741 mmol) up in DMF (2.49 mL ) and added l-bromo-5- chloropentane (39.3 , 0.2992 mmol). Stirred reaction at r.t. for 5 hours and added sodium azide (32.4 mg, 0.4988 mmol). Heated reaction up to 70°C and stirred for 16 hours. Concentrated from toluene onto celite and purified by isco 0-10% MeOH/DCM to give 8-(l-(5-azidopentyl)-lH- pyrazol-4-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,l'-cyclopropane] ( 110 mg, 0.2148 mmol, 86.6 %) as an oil. LCMS (ES+): m/z= 512 [M + H]+

Intermediate 98-1. Preparation of 8-(l-(6-Azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)- l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 98.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (0.053 g, 0.1322 mmol) up in DMF (1.32 mL ) at 0°C. Added potassium 2-methylpropan-2-olate (16.3 mg, 0.1454 mmol) and let warm to r.t. Added l-bromo-6-chlorohexane (21.6 , 0.1454 mmol) 15 minutes later. Added sodium azide (8.59 mg, 0.1322 mmol) and heated to 50°C O/N. Concentrated from toluene onto celite and purified by isco 0-10% MeOH/DCM to give 8-(l-(6-azidohexyl)-lH-pyrazol-4- yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1 (64.0 mg, 0.122 mmol, 92.0%) as an oil. LCMS (ES+): m/z= 527 [M + H]+

Intermediate 99-1. Preparation of (S)-8-(l-(6-Azidohexyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 99. Brought (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (0.045 g, 0.1 157 mmol) up in DMF (1.0 mL) and cooled to 0°C. Added potassium 2-methylpropan-2-olate (12.9 mg, 0.1 157 mmol) and stirred at reduced temp for 10 minutes and room temp for 20 minutes. Then added l-bromo-6- chlorohexane (18.9 , 0.1272 mmol) dropwise atr.t. Stirred for 1hours at.r.t complete by lcms. Added sodium azide (9.77 mg, 0.1504 mmol) and stirred at r.t. for 3 hours. Very slow progress. Heated to 50 °C O/N over half complete. Added another 5 mg sodium azide and continued heating. After ~ 4 hours more complete concnetrated onto celite and purified by isco 0-10% MeOH/DCM to give (S)-8-(l-(6-azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (99-1) (41.0 mg, 0.07976 mmol, 69.0 %) as an oil. LCMS (ES+): m/z= 515 [M + H]+

Intermediate 100-1. Preparation of (S)-8-(l-(10-Azidodecyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 100.

Brought (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (0.0430 g, 0.1 103 mmol) up in DMF (1.1 mL) and cooled to 0°C. Added potassium 2-methylpropan-2-olate (15.4 mg, 0.1378 mmol) and stirred at reduced temp for 15 minutes before adding decane-l,10-diyl bis(4-methylbenzenesulfonate) (J. Org. Chem. 72(15), 5505; 2007) (79.8 mg, 0.1654 mmol). Continued stirring for 30 minutes at reduced temp before warming to r.t. for 2 hours. No SM as I could tell. Added sodium azide (21.5 mg, 0.3308 mmol) and heated to 55°C for 24 hours. Concentrated from toluene onto celite for solid loading onto isco. Isolated (S)-8-(l-(10-azidodecyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4- dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (100-1) (28.0 mg, 0.0491 1 mmol, 44.5 %) as an oil. LCMS (ES+): m/z= 571 [M + H]+

Intermediate 101-1. Preparation of (S)-8-(l-(4-Azidobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 101.

Brought (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (0.045 g, 0.1 157 mmol) up in DMF (1.0 mL) and cooled to 0°C. Added potassium 2-methylpropan-2-olate (12.9 mg, 0.1 157 mmol) and stirred at reduced temp for 10 minutes and room temp for 20 minutes. Then added l-bromo-4- chlorobutane (14.6 , 0 .1272 mmol) dropwise at r.t. Stirred for ~ 1 hours at.r.t complete by lcms. Added sodium azide (9.77 mg, 0.1504 mmol) and stirred at r.t. for 3 hours. Very slow progress. Heated to 50 °C O/N over half complete. Added another 5 mg sodium azide and continued heating. Concentrated onto celite and purified by isco 0-10% MeOH/DCM to give (S)-8-(l-(4-azidobutyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine (100-1) (44.0 mg, 0.09054 mmol, 78.2 %) as an oil. LCMS (ES+): m/z= 487 [M + H]+

Intermediate 102-1. Preparation of (S)-8-(l-(3-Azidopropyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine Scheme 102. Brought (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (0.047 g, 0.1208 mmol) up in dmf (1.20 mL ) and cooled to 0°C. Added potassium 2-methylpropan-2-olate (14.9 mg, 0.1328 mmol) and let gradually warm to r.t. after 20 minutes added l-bromo-3-chloropropane ( 11.9 , 0.1208 mmol) . After 4 hours added sodium azide ( 11.7 mg, 0.1812 mmol) and heated reaction to 50°C O/N. cooled to r.t. and concnetrated. purified by isco 0-10% MeOH/DCM to give Preparation of (S)-8- (1-(3 -azidopropyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (102-1) (40.6 mg, 0.0975 mmol, 80.7%) as an oil. LCMS (ES+): m/z= 516.4 [M + H]+

Intermediate 103-2. Preparation of 2-(2-(2-(2-Azidoethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate. Scheme 103.

Brought 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethanol (103-1) (0.175 g, 0.7936 mmol) and N,N-dimethylpyridin-4-amine (9.69 mg, 0.07936 mmol) up in DCM (8.0 mL) and cooled to 0°C. Added N-ethyl-N-isopropylpropan-2-amine (206 1.19 mmol) followed by 4- methylbenzene-l-sulfonyl chloride (151 mg, 0.7936 mmol) and gradually let warm to r.t. O/N. Messy LCMS after 1 night in MeOH. Added more TEA and ~ 50 mg more TsCl. Continued stirring, after another night LCMS looks good in CH3CN. Diluted with DCM and washed with H4C1solution followed by water. Dried organic over sodium sulfate and concnetrated. Purified by isco 50%- 100% EA hx to give 2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl 4- methylbenzenesulfonate (103-2) (150 mg, 0.4016 mmol, 50.6 %) as an oil.

Intermediate 104-1. Preparation of (S)-8-(l-(2-(2-(2-(2-Azidoethoxy)ethoxy)ethoxy)ethyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine Scheme 104.

Brought (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(lH-pyrazol-4-yl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (64-1) (0.031 g, 0.07972 mmol) up in DMF (797 µ ) and cooled reaction to 0°C. Added potassium 2-methylpropan-2-olate (8.94 mg, 0.07972 mmol) and let warm to r.t. for 10 minutes before recooling and adding 2-(2-(2-(2- azidoethoxy)ethoxy)ethoxy)ethyl 4-methylbenzenesulfonate (103-2) (29.7 mg, 0.07972 mmol). Then let the reaction stir at r.t. for 12 hours before concentrating onto celite and purifying by isco 0-15% MeOH/DCM to give (S)-8-(l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-lH-pyrazol- 4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (104-1) (25.0 mg, 0.04236 mmol, 53.1 %) as an oil. LCMS (ES+): m/z= 591 [M + H]+

Intermediate 105-1. Preparation of 3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propan-l-amine Scheme 105. Brought 8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (95-1) (0.015 g,

0.03099 mmol) and triphenylphosphine (9.75 mg, 0.03718 mmol) up in THF (309 ΐ ) / water

(30.9 µΙ_, ) and stirred O/N. Concentrated and azeotroped from toluene. Purified by isco 1-20% MeOH/DCM w ammonia to give 3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propan-l -amine (105-1) ( 11.0 mg, 0.024 mmol, 78.0%) as an oil. LCMS (ES+): m/z= 458 [M + H ]+

Intermediate 106-1. Preparation of 6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexan-l-amine Scheme 106.

Brought 8-(l-(6-azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (98-1) (0.02 g,

0.03802 mmol) and triphenylphosphine ( 11.9 mg, 0.04562 mmol) up in THF:water 10:1 (420 uL) and stirred at r.t.. Concentrated from toluene 2x and purified by isco 1-20% MeOH/DCM w NH3 to give 6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexan-l-amine (106-1) (4.0 mg, 0.008 mmol, 21.1%) as an oil.LCMS (ES+): m/z= 500 [M + H]+

Intermediate 107-1. Preparation of (S)-3-(4-(6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)propan-l-amine Scheme 107.

Brought (S)-8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (102-1) (22 mg, 0.04661 mmol) and triphenylphosphine (18.3 mg, 0.06991 mmol) up in THF (466 i ) / water (46.6 i ) and stirred O/N at r.t.. Concentrated reaction and purified by isco 0-20% MeOH/DCM w ammonia to give (S)-3-(4-(6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)- lH-pyrazol-l-yl)propan-l-amine (107-1) (20.0 mg, 0.045 mmol, 96.6%) as an oil. LCMS (ES+): m/z= 447 [M + H]+

Intermediate 108-1. Preparation of (S)-6-(4-(6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)hexan-l-amine Scheme 108.

Added (S)-8-( 1-(6-azidohexyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (99-1) (0.021 g, 0.04085 mmol) and triphenylphosphine (10.7 mg, 0.04085 mmol) to a vial and dissolved in thf (0.4 mL)/water (0.02 mL). Stirred at r.t. for 3 nights complete by lcms. Concentrated reaction onto celite and purified by isco 0-30% MeOH/DCM with ammonia to give 4-((6-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl- 4H-benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)hexyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (108-1) (14.0 mg, 0.029 mmol, 70.2%) as an oil. LCMS (ES+): m/z= 488 [M + H]+

Intermediate 109-1. Preparation of (S)-10-(4-(6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)decan-l-amine Scheme 109.

Brought (S)-8-( 1-(1 0-azidodecyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (100-1) (14 mg, 0.02455 mmol) and triphenylphosphine (9.65 mg, 0.03682 mmol) up in THF (245 uL)/Water (25 uL) and stirred reaction overnight. Concentrated from toluene to dryness. Purified by isco, 1-20% MeOH/DCM w H3 to give (S)-10-(4-(6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)decan-l-amine (109-1) (13.0 mg, 0.02389 mmol, 97.7 %) as an oil. LCMS (ES+): m/z= 544 [M + H]+

Intermediate 110-1. Preparation of (S)-2-(2-(2-(2-(4-(6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l- yl)ethoxy)ethoxy)ethoxy)ethanamine Scheme 110. Brought (S)-8-(l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (104-1) (14 mg, 0.02372 mmol) and triphenylphosphine (9.33 mg, 0.03558 mmol) up in THF (237 ) / water

(23.7 µΙ_, ) and stirred at r.t. for 3 nights. Concentrated reaction from toluene 2x and purified by isco 0-20% MeOH/DCM to give (S)-2-(2-(2-(2-(4-(6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- IH-pyrazol- 1- yl)ethoxy)ethoxy)ethoxy)ethanamine (110-1) (9.00 mg, 0.01595 mmol, 67.6 %) as an oil. LCMS (ES+): m/z= 565 [M + H ]+

Intermediate 111-2. Preparation of 7-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptan-l-amine Scheme 111.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (0.10 g, 0.2494 mmol), cesium carbonate (121 mg, 0.3741 mmol) up in DMF (2.5 mL) and added l-bromo-7- chloroheptane (49.4 , 0.2992 mmol). Stirred reaction atr.t. overnight. Added sodium azide (16.2 mg, 0.2494 mmol) and heated reaction to 50°C O/N, when complete concentrated onto celite from toluene and purified by isco (0-15% MeOH/DCM) Isolated 8-(l-(7-azidoheptyl)-lH-pyrazol-4- yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (111-1) (61.0 mg, 0.1 135 mmol, 45.5 %) as an oil. LCMS (ES+): m/z= 541 [M +

H ]+ Brought 8-(l-(7-azidoheptyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (111-1) (0.06 g, 0.1 110 mmol) and triphenylphosphine (37.8 mg, 0.1443 mmol) up in thf (1.10 mL ) / water ( 111

) and stirred at r.t. overnight (saw full conversion from Azide to Amine by lcms). Concentrated reaction onto celite from toluene to remove water. Purified directly via isco 0-20% MeOH/DCM. Isolated 7-(4-(6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptan-l-amine (111-2) (50.0 mg, 0.09726 mmol, 87.7 % ) as an oil. LCMS (ES+): m/z= 515 [M + H ]+

Intermediate 112-1. Preparation of 4-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butan-l-amine Scheme 112.

Brought 8-(l-(4-azidobutyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (96-1) (220 mg, 441

µ οΐ) and triphenylphosphine (148 mg, 567 µ οΐ) up in Tetrahydrofuran ( 2.5ml) / Water

(0.25ml) and stirred at r.t. overnight. Concentrated reaction onto celite from toluene to remove water. Purified by TLC (10% MeOH/DCM) to afford 4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butan-l -amine (112-1) (157 mg, 332 µιηοΐ , 58.8 % ) as an oil. LCMS (ES+): m/z= 473 [M + H ]+

Intermediate 113-1. Preparation of 8-(l-(9-Bromononyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 113.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (100 mg, 249

µπιοΐ ) in DMF(1.5 mL) was added Cs2C03 (121 mg, 373 µιηοΐ ) and 1,9-dibromononane (85.2 mg, 298 µιηοΐ ) . The mixture was stirred for 4h at 30 °C. Water(30 mL) and EA(30 mL*2) were added. The Combined organic layers was dried over Na2S04 and concentrated, purified with prep- TLC (EA=100%) to give 8-(l-(9-bromononyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (113-1) (39.2 mg,

26. 1 % ) as an oil. LCMS (ES+): m/z= 605 [M + H ]+

Intermediate 114-1. Preparation of 8-(l-(9-Azidononyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 114. To a solution of 8-(l-(9-bromononyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (113-1) (250 mg,

412 µηιοΐ) in DMF(5 mL) was added sodium azide (79.9 mg, 1.23 mmol) . The mixture was stirred overnight at 50 °C. Water (50 mL) and EA(50 mL*2) were added. The Combined organic layers was dried over Na2S04 and concentrated to give 8-(l-(9-azidononyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (114-1) (200 mg, 352 µιηοΐ , 85.4 %) to used to next. LCMS (ES+): m/z= 568 [M + H]+

Intermediate 115-1. Preparation of 9-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)nonan-l-amine Scheme 115.

To a solution of 8-(l-(9-azidononyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (114-1) (200 mg, 352 µ οΐ) in THF(3.5 mL) and Water(0.35 mL) was added triphenylphosphine ( 119 mg, 457

µ οΐ) . The mixture was stirred overnight at rt. The mixture was concentrated and purified with Prep-TLC(DCM:MeOH=5:l) to give 9-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)nonan-l -amine (115-1) (141 mg, 261 µιηοΐ , 74.2 %) as an oil. LCMS (ES+): m/z= 542 [M + H]+

Intermediate 116-1. Preparation of 8-(l-(8-Bromooctyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l Scheme 116. Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (250 mg, 623 µιηοΐ) up in DMF (6 mL ) and added Cs2C03 (407 mg, 1.25 mmol) followed by 1,8-dibromooctane (505 mg, 1.86 mmol) . Stirred reaction at r.t. for 4 hours. (116-1) used directly in the next step. LCMS (ES+): m/z= 593 [M + H]+

Intermediate 117-1. Preparation of 8-(l-(8-Azidooctyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)- l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l heme 117.

To crude 8-(l-(8-bromooctyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (116-1) (354 umol) in DMF (1.0 mL) was added sodium azide (26.9 mg, 415 µιηοΐ) and heated reaction to 50°C O/N. The reaction mixture was allowed to cool to room temperature and was partitioned between ethyl acetate (30 mL) and water (100 mL*3). The organic layer was collected, washed with brine (50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to give a crude product. It was the purified by TLC (MeOH/DCM 1:10) to afford 8-(l-(8-azidooctyl)-lH-pyrazol-4-yl)-6- (4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (117-1) (194 mg, 350 µιηοΐ , 98.9 %) as white solid. LCMS (ES+): m/z= 554 [M + H]+ Intermediate 118-1. Preparation of 8-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)octan-l-amine Scheme 118.

Brought 8-(l-(8-azidooctyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (117-1) (200 mg, 360 µιηοΐ) and triphrnylphosphine (94.4 mg, 360 µηιοΐ) up in Tetrahydrofuran (3.6 mL ) / Water (6.48 mg, 360

µ οΐ) and stirred at r.t. overnight. Concentrated and purified by TLC (10% MeOH/DCM) to afford 8-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)octan-l-amine (118-1) (170 mg, 321 µπιοΐ , 81.3 %) as an oil. LCMS (ES+): m/z= 528 [M + H]+

Intermediate 119-1. Preparation of 5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentan-l-amine Scheme 119. Brought 8-(l-(5-azidopentyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (97-1) (135 mg, 263 µιηοΐ) and triphenylphosphine (68.9 mg, 263 µιηοΐ) up in Tetrahydrofuran (2.5ml ) / Water (0.25ml ) and stirred at r.t. overnight. Concentrated reaction and purified by TLC (10% MeOH/DCM) to afford 5-(4-(6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentan-l-amine (119-1) (125 mg, 257 µπιοΐ , 98.4 % ) as an oil. LCMS (ES+): m/z= 486 [M + H ]+

Intermediate 120-1. Preparation of r -Butyl (2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)ethyl)carbamate 1112-53 Scheme 120.

To a solution of 2-(2-((/er/-butoxycarbonyl)amino)ethoxy)ethyl methanesulfonate (120-1)

(commercial) (240 mg, 847 µ οΐ) in DMF (3 mL ) added cesium carbonate (687 mg, 2.1 1 mmol) at RT,then added 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (373 mg, 931

µ οΐ) and stirred for 18 h at RT.The reaction progress was monitored by TLC and LCMS. After completion of reaction quenched it with ice cold water(lOmL) and extracted with ethyl acetate

(10x3mL). Combined the organic layers and dried over anhydrous sodium sulfate,filtered and distilled. The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0- 10%) to give r t-butyl (2-(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4, l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethoxy)ethyl)carbamate (120-2) (200 mg, 340 µιηοΐ , 40.1 % ) as yellow solid.LCMS (ES+): m/z= 646 [M + H ]+ Intermediate 121-1. Preparation of 2-(2-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)ethan-l-amine Scheme 121.

Brought ter t-butyl (2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethoxy)ethyl)carbamate (120-2) (130 mg, 0.2210 mmol) up in dcm (1.5 mL ) and added 2,2,2- trifluoroacetic acid (350 ul, 0.221 mmol) . Stirred at r.t. for 1 hour and then diluted with toluene and concentrated. Lyophilized from 1:1 ACN/water to give 2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethoxy)ethanamine 2,2,2-trifluoroacetate (121-1) ( 115 mg, 0.1910 mmol, 86.4 %) as an oil.

LCMS (ES+): m/z= 488 [M + H]+

Intermediate 122-1. Preparation of 2-(2-((terf-Butoxycarbonyl)(methyl)amino)ethoxy)ethyl methanesulfonate Scheme 122.

122-1 122-2

To a solution of ter t-butyl (2-(2-hydroxyethoxy)ethyl)(methyl)carbamate (122-1) (200 mg, 912 µπιοΐ) in DCM (2 mL ) added TEA (92.2 mg, 912 µπιοΐ ) under nitrogen atmosphere, at 0°C added MESYL CHLORIDE (312 mg, 2.73 mmol) drop wise,stirred the reaction at RT for 1 h.The reaction progress was monitored by TLC and LCMS.The RM was quenched with water (lOmL) and extracted with DCM (10mLx3) .combined organic layers and dried over anhydrous sodium sulfate,filtered and distilled to obtain crude2-(2 -((t r t- butoxycarbonyl)(methyl)amino)ethoxy)ethyl methanesulfonate (122-2) (320 mg, 1.07 mmol, 118 %) as yellow viscous liquid. LCMS (ES+): m/z= 356 [M + H]+

Intermediate 123-1. Preparation of r -Butyl (2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)ethyl)(methyl)carbamate 1112-57 Scheme 123.

To a solution of 2-(2-((t rt-butoxycarbonyl)(methyl)amino)ethoxy)ethyl methanesulfonate (122-2) (320 mg, 1.07 mmol) added cesium carbonate (1.04 g, 3.21 mmol) at RT,then added 6-(4- chlorophenyl)-l-methyl-8-(lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,1 '-cyclopropane] (59-2) (428 mg, 1.07 mmol) and stirred for 18 h at RT.The reaction progress was monitored by TLC and LCMS.. After completion of reaction quenched it with ice cold water(15mL) and extracted with ethyl acetate (15x3mL). Combined the organic layers and dried over anhydrous sodium sulfate,filtered and distilled.The product was purified by silica gel flash chromatography DCM/MeOH 0-10% to give rt-butyl (2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethoxy)ethyl)(methyl)carbamate (123-1) (320 mg, 531 µιηοΐ , 49.6 %)as yellow solid. LCMS (ES+): m/z= 602 [M + H]+

Intermediate 124-1. Preparation of 2-(2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)-N-methylethan-l-amine Scheme 124. Brought tert-butyl (2-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethoxy)ethyl)(methyl)carbamate (123-1) (130 mg, 0.2159 mmol) up in dcm (1.5 mL ) and added 2,2,2-trifluoroacetic acid (350 uL, 0.2159 mmol) . Stirred at r.t. for 1 hour, complete by lcms. Diluted with toluene and concentrated to an oil. dissolved in ACN/water and lyophilized to give 2-(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethoxy)-N-methylethanamine 2,2,2-trifluoroacetate (124-1) ( 112 mg, 0.1818 mmol, 84.2 %) as an oil. LCMS (ES+): m/z= 503 [M + H]+

Intermediate 125-2. Preparation of 2-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethanol Scheme 125.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (59-2) (500 mg, 1.24 mmol) in DMF (10 mL ), Cesium carbonate (1.20 g, 3.72 mmol) was added and stirred for 10 minte followed by addition of 2-bromoethanol (232 mg, 1.86 mmol) and stirred the reaction mixture at room temperature for 16 h, After showed TLC completion, Quenched the reaction mixture inti ice water (20 mL) and extracted with EtOAc (3 x 20 mL). the resulting was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified via Combiflash by using 5% MeOH in DCM as eluent to give 2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethanol (125-2) (310 mg, 696 µιηοΐ , 56.2 %) as colorless liquid. LCMS (ES+): m/z= 445 [M + H]+

Intermediate 126-1. Preparation of 2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethyl methanesulfonate Scheme 126.

To a solution of 2-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethanol (125-2) (310 mg, 696 µ οΐ ) in

DCM (6.0 mL ) , Triethylamine (484 µΐ 3.48 mmol) was added. Then the reaction mixture was cooled to 0 °C and Methanesulfonyl chloride (107 µΐ 1.39 mmol) was added drop wise. The reaction mixture was stirred at rt for lh. After the completion of starting material by TLC, reaction mixture was diluted by DCM (~25ml) and washed with water(~20ml). Combined organic layer was washed with brine solution and dried over anhydrous sodium sulfate and concentrated under reduce pressure. The product was purified by isco 0-10%) DCM/MeOH to give 2-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)ethyl methanesulfonate (126-1) (212 mg, 406 µπιοΐ , 58.2 %) as a white solid.

LCMS (ES+): m/z= 523 [M + H]+ Intermediate 127-2. Preparation of r -Butyl (2-(4-(2-(4-(6-(4-chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethyl)piperazin-l-yl)ethyl)carbamate Scheme 127.

To a solution of 2-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethyl methanesulfonate (126-1) (230 mg, 439 µηιοΐ) , t r t-butyl (2-(piperazin-l-yl)ethyl)carbamate (127-1) (150 mg, 658 µηιοΐ) in

Acetonitrile (5 mL ) , DIEA (227 µΐ 1.31 mmol) was added at rt. The reaction was heated to 85 °C for 16h. The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was quenched with water(15.0 mL) and extracted with ethyl acetate(25x3 mL).The organic layers were dried over sodium sulfate,filtered and concentrated to afford crude. The crude product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-10% ) to give r t-butyl (2-(4-(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)ethyl)piperazin- 1-yl)ethyl)carbamate (127-2) (185 mg, 281 µιηοΐ , 64.2 %) as light brown solid. LCMS (ES+): m/z= 656 [M + H]+

Intermediate 128-1. Preparation of Synthesis of 2-(4-(2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethyl)piperazin-l-yl)ethan-l-amine Scheme 128. To a solution of rt-butyl (2-(4-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethyl)piperazin-l-yl)ethyl)carbamate (127-2) (185 mg, 281 µιηοΐ) in DCM (3 mL ) , 25% TFA in DCM (3 mL ) was added at 0 °C. The reaction mixture was stirred at rt for 2h. The reaction progress was monitored by TLC and LCMS analysis. The solvent was evaporated and triturated with diethylether to afford 2-(4-(2-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)ethyl)piperazin-l-yl)ethanamine with TFA salt (128-1) (180 mg, 268 µιηοΐ , 95.7 %) as light brown solid. LCMS (ES+): m/z= 556 [M + H]+

Intermediate 129-2. Preparation of 8-(l-(6-((ieri-Butyldimethylsilyl)oxy)hexyl)-lH-pyrazol- 4-yl)-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 129. Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (59-2) (150mg, 0.3741 mmol) and (182 mg, 0.561 1 mmol) up in DMF (3.74 mL ) at r.t. Added ((6-bromohexyl)oxy)(fer t- butyl)dimethylsilane (129-1) (125 L, 0.4489 mmol) and stirred at r.t. for 16 hours. Concentrated onto celite and purified by isco 0-15% MeOH/DCM to give 8-(l-(6-((tert- butyldimethylsilyl)oxy)hexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (129-2) (150.0 mg, 0.244 mmol, 65.2%) as an oil. LCMS (ES+): m/z= 615 [M + H]+

Intermediate 130-1. Preparation of 6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexan-l-ol Scheme 130.

Brought 8-(l-(6-((ter t-butyldimethylsilyl)oxy)hexyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (129-2) (150 mg, 0.2437 mmol) up in THF (2.43 mL ) and added 1M tetrabutylammonium fluoride in THF (316 , 0.3168 mmol). Stirred reaction at r.t. O/N. Complete by lcms. Concentrated and purified by isco 0-15% MeOH/DCM to give 6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexan-l-ol (130-1) (100 mg, 0.1995 mmol, 81.9 %) as an oil. LCMS (ES+): m/z= 501 [M + H]+

Intermediate 131-1. Preparation of 6-(4-Chlorophenyl)-8-(l-(6-iodohexyl)-lH-pyrazol-4-yl)- l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 131. Brought lH-imidazole (330 mg, 4.86 mmol) and triphenylphosphine (1.27 g, 4.86 mmol) up in DCM (15.0 mL) and cooled the solution to 0°C. Added diiodine (1.1 g, 4.37 mmol) and stirred for 15 minutes at reduced temperature. Subsequently added a solution of 5-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)pentan-l-ol (130-1) (1.22 g, 2.43 mmol) in DCM (5mL ) and continued stirring at reduced temp for 1 hour. Diluted reaction with EA and quenched with 1:1 sodium thiosulfate solution/sodium bicarbonate solution at reduced temperature stirred for 5 minutes. Partitioned and washed org layer w EtOAc. Then washed combined organics with sodium thiosulfate solution followed by brine. Dried organic layer over sodium sulfate and concentrated. Purified by isco (0-10% MeOH/DCM) to give 6-(4-chlorophenyl)-8-(l-(5-iodopentyl)-lH- pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (131-1) (1.37 g, 2.25 mmol, 92.0 %) as an amorphous solid. LCMS (ES+): m/z= 6 11 [M + H]+

Intermediate 132-1. Preparation of 5-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentan-l-ol Scheme 132. Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (59-2) (0.35 g, 0.8706 mmol) and cesium carbonate (423 mg, 1.30 mmol) up in DMF (3.48 mL ) and added 5- bromopentan-l-ol (, 1.04 mmol) . Stirred at r.t. O/N. Diluted reaction with EtOAc and washed with water and 2 x 5% LiCl solution. Dried organic layer over sodium sulfate and concentrated. Purified by Isco 0-15% MeOH/DCM. Isolated 5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentan-l-ol (132-1) (250 mg, 0.5133 mmol, 59.1 % ) as an oil. LCMS (ES+): m/z= 488 [M +

H ]+

Intermediate 133-1. Preparation of 6-(4-Chlorophenyl)-8-(l-(5-iodopentyl)-lH-pyrazol-4- yl)-l-methylspiro [benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 133.

Brought lH-imidazole (69.4 mg, 1.02 mmol) and triphenylphosphine (267 mg, 1.02 mmol) up in DCM (2.56 mL ) and cooled the solution to 0°C. Added diiodine (258 mg, 1.02 mmol) and stirred for 15 minutes at reduced temperature. Subsequently added a solution of 5-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)pentan-l-ol (132-1) (250 mg, 0.5133 mmol) in DCM (2.56 mL ) and continued stirring at reduced temp for 1 hour. Diluted reaction with EA and quenched with 1:1 sodium thiosulfate solution/sodium bicarbonate solution at reduced temperature stirred for 5 minutes. Partitioned and washed org layer w EtOAc. Then washed combined organics with sodium thiosulfate solution followed by brine. Dried organic layer over sodium sulfate and concentrated. Purified by isco (0-15% MeOH/DCM) to give 6-(4-chlorophenyl)-8-(l-(5-iodopentyl)-lH- pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (133-1) (250 mg, 0.4188 mmol, 81.6 % ) as an amorphous solid. Intermediate 134-1. Preparation of 3-(4-(6-(4-Chloroph methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)propan-l-ol Scheme 134.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] 59-2) (500 mg, 1.24 mmol) in DMF (10 mL ) at R T under nitrogen atmosphere added, cesium carbonate (1.61 g, 4.96 mmol) followed by addition of 3-bromopropan-l-ol (517 mg, 3.72 mmol) ,Then stirred the reaction mixture at R T for 16h. After consumption of SM quenched the reaction with ice cold water (30mL) and extracted with ethyl acetate (30mLx3). Combined the organic layers and dried over anhydrous sodium sulfate, filtered and concentrated. The product was purified by silica gel flash chromatography (DCM/MeOH 0-10%) to give3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propan-l-ol (134-1) (165 mg, 359 µιηοΐ , 28.9 % ) as yellow semisolid. LC/MS (ES+): m/z 459 [M + H ]+ Intermediate 135-1. Preparation 3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)propyl methanesulfonate Scheme 135. To a solution of 3-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)propan-l-ol (134-1) (165 mg, 359

µηιοΐ) in DCM (2 mL ) at 0°C under nitrogen atmosphere, added TEA (217 mg, 2.15 mmol) followed by addition of MESYL CHLORIDE (102 mg, 897 µιηοΐ) ,Then stirred the reaction mixture at R T for lh.The reaction progress was monitored by TLC and LCMS. After consumption of SM reaction mixture was quenched with water (lOmL) and extracted with DCM (10x3mL).

Combined organic layers and dried over anhydrous sodium sulfate,filtered and distilled under reduced pressure. The product was purified by silica gel flash chromatography (4 g Isco gold, DCM/MeOH 0-10%) to give 3-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)propyl methanesulfonate (135-1) (130 mg, 242 µιηοΐ , 67.7 % ) as yellow crystalline solid. LC/MS (ES+): m/z 537 [M + H ]+

Intermediate 136-1. Preparation of 6-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexanal Scheme 136.

Brought 6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4 ,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexan-l-ol (130-1) (875 mg, 1.74 mmol) up in DCM (17.4 mL ) and l,l-bis(acetyloxy)-3-oxo-3H-lX ,2-benziodaoxol-l-yl acetate (882 mg, 2.08 mmol) as a 0.3 M DCM solution. Stirred at r.t. for 3 hours before quenching with iPrOH (~ 1 mL) after 5 minutes partitioned reaction between DCM and sodium thiosulfate solution. Dried over sodium sulfate and concentrated to an oil. Purified by isco 0-10% MeOH/DCM to give 6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexanal (136-1) (800 mg, 1.60 mmol, 92. 1 %) as an oil. LCMS (ES+): m/z= 499 [M + H]+

Intermediate 137-1. Preparation of 5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentanal Scheme 137.

Brought 5-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentan-l-ol (132-1) (545 mg, 1.1 1 mmol) up in DCM (17.0 mL ) and l,l-bis(acetyloxy)-3-oxo-3H-lX ,2-benziodaoxol-l-yl acetate (705.5 mg, 1.66 mmol) as a 0.3 M DCM solution. Stirred at r.t. for 3 hours before quenching with iPrOH (~ 1 mL) after 5 minutes partitioned reaction between DCM and sodium thiosulfate solution. Dried over sodium sulfate and concentrated to an oil. Purified by isco 0-10% MeOH/DCM to give 5-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentanal (137-1) (485 mg, 1.00 mmol, 90. 1 %) as an oil. LCMS (ES+): m/z= 484 [M + H]+

Intermediate 138-2. Preparation of (l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methanol Scheme 138. Brought 8-(l-(6-azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane (98-1) (789 mg, 1.5 mmol), prop-2-yn-l-ol (131 , 2.25 mmol) and N-ethyl-N-isopropylpropan-2-amine (521 , 3.00 mmol) in thf (15.0 mL). Added copper(I) iodide (142 mg, 0.75 mmol) and stirred at r.t. for

12 hours before concentrating to an oil and purifying by isco 0-10% MeOH/DCM to give (l-(6- (4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (138-2) (480 mg, 0.8246 mmol, 54.9 %) as an oil. LCMS (ES+): m/z= 583 [M + H ]+

Intermediate 139-1. Preparation of l-(5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)-lH-l,2,3-triazole-4-carbaldehyde Scheme 139.

Brought ( 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methanol (138-2) (0.48 g, 0.825 mmol) up in DCM (8.3 mL) and added DMP (3.29 mL, 0.990 mmol) as a 0.3M solution in DCM. Stirred at r.t. for 2 hours before quenching with iPrOH (~ 1 mL) for 5 minutes and partitioning between DCM and sodium thiosulfate solution. Dried organic layer over sodium sulfate and concentrated. Purified by isco 0-10% MeOH/DCM to give l-(6-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4-carbaldehyde (139-1) (450 mg, 0.7757 mmol, 94. 1 %) as an oil. LCMS (ES+): m/z= 582 [M + H]+

Intermediate 140-2. Preparation of r -Butyl (2-(l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6- dihydropyridine-3-carboxamido)ethyl)carbamate Scheme 140.

Brought t rt-butyl (2-aminoethyl)carbamate (140-1) (176 mg, 1.10 mmol) , l-(2,6- dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxylic acid (48-3) (0.185 g, 0.7393 mmol) and N-ethyl-N-isopropylpropan-2-amine (513 , 2.95 mmol) up in DMF (3.69 mL ) and cooled to 0°C. Added 2,4,6-tripropyl-l,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (698 mg, 1.10 mmol) (50% in Me-THF) and let gradually warm to r.t. After stirring for 45 minutes partitioned between EA and H4C1 solution. Washed org with 2x NH4C1 and 2x 5% LiCl solution. Dried organic layer over sodium sulfate and concentrated to an oil. Purified by isco 0-15% MeOH/DCM to give tert-butyl (2-(l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxamido)ethyl)carbamate (140-2) (230 mg, 0.5861 mmol, 79.3 %) as an oil. LCMS (ES+): m/z= 415 [M + Na]+

Intermediate 141-1. Preparation of N-(2-aminoethyl)-l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6- dihydropyridine-3-carboxamide 2,2,2-trifluoroacetate Scheme 141. Brought tert-butyl (2-(l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxamido)ethyl)carbamate (0.23 g, 0.5861 mmol) up in DCM (4 mL) and added trifluoroacetic acid ( 1 mL, 12.2 mmol). Stirred the reaction at r.t. for 2 hours and concentrated from toluene 2x. Recovered N-(2-aminoethyl)-l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxamide 2,2,2-trifluoroacetate (141-1) (225 mg, 0.5537 mmol, 94.5 %) crude as an oil. LCMS (ES+): m/z= 293 [M + Na]+

Intermediate 142-1. Preparation of 8-(l -(7-Chloroheptyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 142.

Brought 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (59-2) (0.62 g, 1.54 mmol) and cesium carbonate (752 mg, 2.31 mmol) up in dmf (7.70 mL ) and added l-bromo-7- chloroheptane (328 µΐ 2.00 mmol). Stirred reaction at r.t. O/N. Concentrated reaction from toluene and purified directly by isco 0-10% MeOH/DCM to give 8-(l-(7-chloroheptyl)-lH- pyrazol-4-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,l'-cyclopropane] (142-1) (530 mg, 0.9934 mmol, 64.5 %) as an oil. LCMS (ES+): m/z= 533 [M + H]+ Intermediate 143-1. Preparation of 6-(4-Chlorophenyl)-8-(l -(7-iodoheptyl)-lH-pyrazol-4- yl)-l-methylspiro [benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 143.

Brought 8-(l-(7-chloroheptyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (142-1) (530 mg, 0.9934 mmol) up in DMF (4.96 mL ) and added potassium iodide (1.64 g, 9.93 mmol) . Heated reaction o/n at 50°C. Reaction was very messy. Concentrated down from toluene and purified directly by isco 0-10% MeOH/DCM. Isolated 6-(4-chlorophenyl)-8-(l-(7-iodoheptyl)-lH- pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (143-1) (120 mg, 0.1920 mmol, 19.3 % ) as an oil. LCMS (ES+): m/z= 625 [M + H ]+

Intermediate 144-1. Preparation of 8-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct -7-yn-l- ol Scheme 144.

6-(4-Chlorophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (58-1) (.150 g, 361 µηιοΐ) cesium carbonate (305 mg, 938 µιηοΐ) were dissolved in ACN (2.40 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen oct-7-yn-l-ol (89.5 µ , 0.6284 mmol) was added followed by dicyclohexyl(2',4',6'- triisopropyl-[l,l'-biphenyl]-2-yl)phosphine (17.2 mg, 36.1 µ οΐ) and XPhos-Pd-G3 (30.5 mg, 36. 1 µ οΐ) . The reaction tube was evacuated and backfilled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered through celite and concentrated onto additional celite. Purified by isco 0-5% MeOH/DCM to give 8-(6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (144-1) (40.0 mg, 0.08714 mmol, 18.0 %) as an oil after concentration. LCMS (ES+): m/z= 459 [M + H]+

Intermediate 145-1. Preparation of 8-(8-Azidooct-l-yn-l-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 145.

Brought 8-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l (144-1) (40 mg, 0.08714 mmol) up in CH3CN (871 µΐ ) and added N-ethyl- N-isopropylpropan-2-amine (36.3 µ , 0.2091 mmol) followed by methanesulfonyl chloride (8.37 µ , 0.1089 mmol) at 0°C. SM crashed out of solution so warmed to r.t. Stirred at r.t. O/N. Added

5 uL more MsCl and stirred for 4 hours more. Then added sodium azide ( 11.3 mg, 0 .1742 mmol) and heated to 50°C O/N. Added ~ 6 mg more azide and continued stirring until afternoon. Concentrated and dissolved in ~ 60 mL EA and washed with water x2 and then brine. Dried org over sodium sulfate and concentrated organic layer. Purified by isco 0-10% MeOH/DCM to give 8-(8-azidooct- 1-yn- 1-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropane] (145-1) (19.0 mg, 0.039 mmol, 45.1%) as an oil. LCMS

(ES+): m/z= 484 [M + H ]+

Intermediate 146-2. Preparation of 7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl methanesulfonate Scheme 146.

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (59-2) (1.0 g, 2.49 mmol) in DMF (10.0 mL ) , Cesium carbonate (2.43 g, 7.47 mmol) was added followed by the addition of 7-bromoheptan-l-ol (971 mg, 4.98 mmol) at 0 °C. The reaction mixture was stirred at rt for

24h.The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was quenched with ice water (15.0 mL) and extracted with ethyl acetate(30.0 mLx3). The organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated to afford 7-(4-

(6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptan-l-ol (850 mg, 1.65 mmol, 66.4 % ) as brown solid.

LCMS (ES+): m/z= 515 [M + H ]+

To a solution of 7-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4 ,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptan-l-ol (146-1) (200 mg, 388

µ οΐ ) in DCM (4 mL ), Triethylamine (322 µΐ 2.32 mmol) was added followed by the addition of Methanesulfonyl chloride (89.1 µΐ 1.16 mmol) at 0 °C. The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was diluted with DCM(25.0 mL) and washed with waster(15.0 mLx3). The organic layers were dried over sodium sulfate,filtered and concentrated to afford 7-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptyl methanesulfonate (146-2) (210 mg, 354 µιηοΐ , 91.3 %) as brown solid. LCMS (ES+): m/z= 593 [M + H]+

Intermediate 147-1. Preparation of 6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl methanesulfonate Scheme 147.

To a solution of 6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexan-l-ol (130-1) (170 mg, 339 µιηοΐ) in DCM (4 mL ) added TEA (205 mg, 2.03 mmol) under nitrogen atmosphere, at 0°C added TEA (205 mg, 2.03 mmol) drop wise,stirred the reaction at RT for 1 h.The reaction progress was monitored by TLC and LCMS.The reaction was quenched with water(12mL) and extracted with DCM (12mLx3) .combined organic layers and dried over anhydrous sodium sulfate. The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-10%) to give 6- (4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl methanesulfonate (147-1) (140 mg, 241 µπιοΐ, 71.4 %)as yellow crystalline solid. LCMS (ES+): m/z= 579 [M + H]+ Intermediate 148-1. Preparation of 6-((4-(l-Methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)amino)hexan-l-ol Scheme 148.

Brought 6-(4-chlorophenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol -4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (209-2) (150mg, 0.3615 mmol) ,6- aminohexan-l-ol (148-1) (50.8 mg, 0.4338 mmol) ,BrettPhos (19.4 mg, 0.03615 mmol) and G3 BrettPhos Precatalyst (28.8 mg, 0.03615 mmol) up in dioxane (1.6 mL) and added lithium bis(trimethylsilyl)amide ( 1.22 mL, 1.22 mmol) (1M in THF). Purged vial with Ar and then heated to 90°C overnight. In the morning concentrated on celite from toluene. Purifed by isco 0-15 MeOH/DCM to give 6-((4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (148-2) (44.0 mg, 0.08877 mmol, 32.1 %) as an oil. LCMS (ES+): m/z= 495 [M + H]+

Intermediate 149-1. Preparation of N-(6-Azidohexyl)-4-(l-methyl-8-(l-methyl-lH-pyrazol-

4-yl)spiro[benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4, 1'-cyclopropan] -6-yl)aniline Scheme 149. Brought 6-((4-(l -methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l (148-2) (44 mg, 0.08877 mmol) and N-ethyl-N-isopropylpropan-2-amine (30.8 uL, 0 .1775 mmol) up in DCM (443 µ ) and cooled to 0°C. Added methanesulfonyl chloride (7.50 , 0.09764 mmol) and continued at reduced temp for 45 minutes. Complete by lcms.

Concentrated off DCM and redissolved crude material in DMF (443 µΙ_, ) and added sodium azide (9.23 mg, 0.1420 mmol) and heated the vial for 16 hours at 45°C. Concentrated from toluene onto celite and purified by isco 0-10% MeOH/DCM to give N-(6-azidohexyl)-4-(l-methyl-8-(l- methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (149-1) (35.0 mg, 0.06722 mmol, 75.7 %) as an oil. LCMS (ES+): m/z= 521 [M + H]+

Intermediate 150-2. Preparation of 4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)but-3-yn-l- ol heme 150. 8-Bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (58- 1) (.350 g, 0.8436 mmol), but-3-yn-l-ol (150-1) (0.095 mL, 1.26 mmol) and triethylamine (5 mL,

35.8 mmol) were taken up in ACN (8 mL ) and argon was bubbled through the solution for 15 min. bis(triphenylphosphinepalladium) acetate (63.1 mg, 0.08436 mmol) was added, the atmosphere purged with argon and the vial sealed. The reaction was heated to 70 °C for 45 min, diluted with EtOAc, filtered through a plug of celite and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 4-(6-(4-chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)but-3-yn-l-ol (150-2) (330 mg, 97 %) as a white solid. LC/MS (ES+): m/z 403.2 [M + H]+

Intermediate 151-2. Preparation of 8-(4-Azidobut-l -yn-l -yl)-6-(4-chloroph methylspiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 151.

To a mixture of 4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l (150-2) (339 mg, 0.843 mmol), Zn(N 3)2/bis-pyridine complex (151-1) (516 mg, 1.68 mmol), triphenylphosphine (440 mg, 1.68 mmol), and imidazole (229 mg, 3.37 mmol) in DCM (3.83) was added diisopropyl azodicarboxylate (0.330 mL, 1.68 mmol) dropwise at room temperature. The mixture was stirred for 16 h . and concentrated. The residue was purified by column chromatography (silica, 0-15 % MeOH in DCM to give 8-(4-azidobut-l-yn-l-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1 diazepine-4, 1'- cyclopropane (151-2) (0.299 g, 83 %) as a white solid. LC/MS (ES+): m/z 428.3 [M + H]+ Intermediate 152-2. Preparation of 10-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)dec-9-yn-l- ol Scheme 152.

8-Bromo-6-(4-chlorophenyl)- 1-methylspiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,l'-cyclopropane] (58-1) (.350 g, .8463 mmol) dec-9-yn-l-ol (152-1) (0.222 mL, 35.8 mmol) and triethylamine (5 mL, 35.8 mmol) were taken up in ACN (8 mL ) and argon was bubbled through the solution for 15 min. bis(triphenylphosphinepalladium) acetate (63.1 mg, 0.08436 mmol) was added, the atmosphere purged with argon and the vial sealed. The reaction was heated to 70 °C for 2 hours, diluted with EtOAc, filtered and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 10-(6-(4-chlorophenyl)-l- methylspiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)dec-9-yn- 1-ol (152-2) (330 mg, 97 %) as a white solid. LC/MS (ES+): m/z 488.7 [M + H]+

Intermediate 153-1. Preparation of 8-(10-Azidodec-l -yn-l -yl)-6-(4-chloroph methylspiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 153. To a mixture of 10-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l cyclopropane] (152-2) (.3 g, 0.6159 mmol) , Zn(N3)2/bis-pyridine complex (151-1) (189 mg, .6159), triphenylphosphine (322 mg, 1.23 mmol) and lH-imidazole (167 mg, 2.46 mmol) in DCM (3 ml)) was added (E)-diisopropyl diazene-l,2-dicarboxylate (241 , 1.23 mmol) dropwise. The mixture was stirred for 16 hours at room temperature and concentrated. The residue was purified by column chromatography (0-15 % MeOH in DCM) to give 8-( 10-azidodec- 1-yn- 1-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l cyclopropane] (153-1) (150 mg, 47.6 %) as a white solid. LC/MS (ES+): m/z 512.3 [M + H]+

Intermediate 154-2. Preparation of tert - y 4-(3-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H- pyrazol-l-yl)propoxy)propyl)piperazine-l-carboxylate heme 154.

Sodium hydride (12.5 mg, 0.3141 mmol) was added to a solution of t rt-butyl 4-(3- hydroxypropyl)piperazine-l-carboxylate (154-1) (76.7 mg, 0.3141 mmol) in DMF (2.09 mL ) at 0 °C. The bath was removed and the reaction stirred for 30 min. 6-(4-chlorophenyl)-8-(l-(3- chloropropyl)- lH-pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (89-1) (0.1 g, 0.2094 mmol) was added, the reaction stirred for 16 hours and concentrated. The product was purified by column chromatography (silica, 0-15% MeOH in DCM) to give tert-butyl 4-(3-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propoxy)propyl)piperazine-l-carboxylate (154-2) (22.0 mg, 15.3 %) LC/MS (ES+): m/z 685.3 [M + H]+ Intermediate 155-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(3-(3-(piperazin-l- yl)propoxy)propyl)-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 155.

rt-Butyl 4-(3 -(3 -(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)propoxy)propyl)piperazine- 1- carboxylate (154-2) (0.016 g, 0.02334) was stirred in a mixture of TFA (0.07 mL, 0.9120 mmol) and DCM (0.233 mL) for 16 hours and concentrated to give 6-(4-chlorophenyl)-l-methyl-8-(l-(3- (3-(piperazin-l-yl)propoxy)propyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l '-cyclopropane] isolated as the TFA salt (155-1) (10.0 mg, 61.3 %) as a brown gum. LC/MS (ES+): m/z 585.4 [M + H]+

Intermediate 156-2. Preparation of r -butyl 4-(2-(4-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)butoxy)ethyl)piperazine-l-carboxylate Scheme 156. Sodium hydride (12.5 mg, 0.3141 mmol) was added to a solution of t rt-butyl 4-(2- hydroxyethyl)piperazine-l-carboxylate (156-1) (70.2 mg, 0.3051 mmol) in DMF (2.03 mL ) at 0 °C. The bath was removed and the reaction stirred for 30 min. 6-(4-chlorophenyl)-8-(l-(3- chlorobutyll)- lH-pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (86-1) (.1 g, 0.2034 mmol) was added, the reaction stirred for 16 hours and concentrated. The product was purified by column chromatography (silica, 0-15% MeOH in DCM) to give tert-butyl 4-(2-(4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butoxy)ethyl)piperazine-l-carboxylate (156-2) (29.0 mg, 20.8 %)LC/MS (ES+): m/z 685.6 [M + H]+ Intermediate 157-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-(4-(2-(piperazin-l- yl)ethoxy)butyl)-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 157.

rt-Butyl 4-(2-(4-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)butoxy)ethyl)piperazine- 1-carboxylate (156-2) (0.029 g, 0.04231) was stirred in a mixture of TFA (0.126 mL, 1.64 mmol) and DCM ( 0.423 mL) for 3 hours and concentrated to give 6-(4-chlorophenyl)-l-methyl-8-(l-(4-(2- (piperazin-l-yl)ethoxy)butyl)-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (157-1) (25.0 mg, 84.7 %) LC/MS (ES+): m/z 585.6 [M + H]+ Intermediate 158-1. Preparation of 8-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct -7-yn-l- ol Scheme 158.

8-Bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,1 -cyclopropane] (58-1) (.25 g, 0.6043 mmol) and oct-7-yn-l-ol (158-1) (0.095 mL, .90645 mmol) and triethylamine (5 mL, 35.8 mmol were taken up in ACN (8 mL ) and argon was bubbled through the solution for 15 min. bis(triphenylphosphinepalladium) acetate (63.1 mg, 0.08436 mmol) was added, the atmosphere purged with argon and the vial sealed. The reaction was heated to 70 °C for 4 hours. After cooling the reaction was diluted with EtOAc, filtered and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 8-(6- (4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]- 8-yl)oct-7-yn-l-ol (158-2) (201 mg, 72.5 %)as a white solid. LC/MS (ES+): m/z 460.6 [M + H]+

Intermediate 159-1. Preparation of 8-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l ,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct -7-yn-l- yl methanesulfonate Scheme 159. Methanesulfonyl chloride (89.8 mg, 0.7843 mmol) was added to solution of 8-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)oct-7-yn-l-ol (158-2) (.3 g, 0.6536 mmol) and triethylamine (109 µ Τ_, 0.7843 mmol). The solution was stirred for six hours and concentrated to give 8-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)oct-7-yn- 1-yl methanesulfonate (159-1) (322 mg, 91.7 %) as a clear oil. LC/MS (ES+): m/z 538.7 [M + H]+

Intermediate 160-1. Preparation of r -Butyl 4-(8-(6-(4-chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct -7-yn-l- yl)piperazine-l-carboxylate Scheme 160.

8-(6-(4-Chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)oct-7-yn-l-yl methanesulfonate (159-1) (0.351 g, 0.6516 mmol) and t rt-butyl piperazine-l-carboxylate (79-1) (182 mg, 0.9774 mmol) were taken up in DMF (3.25 mL ) . N- ethyl-N-isopropylpropan-2-amine (169 , 0.9774 mmol) was added and the reaction heated to 60 °C for 16hrs, concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give rt-butyl 4-(8-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)oct-7-yn- 1- yl)piperazine-l-carboxylate (160-1) (285 mg, 69.8 %) LC/MS (ES+): m/z 628.5 [M + H]+

Intermediate 161-1. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(8-(piperazin-l-yl)oct-l- yn-l-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 161. t r t-Butyl 4-(8-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct-7-yn-l-yl)piperazine-l-carboxylate (160-1) (0.06 g, 0.09566 mmol) was stirred in a mixture of TFA (0.145 mL, 1.91 mmol) and DCM ( 1 mL) for 2 hours and concentrated to give 6-(4-chlorophenyl)-l-methyl-8-(8-(piperazin-l-yl)oct-l-yn-l- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (161-1) (60.0 mg, 0.09358 mmol, 97.8 %) LC/MS (ES+): m/z 527.7 [M + H]+

Intermediate 162-2. Preparation of 6-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)hex-5-yn-l- ol Scheme 162.

8-Bromo-6-(4-chlorophenyl)- 1-methylspiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,l'-cyclopropane] (58-1) (.380 g, .9185 mmol) hex-5-yn-l-ol (162-1) (0.155mL, 1.37 mmol) and triethylamine (5 mL, 35.8 mmol) were taken up in ACN (8 mL ) and argon was bubbled through the solution for 15 min. bis(triphenylphosphinepalladium) acetate (68.6 mg, 0.09185 mmol) was added, the atmosphere purged with argon and the vial sealed. The reaction was heated to 70 °C for 7 hours, diluted with EtOAc, filtered and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,lcyclopropane] (162-2) (322 mg, 81.5 %) as an off white solid. LC/MS (ES+): m/z 432.6 [M + H]+

Intermediate 163-1. Preparation of 8-(6-Azidohex-l -yn-l -yl)-6-(4-chloroph methylspiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 163.

To a mixture of 6-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)hex-5-yn-l-ol (162-2) (.438 g, 1.01 mmol) , Zn(N3)2/bis- pyridine complex (151-1) (621 mg, .2.02 mmol), triphenylphosphine (529 mg, 2.02 mmol) and lH-imidazole (275 mg, 4.04 mmol) in DCM (4 ml)) was added (E)-diisopropyl diazene-1,2- dicarboxylate 397 , 2.02 mmol) dropwise. The mixture was stirred for 16 hours at room temperature and concentrated. The residue was purified by column chromatography (0-15 % MeOH in DCM) to give 8-(6-azidohex-l-yn-l-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (163-1) (150 mg, 47.6 %) as a white solid. LC/MS (ES+): m/z 457.5 [M + H]+

Intermediate 164-1. Preparation of 6-(4-(l-Methyl-8-(l-methyl-l H-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)hex-5- yn-l-ol Scheme 164. 6-(4-Chlorophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (209-2) (0.150 g, 361 µηιοΐ) and cesium carbonate (305 mg, 938 µηιοΐ ) were taken up in ACN (2.40 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen. Hex-5-yn-l-ol (162-1) (52.2 µ , 469 µηιοΐ) was added followed by dicyclohexyl(2',4',6'- triisopropyl-[l,l'-biphenyl]-2-yl)phosphine (17.2 mg, 36.1 µηιοΐ) and XPhos-Pd-G3 (30.5 mg, 36.1 µηιοΐ) . The reaction tube was evacuated and back-filled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered through celite and concentrate. Purification of the residue by column chromatography (silica, 0-10% MeOH in DCM) to give 6-(4-(l-methyl-8-(l-methyl-lH- pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)hex-5-yn-l-ol (164-1) (96.0 mg, 55.8 %) as a clear oil. LC/MS (ES+): m/z 477.5 [M + H]+

Intermediate 165-1. Preparation of 6-(4-(l -Methyl-8-( l -methyl- l H -pyrazol-4-yl)-4H - benzo [ ] [l ,2,4]triazolo[4,3-a ] [l ,4]diazepin-6-yl)phenyl)hex-5-yn - l -yl methanesulfonate Scheme 165 6-(4-(l-Methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)hex-5-yn-l-ol (164-1) (.081 g, 0.1699 mmol) was dissolved in DCM (849 µΙ_, ) triethylamine (47.2 µ , 0.3398 mmol) was added and the solution cooled to 0°C. Methanesulfonyl chloride (15.7 µ , 0.2038 mmol) was added, the reaction stirred for 30 min and concentrated to give 6-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4-yl)-4H- benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-6-yl)phenyl)hex-5-yn- 1-yl methanesulfonate (165-1) (89.8 mg, 100 %) as a clear oil.LC/MS (ES+): m/z 529.3 [M + H]+

Intermediate 166-1. Preparation 6-(4-(6-Azidohex-l-yn-l-yl)phenyl)-l-methyl-8-(l-methyl- lH -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 166 6-(4-( 1-methyl-8-(l -methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,1 -cyclopropane] (165-1) (0.09 g, 162 µιηοΐ) was dissolved in DMF (0.809 mL) and sodium azide (21.0 mg, 324 µιηοΐ) was added. The reaction was heated to 50 °C for 14 hours and concentrated. The residue was purified by column chromatography (silica, 0-10% MeOH in DCM) to give 6-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l-cyclopropane] (166-1) (0.09 g, 81%) as a clear oil. LC/MS (ES+): m/z 502.5 [M + H]+

Intermediate 167-1. Preparation of 8-(4-(l-Methyl-8-(l-methyl-l H-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)oct-7-yn- l-ol Scheme 167.

6-(4-Chlorophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l-cyclopropane] (209-2) (0.150 g, 361 µ οΐ) and cesium carbonate (305 mg, 938 µ οΐ ) were dissolved in ACN (2.40 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen. Oct-7-yn-l-ol (158-1) (66.4 µ , 469 µ οΐ) was added followed by dicyclohexyl(2 (17.2 mg, 36.1 µ οΐ) and XPhos-Pd-G3 (30.5 mg, 36.1 µ οΐ) . The reaction tube was evacuated and back-filled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered through celite and concentrated. The product was purified by column chromatography (silica, 0-5% MeOH in DCM) to give 8-(4-( 1-methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l-cyclopropane] (167-1) (81.0 mg, 44.5 %) as a clear oil. LC/MS (ES+): m/z 505.5 [M + H]+

Intermediate 168-1. Preparation of 8-(4-(l-Methyl-8-(l-methyl-l H-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)oct-7-yn- 1-yl methanesulfonate Scheme 168.

8-(4-(1-Methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)oct-7-yn-l-ol (167-1) (0.1 g, 0.1981 mmol) was dissolved in DCM ( 1.32 mL ) . Triethylamine (52.9 , 0.3962 mmol) was added and the solution cooled to 0 °C. Methanesulfonyl chloride (18.3 , 0.2377 mmol) was added, the reaction stirred for 30 min and concentrated to give 8-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)oct-7-yn- 1- yl methanesulfonate (168-1) ( 115 mg, 100 %) as a clear oil.LC/MS (ES+): m/z 583.3 [M + H]+

Intermediate 169-1. Preparation of 6-(4-(8-Azidooct-l-yn-l-yl)phenyl)-l-methyl-8-(l- methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 169. 8-(4-(l-Methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l-cyclopropane] (168-1) (.1 15 g, 197 mol) was dissolved in DMF ( 1 mL) and sodium azide (25.6 mg, 394 µιηοΐ) was added. The reaction was heated to 50 °C for 1 hour and concentrated. The residue was purified by column chromatography (silica 0-10% MeOH in DCM) to give 8-(4-( 1-methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l-cyclopropane] (169-1) (0.065 g, 63 %) as a clear oil. LC/MS (ES+): m/z 531.1 [M + H]+

Intermediate 170-1. Preparation of 10-((4-(l-Methyl-8-(l-methyl-li/-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)amino)decan-l-ol Scheme 170. 6-(4-Chl orophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (0.3 g, 723 µιηοΐ), 10-aminodecan-l-ol (170-1) (125 mg, .723 µιηοΐ), LHMDS (2.45 mL, 2.45 mmol),

BrettPhos (39 mg, .0723 µιηοΐ), and BrettPhos P d G3 (58 mg, .0723 µιηοΐ) were placed in a sealable reaction tube under argon. Dioxane (4.82 mL ) (degassed with argon for 15 min) was added, the tube sealed and heated to 90 °C for 14 hrs. The reaction was diluted with EtOAc, filtered and concentrated. The residue was purified by column chromatography (siica, 0-10% MeOH in DCM) to give 10-((4-(l-methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)amino)decan-l-ol (170-2) (40 mg, 10.0 %) of an off white solid. LC/MS (ES+): m/z 552.6 [M + H ]+

Intermediate 171-1. Preparation of N-(10-Azidodecyl)-4-(l-methyl-8-(l-methyl-l H -pyrazol- 4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)aniline Scheme 171. 10-((4-(l-Methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l -cyclopropane] (170-2) (0.04 g, 0.07249 mmol) and N-ethyl-N- isopropylpropan-2-amine (25.2 , 0.1449 mmol) were dissolved in DCM (483 µΙ_, ) and cooled to 0 °C. Methanesulfonyl chloride (7.22 µ , 0.09423 mmol) was added and the bath removed. The reaction was stirred for 2 hours and the solvent was removed. The residue was dissolved in DMF (0.5 mL), sodium azide (9.41 mg, 0.1449 mmol) was added and the reaction was stirred for 16 hours at 50 °C. After cooling the reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give N-(10-azidodecyl)-4-(l-methyl-8-(l- methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1-cyclopropane] (171-1) (21.0 mg, 50.2 %) as a white solid. LC/MS (ES+): m/z 578.2 [M + H]+

Intermediate 172-2. Preparation of 10-(4-(l-Methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)dec-9- yn-l-ol Scheme 172. 6-(4-Chlorophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (0.150 g, 361

µ οΐ) and cesium carbonate (305 mg, 938 µιηοΐ ) were taken up in ACN (2.40 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen. Dec-9-yn-l-ol (172-1) (164 µ , 0.9399 mmol) was added followed by dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (17.2 mg, 36.1 µ οΐ) and

XPhos-Pd-G3 (30.5 mg, 36.1 µ οΐ) . The reaction tube was evacuated and backfilled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered through celite and concentrated. The residue was purified by column chromatogaphy (silica, 0-5% MeOH in DCM) to give 10-(4-(l- methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1- cyclopropane] (172-2) (129 mg, 33.5 %) as an off white solid. LC/MS (ES+): m/z 533.8 [M + H ]+

Intermediate 173-1. Preparation of 6-(4-(10-Azidodec-l-yn-l-yl)phenyl)-l-methyl-8-(l- methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 173 10-(4-( 1-Methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)dec-9-yn-l-ol (172-2) (0.130 g, 0.2440 mmol) and DIEA (84.9 L , 0.488 mmol) were dissolved in DCM (1.62 mL ) and cooled to 0 °C. Methanesulfonyl chloride (24.5 , 0.3 172 mmol) was added and the bath removed. The reaction was stirred for 2 hours and the solvent was removed. The residue was dissolved in DMF (1.62 mL), sodium azide (31.7 mg, 0.488 mmol) was added and the reaction was stirred for 16 hours at 50 °C. After cooling the reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(4-(10-azidodec-l-yn-l-yl)phenyl)-l- methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (173-1) (100 mg, 73.5 %) as a white solid. LC/MS (ES+): m/z 559.2 [M + H]+

Intermediate 174-1. Preparation of 4-(4-(l-Methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3- yn-l-ol Scheme 174. 6-(4-Chl orophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (.300 g, 7230 mmol) cesium carbonate (609 mg, 1.87 mmol) were dissolved in ACN (4.82 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen. But-3-yn-l-ol (150-1) (71.0 L , 0.9399 mmol) was added followed by dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (34.4 mg, .0723 mmol) and XPhos-Pd-G3 (61.1 mg, .0723 mmol). The reaction tube was evacuated and backfilled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered and concentrate. The residue was purified by column chromatography (silica 0-5% MeOH in DCM) to give 4-(4-(l-methyl-8-(l-methyl-lH- pyrazol-4-yl)spiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)but-3-yn-l-ol (174-1) (0.222 g, 68.5%) as a clear oil. LC/MS (ES+): m/z 449.5 [M +

H ]+

Intermediate 175-1. Preparation of 6-(4-(4Aazidobut-l-yn-l-yl)phenyl)-l-methyl-8-(l- methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 175. 4-(4-(l-Methyl-8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3-yn-l-ol (174-1) (0.035 g, 0.07803 mmol) and N-ethyl-N-isopropylpropan-2-amine (27 , 0 .1560 mmol) were dissolved in DCM (0.5 mL ) and cooled to 0 °C. Methanesulfonyl chloride (7.83 , 0.1014 mmol) was added and the bath removed. The reaction was stirred for 2 hours and the solvent was removed. The residue was dissolved in DMF (0.5 mL), sodium azide (10.1 mg, 0.1560 mmol) was added and the reaction was stirred for 16 hours at 50 °C. After cooling the reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(4-(4-azidobut-l- yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l-cyclopropane] (175-1) (12.0 mg, 32.5 %) as a white solid. LC/MS (ES+): m/z 475.1 [M + H]+

Intermediate 176-1. Preparation of 10-(4-(l-Methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)dec-9- yn-l-amine Scheme 176. 6-(4-( 10-Azidodec- 1-yn- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH -pyrazol -4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (173-1) (0.067 g, 0.1201 mmol) and triphenylphosphine (47.2 mg, 0.1801 mmol) were stirred for 16 hours in a mixture of

THF (600 µΙ_, ) and water (120 µΙ_, ) . The reaction was concentrated and purified by column chromatography (silica, 0-15% MeOH in DCM) to give 10-(4-(l-methyl-8-(l -methyl- lH-pyrazol-

4-yl)spiro[benzo[f][l,2,4]triazolo[4,3 -a] [l,4]diazepine-4,l -cyclopropane] (176-1) (30.0 mg, 47.0

% ) as a white solid. LC/MS (ES+): m/z 532.5 [M + H ]+

Intermediate 177-2. Preparation of 2,2-Dimethyl-4-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3- yn-l-ol Scheme 177. 6-(4-Chl orophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (.300 g, 7230 mmol) cesium carbonate (609 mg, 1.87 mmol) were dissolved in ACN (4.82 mL ) (degassed by bubbling argon into solution for 15 min) in an oven dried reaction tube fitted with a septa under nitrogen. 2,2-dimethylbut-3-yn-l-ol (177-1) (92 mL, 0.9399 mmol) was added followed by dicyclohexyl(2',4',6'-triisopropyl-[l ,r-biphenyl]-2-yl)phosphine (34.4 mg, .0723 mmol) and

XPhos-Pd-G3 (61.1 mg, .0723 mmol). The reaction tube was evacuated and backfilled with argon three times. The septa was replaced with a teflon scew-cap and the reaction heated in a 90 °C bath for 14 hrs. The reaction was diluted with EtOAc, filtered and concentrate. The residue was purified by column chromatography (silica 0-5% MeOH in DCM) to give 2,2-dimethyl-4-(4-(l-methyl-8-

( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]- 6-yl)phenyl)but-3-yn-l-ol (177-2) (0.223 g, 64.8%) as a clear oil. LC/MS (ES+): m/z M l .9 [M +

H ]+

Intermediate 178-1. Preparation of 2,2-Dimethyl-4-(4-(l-methyl-8-(l-methyl-l H-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3- yn-l-yl methanesulfonate Scheme 178.

2,2-Dimethyl-4-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)but-3 -yn- 1- ol (177-2) (.225 g, 472 µιηοΐ) was dissolved in DCM (2.35 mL ) and DIEA (106 , 613 µιηοΐ) was added. The reaction was cooled to 0°C and methanesulfonyl chloride (43.7 , 566 µ οΐ) was added. The solution was stirred at 0°C for 5 min followed by 2 hours at room temperature. The solvent was removed under reduced pressure the residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 2,2-dimethyl-4-(4-(l-methyl-8-(l-methyl- lH -pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan ]-6- yl)phenyl)but-3-yn-l-yl methanesulfonate (17801) (200 mg, 76.6%) of a clear oil. LC/MS (ES+): m/z 555.3 [M + H ]+

Intermediate 179-1. Preparation of 6-(4-(4-Azido-3,3-dimethylbut-l-yn-l-yl)phenyl)-l- methyl-8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 179.

2,2-Dimethyl-4-(4-(l-methyl-8-(l -methyl- lH -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)but-3 -yn- 1- yl methanesulfonate (178-1) (0.04 g, 72. 1 µ οΐ) and sodium azide (9.36 mg, 144 µ οΐ) were taken up in DMF (360 µΐ ) . The atmosphere was purged with argon and the reaction heated for 16 hours at 90°C. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 6-(4-(4-azido-3 ,3-dimethylbut- 1-yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane (179-1) (10.0 mg, 27.7 %) as a white solid. LC/MS (ES+): m/z 502.7 [M + H ]+ Intermediate 180-1. Preparation of r -Butyl 4-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)piperazine-l-carboxylate Scheme 180.

6-(4-Chlorophenyl)- l-methyl-8-(l -methyl- lH -pyrazol -4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (0.5 g, 1.20 mmol), ter t-butyl piperazine-l-carboxylate (79-1) (288 mg, 1.55 mmol), sodium 2-methylpropan-

2-olate (148 mg, 1.55 mmol), RuPhos (55.9 mg, 120 µιηοΐ) and RuPhos P d G3 (100 mg, 120 µιηοΐ) were taken up in THF (2.40 mL ) (degassed by bubbling argon for 30 min) and the atmosphere purged with argon. The vial was sealed and the reaction heated to 85°C for 16 hours. After cooling, the reaction was diluted with EtOAc, filtered and concentrated. The residue was purified by column chromatography (silica, 0-10%) MeOH in DCM) to give ter t-butyl 4-(4-(l-methyl-8-(l- methyl- lH -pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)piperazine-l-carboxylate (180-1) (670 mg, 1.18 mmol, 98.9 % ) as an off white solid. LC/MS (ES+): m/z 566.1 [M + H ]+

Intermediate 181-1. Preparation of l-Methyl-8-(l-methyl-l H -pyrazol-4-yl)-6-(4-(piperazin- l-yl)phenyl)spiro [benzo [ ] [1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropane] Scheme 181. t r t-Butyl 4-(4-(l -methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)piperazine-l-carboxylate (180-1) (0.670 g, 1.18 mmol) was dissolved in dioxane (2.94 mL ) and added to a solution of hydrogen chloride (2.95 mL, 11.8 mmol) (4 M dioxane). After stirring for 2 hours the solvent is removed to give 1-methyl- 8-(l-methyl-lH-pyrazol-4-yl)-6-(4-(piperazin-l-yl)phenyl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l '-cyclopropane] (181-1) isolated as the HC1 salt (520 mg, 94.8 %) as a dark red solid. LC/MS (ES+): m/z 465.5 [M + H]+

Intermediate 182-2. Preparation of 6-(4-(4-(2-Chloroethyl)piperazin-l-yl)phenyl)-l-methyl-

8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 182. 1-Methyl-8-(l -methyl- lH-pyrazol-4-yl)-6-(4-(piperazin- 1- yl)phenyl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] hydrochloride 181-1) (0.130 g, 259 µιηοΐ), potassium carbonate (71.5 mg, 518 µιηοΐ) and l-bromo-2- chloroethane (182-1) (32.2 , 388 µιηοΐ) were stirred in DMF (647 µL· ) for 16 hours. The reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(4-(4-(2-chloroethyl)piperazin-l-yl)phenyl)-l-methyl-8-(l -methyl- 1H - pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (182-2) (0.044 g, 44%) as a yellow oil. LC/MS (ES+): m/z 527.5 [M + H]+

Intermediate 183-1. Preparation of 6-(4-(4-(2-Azidoethyl)piperazin-l-yl)phenyl)-l-methyl- 8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 183.

6-(4-(4-(2-Chloroethyl)piperazin- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol -4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (182-2) (.044 g, 83.4

µ οΐ) and sodium azide (10.7 mg, 166 µ οΐ) were taken up in DMF (417 µΙ_, ) and heated to 50°C for 24 hrs. The reaction was diluted with EtOAc and concentrated. The residue was purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(4-(4-(2-azidoethyl)piperazin- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropane] (183-1) (36.0 mg, 80.8 %) as an off white solid. LC/MS (ES+): m/z 534.5 [M + H]+ Intermediate 184-2. Preparation of r -Butyl 6-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)-2,6- diazaspiro [3.3] heptane-2-carboxylate Scheme 184.

6-(4-Chlorophenyl)-l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l-cyclopropane] (209-2) (.2 g, 482 µηιοΐ), t rt-butyl 2,6-diazaspiro[3.3]heptane-2-carboxylate oxalate (184-1) (166 mg, 578 µιηοΐ), sodium 2-methylpropan-2-olate (101 mg, 1.06 mmol), RuPhos (22.4 mg, 48.2 µιηοΐ), and RuPhos Pd G3 (40.3 mg, 48.2 µιηοΐ) were placed in a sealable reaction tube and placed under argon. THF (964

) (degassed with argon for 15 min) was added, the tube sealed and heated to 90 °C for 14 hrs. The reaction was diluted with EtOAc, filtered through celite and concentrated. The residue was purified by column chromatography (silica, 0-10% MeOH in DCM) to give t rt-butyl 6-(4-(l- methyl-8-( 1-methyl- lH -pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-6-yl)phenyl)-2,6-diazaspiro[3.3]heptane-2-carboxylate (184-2) (75.0 mg, 27.0 %) of an off white solid. LC/MS (ES+): m/z 578.1 [M + H]+

Intermediate 185-1. Preparation of 6-(4-(2,6-Diazaspiro[3.3]heptan-2-yl)phenyl)-l-methyl- 8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 185. t rt-Butyl 6-(4-(l -methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l-cyclopropane] (184-2) (75 mg, 130 µιηοΐ) was dissolved in DCM ( 1 mL ) and 2,2,2-trifluoroacetic acid (396 , 5.19 mmol) was added. After 2 hours the reaction was concentrate. The residu was purified by column flash chromatography (silica, 0-15% MeOH in

DCM with H 3) to give 6-(4-(2,6-diazaspiro[3.3]heptan-2-yl)phenyl)-l-methyl-8-(l-methyl-lH- pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (185-1) (44.0 mg, 71.0 %) as a brown oil. LC/MS (ES+): m/z All [M + H]+

Intermediate 186-1. Preparation of 6-(4-(4-(6-Chlorohexyl)piperazin-l-yl)phenyl)-l- methyl-8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 186. 1-Methyl-8-( 1-methyl- lH-pyrazol-4-yl)-6-(4-(piperazin- 1-yl)phenyl)-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine hydrochloride (182-1) (0.150 g, 315 µιηοΐ) was taken up in DMF ( 0.640 mL). cesium carbonate (215 mg, 661 µιηοΐ) was added followed by 1-bromo- 6-chlorohexane (51.6 L, 346 µιηοΐ) and the reaction stirred for 12 hours. The reaction was concentrated and the residue purified by column chromatography (silica 0-10% MeOH in DCM) to give 6-(4-(4-(6-chlorohexyl)piperazin- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (186-2) (0.034 mg, 19.5%). LC/MS (ES+): m/z 583.6 [M + H]+

Intermediate 187-1. Preparation of 6-(4-(4-(6-Azidohexyl)piperazin-l-yl)phenyl)-l-methyl- 8-(l-methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropane] Scheme 187. 6-(4-(4-(6-Chlorohexyl)piperazin- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (186-2) (0.034 g, 58.3

µ οΐ ) was dissolved in DMF (291 µΙ_, ) and sodium azide (7.54 mg, 116 µιηοΐ ) was added. The atmosphere was purged with argon and the reaction heated to 50°C for 8 hours. The reaction was concentrated and the residue purified by column chromatography (silica, 0-15% MeOH in DCM) to give 6-(4-(4-(6-azidohexyl)piperazin- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (187-1) (24.0 mg, 69.9

% ) of a white solid. LC/MS (ES+): m/z 590.6 [M + H ]+

Intermediate 188-2. Preparation of (R )-2-(2-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)- 5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethyl)isoindoline-l,3-dione Scheme 188. (R)-7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one 2,2,2-trifluoroacetate (74-1) (0.025 g, 0.04784 mmol) and 2-(l,3-dioxoisoindolin-2-yl)ethanesulfonyl chloride (188-1) (17.0 mg, 0.06219 mmol) were taken up in DCM (239 µΙ_, ) under nitrogen and cooled to 0 °C. Triethylamine (19.9 µ , 0 .1435 mmol) was added and the reaction stirred for 4 hours at room temperature. The solvent was removed and the product was purified by column chromatography (silica, 0-15% MeOH in DCM) to give (R)- 2-(2-((4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2- c]pyridin-2-yl)methyl)-3 -methylpiperazin- 1-yl)sulfonyl)ethyl)isoindoline- 1,3-dione (188-2) ( 0.019 g, 61.6%) as a white solid. LC/MS (ES+): m/z 646.5 [M + H]+

Intermediate 189-1. Preparation of (R)-2-((4-((2-Aminoethyl)sulfonyl)-2-methylpiperazin- l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one Scheme 189.

(R)-2-(2-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)ethyl)isoindoline-l,3- dione (188-2) (0.006 g, 0.009291 mmol) was taken up in a solution of hydrazine hydrate ( 1 mL) (0.2 M MeOH) and stirred for 16 hours. The solvent was removed and the residue purified by reverse phase HPLC to give (R)-2-((4-((2-aminoethyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)- 7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (189-1) isolated as the TFA salt (3.00 mg, 41.7 %) as a thin film. LC/MS (ES+): m/z 516.5 [M + H]+

Intermediate 190-3. Preparation of 2-(10-(l,3-Dioxoisoindolin-2-yl)decyl)isothiouronium bromide Scheme 190

A suspension of 2-(10-bromodecyl)isoindoline-l,3-dione (190-1) (366 mg, 1 mmol) and thiourea (190-2) (76.1 mg, 1 mmol) in EtOH ( 1 mL) were heated to 70 °C in a glass vial under nitrogen for 3 hours. The solvent was removed and the white solid triturated with ether, filtered, and washed with additional ether to give 2-(10-(l,3-dioxoisoindolin-2-yl)decyl)isothiouronium bromide (190-3) (235 mg, 0.53 11 mmol, 53.1 %) as a white powder. LC/MS (ES+): m/z 326.8 [M + H]+

Intermediate 191-1. Preparation of 10-(l,3-Dioxoisoindolin-2-yl)decane-l-sulfonyl chloride Scheme 191. N-Chlorosuccinimide (240 mg, 1.80 mmol), ACN (.723 mL) and 2 M HC1 (0.126 mL) were cooled in an ice bath. To this suspension 10-(l,3-dioxoisoindolin-2-yl)decyl carbamimidothioate hydrobromide 190-3) (0.2 g, 0.4520 mmol) (was added in small portions over 10 minutes. The bath was removed and the reaction stirred for 16 hours. Water was added and the white precipitate collected and dried to givel0-(l,3-dioxoisoindolin-2-yl)decane-l-sulfonyl chloride (191-1) (89.0 mg, 51.1 %) as a white solid. LC/MS (ES+): m/z 386.3 [M + H]+

Intermediate 192-1. Preparation of (R )-2-(10-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)- 5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)decyl)isoindoline-l,3-dione Scheme 192

(R)-7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (74-1) (0.04 g, 0.08989 mmol) was dissolved in DMF (0.5 mL) and triethylamine (45 mg, 0.4494 mmol) was added. The solution was stirred for 5 minutes and 10-(l,3-dioxoisoindolin-2-yl)decane-l-sulfonyl chloride (191-1) (52.0 mg, 0.1348 mmol) was added in one portion. The reaction was stirred for 16 hours at room temperature purified by reverse phase HPLC to give (R)-2-(10-((4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazin-l-yl)sulfonyl)decyl)isoindoline-l,3-dione (192-1) (44.0 mg, 64.6 %) as a white solid. LC/MS (ES+): m/z 758.7 [M + H]+

Intermediate 193-1. Preparation of (R)-2-((4-((10-Aminodecyl)sulfonyl)-2-methylpiperazin- l-yl)methyl )-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5 H )-one Scheme 193.

(R)-2-(10-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)decyl)isoindoline-l,3- dione (192-1) (.03 g, 0.03958 mmol) was dissolved in 0.2 M methanolic hydrazine (989 , stirred for 16 hours and concentrated. The residue was purified by column chromatography (silica, 0- 15% MeOH in DCM) to give (R)-2-((4-((10-aminodecyl)sulfonyl)-2-methylpiperazin-l- yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (193- 1) (9.00 mg, 0.01433 mmol, 36.2 %) as a thin film. LC/MS (ES+): m/z 628.6 [M + H]+

Intermediate 194-1. Preparation of 2-(12-Bromododecyl)isoindoline-l,3-dione Scheme 194 Potassium l,3-dioxoisoindolin-2-ide (194-1) (2 g, 10.7 mmol) was suspended in DMF (10.7 mL) and 1,12-dibromododecane (194-2) (7 g, 21.4 mmol) was added. The reaction was stirred for 16 hours at room temperature. Acetone (50 mL) was added and the precipitated salts filtered. The acetone was removed under reduced pressure and water was added. The aqueous layer was extracted with diethyl ether (2 x 50 mL) and the combined organic layers washed with 5% aq LiCl solution, brine, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (silica, 0-100% EtOAc in hexane) to give 2-(10-bromodecyl)isoindoline- 1,3-dione (194-3) (0.640 g, 15.2 %) as a white solid. LC/MS (ES+): m/z 394.3 [M + H]+

Intermediate Preparation of 12-(l,3-Dioxoisoindolin-2-yl)dodecane-l-sulfonyl chloride Scheme 195. N-Chlorosuccinimide (534 mg, 4.00 mmol), ACN (1.6 mL) and .28 mL of 2 M HC1 were cooled in an ice bath. To this suspension 12-(l,3-dioxoisoindolin-2-yl)dodecyl carbamimidothioate hydrobromide (195-1) (470 mg, 1 mmol) was added in small portions over 10 minutes. The bath was removed and the reaction was stirred for 16 hours. Water was added and a white precipitate forms. The mixture was filtered, washed with water and dried to give 12-(1,3- dioxoisoindolin-2-yl)dodecane-l-sulfonyl chloride (195-2) (330 mg, 79.9 %) as a white solid. LC/MS (ES+): m/z 414.5 [M + H]+

Intermediate 196-1. Preparation of (R)-2-(12-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)- 5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)dodecyl)isoindoline-l,3-dione Scheme 196

(R)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one 2,2,2-trifluoroacetate (74-1) (.025 g, 0.04784 mmol) was dissolved in DMF (0.5 mL) and triethylamine (19.9 , 0.1435 mmol) was added. The solution was stirred for 5 min and 12-(l,3-dioxoisoindolin-2-yl)dodecane-l-sulfonyl chloride (195-2) (39.6 mg, 0.09568 mmol) was added in one portion. The reaction was stirred for eight hours and the solvent removed. The product was purified by column chromatography (silica, 0-15% MeOH in DCM) to give (R)-2-(12-((4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)dodecyl)isoindoline- 1,3-dione (196-1) (29.0 mg, 77.1 %) as a white solid. LC/MS (ES+): m/z 786.6 [M + H]+ Intermediate 197-1. Preparation of (R)-2-((4-((12-Aminododecyl)sulfonyl)-2- methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2- c]pyridin-4(5H )-one Scheme 197

(R)-2-( 12-((4-((7-(3-(cyclopropylmethoxy) dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)dodecyl)isoind 1,3-dione (196-1) (.02 g, 0.02544 mmol) was dissolved in 0.2 M methanolic hydrazine (636 µ ) and stirred for 16 hours. The solvent was removed and the residue was purified column chromatography (silica, 0-15% MeOH in DCM) to give (R)-2-((4-((12-aminododecyl)sulfonyl)- 2-methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2- c]pyridin-4(5H)-one (197-1) (10.0 mg, 0.01524 mmol, 60.2 %) as a thin film. LC/MS (ES+): m/z 656.7 [M + H]+

Intermediate 198-2. Preparation of 2-(6-(l,3-Dioxoisoindolin-2-yl)hexyl)isothiouronium bromide Scheme 198 A suspension of 2-(6-bromohexyl)isoindoline-l,3-dione (198-1) (310 mg, 1 mmol) and (190-2) thiourea (76. 1 mg, 1 mmol) in EtOH ( 1 mL) were heated to 70 °C for 3 hours. The solvent was removed and the white solid triturated with ether, filtered, and washed with additional ether to give 2-(6-(l,3-dioxoisoindolin-2-yl)hexyl)isothiouronium bromide (198-2) (344 mg, 89. 1%) as a white powder.MH+ 306.7 LC/MS (ES+): m/z 306.7 [M + H]+

Intermediate 199-1. Preparation of 6-(l,3-Dioxoisoindolin-2-yl)hexane-l-sulfonyl chloride heme 199

N-Chlorosuccinimide (240 mg, 1.80 mmol), ACN (1.6 mL) and 0.28 mL of 2 M HC1 were cooled in an ice bath. To this suspension 10-(l,3-dioxoisoindolin-2-yl)decyl carbamimidothioate hydrobromide (198-2) (.2 g, 452 µ οΐ) was added in small portions over 10 minutes. The bath was removed and the reaction stirred for 16 hours. Water was added, the white precipitate was collected and dried to give 6-(l,3-dioxoisoindolin-2-yl)hexane-l-sulfonyl chloride (199-1) (45.0 mg, 0.1364 mmol, 26.4 %) as a white solid. LC/MS (ES+): m/z 330.2 [M + H]+

Intermediate 200-1. Preparation of (R)-2-(6-((4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)- 5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)hexyl)isoindoline-l,3-dione Scheme 200 (R)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (74.1) 696 mmol. The solution was stirred for 5 min and 6-(l,3-dioxoisoindolin-2-yl)hexane-l-sulfonyl chloride (199-1) (28.9 mg, 0.08764 mmol) was added in one portion. After stirring for 30 min the reaction was concentrated and the product purified by column chromatography (silica, 0-15% MeOH in DCM) to give (R)-2-(6-((4- ((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2- yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)hexyl)isoindoline-l,3-dione (200-1) (32.0 mg, 67.6 %) as a white solid. LC/MS (ES+): m/z 702.6 [M + H]+

Intermediate 201-1. Preparation of (R)-2-((4-((6-Aminohexyl)sulfonyl)-2-methylpiperazin- l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5 H )-one Scheme 201

(R)-2-(6-((4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)sulfonyl)hexyl)isoindoline-l,3- dione (200-1) (.040 g, 0.05699 mmol) was dissolved in 0.2 M methanolic hydrazine (1.42 mL) and stirred for 16 hours. The solvent was removed and the residue was purified column chromatography (silica, 0-15% MeOH in DCM) to give (R)-2-((4-((6-aminohexyl)sulfonyl)-2- methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2- c]pyridin-4(5H)-one (201-1) (6.00 mg, 18.4 %) as a thin film. LC/MS (ES+): m/z 572.5 [M + H]+ Intermediate 202-2. Preparation of (R)-2-((4-((2-(2-(2-Azidoethoxy)ethoxy)ethyl)sulfonyl)- 2-methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2- c]pyridin-4(5H )-one Scheme 202

7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-(piperazin-l-ylmethyl)furo[3,2- c]pyridin-4(5H)-one hydrochloride (74-1) (.04 g, 92.8 µ ιοΐ) was dissolve in DFM ( 1 mL) and triethylamine (64.5 , 463 µη οΐ) was added followed by 2-(2-(2- azidoethoxy)ethoxy)ethanesulfonyl chloride (202-1) (33.2 mg, 129 µιηοΐ) . The reaction was stirred for 1 hour, diluted with DMSO and purified by reverse phase HPLC to give (R)-2-((4-((2- (2-(2-azidoethoxy)ethoxy)ethyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)-7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (202-2) (9 mg 15%). LC/MS (ES+): m/z 630.6 [M + H]+

Intermediate 203-1. Preparation of r -Butyl (R)-4-((7-(3,4-dimethoxyphenyl)-5-methyl-4- oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate Scheme 203 (R)- r t-Butyl 4-((7-bromo-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-

3-methylpiperazine-l-carboxylate (70.2) (.250 g, 567 µηιοΐ) and (3,4-dimethoxyphenyl)boronic acid (203-1) (205 mg, 1.13 mmol) were dissolved in THF (558 uL) in a resalabe tube fitted with a septa and degassed 0.5 M tripotassium orthophosphate (2.38 mL, 1.19 mmol) was added. Nitrogen was bubbled through the solution for 10 min followed by the addition of XPhos-Pd-G3 (47.9 mg,

56.7 µ οΐ) and XPhos (27.0 mg, 56.7 µ οΐ) . The septa was quickly replaced by a screw cap and the reaction heated to 80°C for 1 hour. After the reaction cooled EtOAc was added, the reaction filtered and concentrated. Purification of the residue by column chromatography (silica) 0-15% MeOH in DCM to give (R)- r t-butyl 4-((7-(3,4-dimethoxyphenyl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate (203.2) (0.088 g 64%) as a clear oil. LC/MS (ES+): m/z 498.5 [M + H]+

Intermediate 204-1. Preparation of (R)-7-(3,4-Dimethoxyphenyl)-5-methyl-2-((2- methylpiperazin-l-yl)methyl)furo[3,2-c]pyridin-4(5 H )-one Scheme 204

(R)- r t-Butyl 4-((7-(3,4-dimethoxyphenyl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2- c]pyridin-2-yl)methyl)-3-methylpiperazine-l-carboxylate (203-2) (.230 g, 0.4622 mmol) was taken up in dioxane (1.84 mL ) and hydrogen chloride (1.84 mL, 7.36 mmol, 4 M dioxane) was added. The reaction was stirred for 3 hours and concentrated to give (R)-7-(3,4-dimethoxyphenyl)- 5-methyl-2-((2-methylpiperazin-l-yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (204-1) (200 mg, 100 %) as a white solid. LC/MS (ES+): m/z 398.3 [M + H]+

Intermediate 205-2. Preparation of -Butyl (R)-(ll-(4 -((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro [3,2-c]pyridin-2- yl)methyl)-3-methylpiperazin-l-yl)-ll-oxoundecyl)carbamate Scheme 205

1l -((t rt-Butoxycarbonyl)amino)undecanoic acid (205.1) (62 mg, 0.206 mmol) was taken up in DMF (1.79 mL ) and cooled to 0°C. HATU (78.3 mg, 0.206 mmol) was added and the reaction stirred for 30 min at which time diisopropylethylamine (93.8 0.5391 mmol) was added followed by (R)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (204-1) (.075 g, 0.172 mmol). The bath was removed and the reaction stirred 16 hours and concentrated. The residue was purified by column chromatography, (silica, 0-15% MeOH in DCM) to give give(R)-fer t-butyl (l l-(4-((7- (3,4-dimethoxyphenyl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3- methylpiperazin-l-yl)-l l-oxoundecyl)carbamate (205-2) (92.0 mg, 78.6 %) LC/MS (ES+): m/z 681.7 [M + H]+

Intermediate 206-1. Preparation of (R)-2-((4-(ll-Aminoundecanoyl)-2-methylpiperazin-l- yl)methyl)-7-(3,4-dimethoxyphenyl)-5-methylfuro[3,2-c]pyridin-4(5 H )-one cheme 206

(R)-ter t-Butyl ( 1l-(4-((7-(3,4-dimethoxyphenyl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin- 2-yl)methyl)-3-methylpiperazin-l-yl)-l l-oxoundecyl)carbamate (205-2) (0.029 g, 0.1351 mmol) was dissolved in dioxane (0.5 mL) and HCl ( 0.5 mL, 4 M dioxane) was added and the reaction stirred for 3 hours and concentrated to give (R)-2-((4-(l l-aminoundecanoyl)-2-methylpiperazin- l-yl)methyl)-7-(3,4-dimethoxyphenyl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (206-1) (77.0 mg,

0.1325 mmol, 98.2 %) as a white solid. LC/MS (ES+): m/z 582.3 [M + H]+

Intermediate 207-2. Preparation of tert-Butyl (R)-(ll-(4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2- yl)methyl)-3-methylpiperazin-l-yl)-ll-oxoundecyl)carbamate Scheme 207 1l -((t rt-Butoxycarbonyl)amino)undecanoic acid (207-1) (64.3 mg, 0.2135 mmol) and N-ethyl- N-isopropylpropan-2-amine (157 , 0.854 mmol) were dissolved in DMF (2.13 mL ), stirred for 10 min and (R)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-2-((2-methylpiperazin-l- yl)methyl)furo[3,2-c]pyridin-4(5H)-one hydrochloride (204-1) (.095 g, 0.2135 mmol) was added. The reaction was stirred for 2 hours, concentrated and residue purified by column chromatography (silica, 0-10% MeOH in DC to give (R)- -butyl (l l-(4-((7-(3-(cyclopropylmethoxy)pyridin-4- yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-l 1- oxoundecyl)carbamate (207-2) ( 111 mg, 75.5 %). LC/MS (ES+): m/z 693 .4 [M + H]+

Intermediate 208-1. Preparation of (R)-2-((4-(ll-Aminoundecanoyl)-2-methylpiperazin-l- yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5 H )-one Scheme 208 (R)- rt-Butyl ( 1l-(4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-l l-oxoundecyl)carbamate (207-

2) (.080 g, 0.1 156 mmol) was dissolved in dioxane (289 µΙ_, ) hydrogen chloride (300 µ , 1.20 mmol, 4 M dioxane) was added. The reaction was stirred for 30 min and concentrated to give (R)- 2-((4-(l l-aminoundecanoyl)-2-methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin- 4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (208-1) (60.0 mg, 87.7 %) as a white solid. LC/MS (ES+): m/z 592.6 [M + H]+ Intermediate 209-2. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] Scheme 209.

8-Bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,l'-cyclopropane] (58-1) (0.06 g, 0.1450 mmol) l-methyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (209-1) (30.1 mg, 0.145 mmol)and potassium carbonate (60.1 mg, 0.435 mmol) were dissolved in dioxane (580 µ ) and water (290 µ ) (Ar was bubbled through solvents for 10 minutes prior) tetrakis (86.2 mg, 0.07468 mmol) was added and the vial was purged with Ar and sealed. The reaction was stirred at 95°C for 2 hours, at which time LCMS showed reaction was complete. Azeotroped reaction onto celite from toluene and purified on isco 4g column DCM - 10% MeOH/DCM to give 6-(4-chlorophenyl)-l-methyl-8-(l-methyl-lH- pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (209-2) (35.0 mg, 0.08435 mmol, 58.2 %) as a white solid. LCMS (ES+): m/z= 415 [M + H ]+

Intermediate 210-2. Preparation of 8-((4-(l-Methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)amino)octan-l-ol Scheme 210

Brought 6-(4-chlorophenyl)- 1-methyl-8-( 1-methyl- lH -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (209-2) (27 mg, 0.06507 mmol), 8-aminooctan-l-ol (210-1) ( 11.3 mg, 0.07808 mmol), BrettPhos (3.49 mg, 0.006507 mmol) and Brettphos G3 (5.19 mg, 0.006507 mmol) up in dioxane (433 i ) and added 1M (in THF) lithium bis(trimethylsilyl)amide (221 0.2212 mmol) all under Ar. Sealed reaction and heated to reflux overnight. Concentrated onto celite and purified by isco 0-20% MeOH/DCM to give 8-((4-(l-methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)amino)octan-l-ol (210-2) ( 11.0 mg, 0.02100 mmol, 32.3 %) as an oil. LC/MS (ES+): m/z 524.5 [M + H ]+ Intermediate 211-1. Preparation of 8-((4-(l-Methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)amino)octyl methanesulfonate Scheme 211

210-2 211-1 8-((4-(l-Methyl-8-(l-methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)amino)octan-l-ol (210-2) (.06 g, 0.1 145 mmol) is dissolved in DCM (572 µΙ_, ) N-ethyl-N-isopropylpropan-2-amine (39.7 , 0.229 mmol) is added and the solution cooled to 0 C . methanesulfonyl chloride (13.2 , 0.1717 mmol) is added, the reaction stirred for 30 min, concentrated and purified by silica gel flash chromatography (4 g Isco gold, DCM/MeOH 0-15%) to give 8-((4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)amino)octyl methanesulfonate (211-1) (45.0 mg, 0.07478 mmol, 65.3 %) as a clear oil. LC/MS (ES+): m/z 602 [M + H]+

Intermediate 212-1. Preparation of N-(8-Azidooctyl)-4-(l-methyl-8-(l-methyl-l H -pyrazol- 4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)aniline Scheme 212 211-1 212-1 8-((4 1-Methyl-8-( 1-methyl- 4,l'-cyclopropan]-6-yl)phenyl)amino)octyl methanesulfonate (211-1) (.02 g, 0.03323 mmol) wass dissolved in DMF (166 µΐ ), sodium azide (4.32 mg, 0.06646 mmol) was added, the reaction stirred for 16 hours at 50 C and concentrated. The residuet was purified by silica gel flash chromatography (4 g Isco gold, DCM/MeOH 0-15%) to give N-(8-azidooctyl)-4-(l-methyl-8-(l- methyl-lH-pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l (212-1) (10.0 mg,

0.01822 mmol, 54.9 %) LC/MS (ES+): m/z 559 [M + H]+

Intermediate 213-2. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(4-(prop-2-yn-l- yl)piperazin-l-yl)isoindoline-l,3-dione Scheme 213

2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione 213-1 (889 mg, 3.22 mmol), 1- (prop-2-yn-l-yl)piperazine (213-1) (.4 g, 3.22 mmol) and N-ethyl-N-isopropylpropan-2-amine (1.12 mL, 6.44 mmol) were dissolved in N-Methyl-2-pyrrolido (6.44 mL ) and heated in a mw at 120°C for 1hr. The reaction was diluted with water, extracted with EtOAc, and washed with water, 5 % LiCl, and brine. Dried over sodium sulfate and attempted purification by silica gel flash chromatography (24 g Isco gold, DCM/MeOH 0-10%) was unsuccessful. The impure fractions were concentrated, dissolved in DMSO and purified by reverse phase HPLC to give 2-(2,6- dioxopiperidin-3 -yl)-4-(4-(prop-2-yn- 1-yl)piperazin- 1-yl)isoindoline- 1,3-dione (213-2) (556 mg, 1.46 mmol, 45.5 %) as a fluffy yellow powder. ) LC/MS (ES+): m/z 381.7 [M + H]+

Intermediate 214-2. Preparation of r -Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l- oxoisoindolin-4-yl)(methyl)amino)piperidine-l-carboxylate Scheme 214

2 14-1 2 14-2

Add t rt-butyl 4-((2-(2,6-dioxopiperidin-3 -yl)- 1-oxoisoindolin-4-yl)amino)piperidine- 1- carboxylate (214-1) (155 mg, 0.3502 mmol) to 5 mL DCM in RBF at rt. To above suspension were added acetic acid (30.0 µΐ 0.5253 mmol) and formaldehyde (52.0 µΐ 0.7004 mmol) to form a clear solution. Stir reaction mixture for 10-15 min, then add NaBH(OAc)3 ( 111 mg, 0.5253 mmol) in one portion. Reaction solution turns to a bit cloudy after few minutes. LCMS indicate most of starting materials were consumed after stir overnight. FCC collect white solid t rt-butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4-yl)(methyl)amino)piperidine-l-carboxylate (214-2) (78.0 mg, 0.1708 mmol, 49.0 %). LC/MS (ES-): m/z 455.3 (M-H)

Intermediate 215-2. Preparation of 3-(4-(Methyl(piperidin-4-yl)amino)-l-oxoisoindolin-2- yl)piperidine-2,6-dione Scheme 215 215-1 215-2

t r t-Butyl 4-((2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindolin-4- yl)(methyl)amino)piperidine-l-carboxylate (215-1) (.078 g, 0.1708 mmol) was stirred for 2hs in a solution of DCM (1ml) and 2,2,2-trifluoroacetic acid (300 , 3.91 mmol). The solvent was removed to give 3-(4-(methyl(piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (215-1 )(65.0 mg, 0.1381 mmol, 80.9 %) as a brown gum. LC/MS (ES-): m/z 357 (M-H)-

Intermediate 216-2. Preparation of 3-(l-Oxo-4-(piperazin-l-yl)-l,3-dihyd pyrrolo [3,4-c] pyridin-2-yl)piperidine-2,6-dione Scheme 216

30-2 216-1 Charge 3-(4-chloro-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (30-2) (500 mg, 1.78 mmol) and tert-butyl piperazine-l-carboxylate (430 mg, 2.3 1 mmol) to a 20mL vial under inert atmosphere. Dissolve above solid mixture in Dimethyl Sulfoxide (5 mL ) with gentle stirring at room temp, followed by nitrogen purge for 5 min. Ethyldiisopropylamine (619 , 3.56 mmol) was added then the reaction mixture was heated to 70°C, and LCMS indicate only about 20% conversion after overnight. The temp was ramped up to 95 °C with additional 0.6 eq. of Ethyldiisopropylamine ( 1 mmol). After another 16 hrs, the reaction mixture was allowed to cool down and then pour to 0.1 M citric acid aq solution 20mL, form a light beige suspension. Solid product was collected via filtration to afford crude t rt-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxo- 2,3-dihydro-lH-pyrrolo[3,4-c]pyridin-4-yl)piperazine-l-carboxylate (216-1) (630 mg, 1.46 mmol, 82.4 %) directly used for next step. LC/MS (ES+): m/z 430.6 (M+H)+ Crude t rt-butyl 4-(2-(2,6-dioxopiperidin-3-yl)-l-oxo-2,3-dihydro-lH-pyrrolo[3,4- c]pyridin-4-yl)piperazine-l-carboxylate (216-1) (630 mg)was suspended in 5 mL DCM at 0°C, followed by TFA (496 uL, 7.29 mmol). The reaction mixture turned to clear light beige solution and LCMS indicate starting materials were all consumed in 4 hrs. The solvent was removed under reduced pressure and crude product (216-2) obtained as gummy solid was used directly for next step without further purification. LC/MS (ES+): m/z 330.2 (M+H)+

Example 1. Preparation of (S)-6-(4-Chlorophenyl)-l,4-dimethyl-8-(l-methyl-lH-pyrazol-4- yl)-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (Compound 1) Scheme 216.

63-1 Compound 1 (S)-8-Bromo-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine (63-1) (0.030 g, 0.07468 mmol) l-methyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (209-1) (15.5 mg, 0.07468 mmol) and potassium carbonate (20.6 mg, 0.1493 mmol) were dissolved in dioxane (298 and water (149 (Ar was bubbled through solvents for 10 minutes prior) tetrakis (8.62 mg, 0.007468 mmol) was added and the vial was purged with Ar and sealed. The reaction was stirred at 95°C for 2 hours, at which time LCMS showed reaction was complete. Azeotroped reaction onto celite from toluene and purified by isco 0 - 10% MeOH/DCM to give (S)-6-(4-chlorophenyl)-l,4-dimethyl-8-(l-methyl-lH-pyrazol-4-yl)- 4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (Compound 1) (25.0 mg, 0.06205 mmol, 83.3 %) as a white solid. LCMS (ES+): m/z= 403 [M + H]+ 1H NMR (400 MHz, Chloroform-d) δ 7.79 - 7.70 (m, 2H), 7.65 - 7.52 (m, 3H), 7.48 - 7.41 (m, 2H), 7.39 - 7.32 (m, 2H), 4.22 (q, J = 6.7 Hz, 1H), 3.95 (s, 3H), 2.66 (s, 3H), 2.08 (d, J = 6.7 Hz, 3H), 1.84 (s, 1H).

Example 2. Preparation of 6-(4-Chlorophenyl)-l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (Compound 2) Scheme 217.

8-Bromo-6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,l'-cyclopropane] (58-1) (0.06 g, 0.1450 mmol) l-methyl-4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-lH-pyrazole (209-1) (30.1 mg, 0.145 mmol)and potassium carbonate (60.1 mg, 0.435 mmol) were dissolved in dioxane (580 and water (290 (Ar was bubbled through solvents for 10 minutes prior) tetrakis (86.2 mg, 0.07468 mmol) was added and the vial was purged with Ar and sealed. The reaction was stirred at 95°C for 2 hours, at which time LCMS showed reaction was complete. Azeotroped reaction onto celite from toluene and purified on isco 4g column DCM - 10% MeOH/DCM to give 6-(4-chlorophenyl)-l-methyl-8-(l-methyl-lH- pyrazol-4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (Compound 2) (35.0 mg, 0.08435 mmol, 58.2 %) as a white solid. LCMS (ES+): m/z= 415 [M + H]+ 1H NMR (400 MHz, DMSO-de) δ 8.1 1 (d, J = 0.8 Hz, 1H), 7.90 - 7.80 (m, 2H), 7.68 (d, J = 8.5 Hz, 1H), 7.57 - 7.48 (m, 2H), 7.47 - 7.34 (m, 3H), 3.76 (s, 3H), 2.53 (s, 3H), 1.66 (ddd, J = 10.0, 7.1, 5.1 Hz, 1H), 1.33 (ddd, J = 9.9, 7.2, 5.2 Hz, 1H), 0.71 (dddd, J = 21.7, 10.0, 7.2, 5.0 Hz, 2H). Example 3. Preparation of 3-(4-((l-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 1) Scheme 218.

Degronimer 1

Brought 3-(l-oxo-4-(piperidin-4-ylamino)isoindolin-2-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (29-2) ( 111 mg, 0.2450 mmol) , 6-(4-chlorophenyl)-8-(l-(3-chloropropyl)-lH- pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (89- 1) (0.090 g, 0 .1885 mmol) and N-ethyl-N-isopropylpropan-2-amine (131 , 0.754 mmol) up in nmp (1.88 mL ) and heated to 75°C for two nights. Cooled to r.t. and loaded reaction mixture directly onto reversed phase isco 10-lOOACN/water + 0 .1% TFA. to give the TFA salt of 3-(4-((l- (3-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)propyl)piperidin-4-yl)amino)-l-oxoisoindolin-2- yl)piperidine-2,6-dione (Degronimer 1) (7.00 mg, 0.008936 mmol, 4.76 %) as a whitish solid. LCMS (ES+): m/z 784 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.00 (s, 1H), 9.30 - 9.13 (m, 1H), 8.29 - 8.16 (m, 1H), 8.02 - 7.88 (m, 2H), 7.78 - 7.73 (m, 1H), 7.60 - 7.55 (m, 2H), 7.50 - 7.47 (m, 2H), 7.43 (d, J = 2.2 Hz, 1H), 7.33 - 7.25 (m, 1H), 6.97 (d, J = 7.4 Hz, 1H), 6.83 (d, J = 8.0 Hz, 1H), 5.22 - 5.04 (m, 1H), 4.30 - 4.03 (m, 4H), 3.43 - 3.39 (m, 2H), 3.31 - 2.90 (m, 4H), 2.69 - 2.55 (m, 4H), 2.38 - 1.82 (m, 6H), 1.81 - 1.68 (m, 2H), 1.63 - 1.60 (m, 2H), 1.41 - 1.38 (m, 2H), 0.90 - 0.63 (m, 2H).

Example 4. Preparation of 3-(4-((l-(4-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 2) Scheme 219. Degronimer 2 Brought 3-(l-oxo-4-(piperidin-4-ylamino)isoindolin-2-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (29-2) (90.5 mg, 0.1983 mmol) , 8-(l-(4-chlorobutyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (86-1) (75 mg, 0.1526 mmol) and N-ethyl-N-isopropylpropan-2-amine (106 , 0.6104 mmol) up in NMP (1.52 mL ) and heated in a sealed vial at 75°C for two nights. Cooled reaction and loaded directly onto reversed phase isco 10-100% ACN/water + 0.1% TFA to give the TFA salt of 3-(4-((l-(4-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)butyl)piperidin-4-yl)amino)-l-oxoisoindolin-2- yl)piperidine-2,6-dione as the TFA salt (Degronimer 2) (12.0 mg, 0.01504 mmol, 9.91 %) as a white solid. LCMS (ES+): m/z 798 [M + H]+ ¾ NMR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 9.05 (s, 1H), 8.23 (s, 1H), 7.98 - 7.86 (m, 2H), 7.78 - 7.69 (m, 1H), 7.64 - 7.54 (m, 2H), 7.54 - 7.46 (m, 2H), 7.45 - 7.42 (m, 1H), 7.29 (t, J = 7.6 Hz, 1H), 6.98 (d, J = 13.8, 7.4 Hz, 1H), 6.84 (d, J = 13.3, 8.2 Hz, 1H), 5.20 - 5.04 (m, 1H), 4.35 - 4.04 (m, 4H), 3.69 - 3.54 (m, 1H), 3.53- 3.48 (m, 2H), 3.16 - 2.83 (m, 4H), 2.73 - 2.55 (m, 5H), 2.36 - 1.68 (m, 8H), 1.65 - 1.51 (m, 2H), 1.40 - 1.33 (m, 2H), 0.86 - 0.65 (m, 2H).

Example 5. Preparation of 3-(4-((l-(5-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 3) Scheme 220.

Degronimer 3

Brought 3-(l-oxo-4-(piperidin-4-ylamino)isoindolin-2-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (29-2) (92.0 mg, 0.2017 mmol) and 8-(l-(5-chloropentyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (83-1) (0.085 g, 0 .1681 mmol) up in toluene ~4 mL and azeotroped twice. Added M P (840 µ ) followed by N-ethyl-N-isopropylpropan-2-amine ( 117 , 0.6724 mmol) and added 4 A MS before sealing vial and heating at 75 °C for three nights. Loaded the reaction mixture directly onto reversed phase isco and isolated the TFA salt of 3-(4-((l-(5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione isolated as the TFA salt (Degronimer 3) (22.0 mg, 0.0271 1 mmol, 16.1 %) as a whitish solid. LCMS (ES+): m/z 812 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 9.09 (s, 1H), 8.22 (s, 1H), 7.92 (s, 2H), 7.79 - 7.70 (m, 1H), 7.63 - 7.55 (m, 2H), 7.49 (d, J = 8.7, 2.2 Hz, 2H), 7.44 (d, J = 2.2 Hz, 1H), 7.34 - 7.24 (m, 1H), 6.98 (d, J = 13.2, 7.4 Hz, 1H), 6.84 (d, J = 12.2, 8.1 Hz, 1H), 5.21 - 5.06 (m, 1H), 4.17 - 4.04 (m, 4H), 3.62 - 3.42 (m, 3H), 3.10 - 2.90 (m, 4H), 2.69 - 2.56 (m, 4H), 2.38 - 2.18 (m, 2H), 2.17 - 1.88 (m, 4H), 1.85 - 1.70 (m, 3H), 1.69 - 1.56 (m, 2H), 1.44 - 1.31 (m, 2H), 1.31 - 1.15 (m, 2H), 0.92 - 0.65 (m, 2H).

Example 6. Preparation of 3-(4-((l-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione Degronimer 4 Scheme 221. O Brought 8-(l-(6-chlorohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (78-1) (56.8 mg, 0.1095 mmol) , 3-(l-oxo-4-(piperidin-4-ylamino)isoindolin-2-yl)piperidine-2,6-dione 2,2,2- trifluoroacetate (29-2) (50 mg, 0.1095 mmol) and N-ethyl-N-isopropylpropan-2-amine (76.2 , 0.438 mmol) up in NMP (438 ΐ ) . Heated for 2 days at 75°C. Purified by prep HPLC directly 5- 99% ACN/Water w 0.1% TFA to give the TFA salt of 3-(4-((l-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propyl)piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione as the TFA salt (Degronimer 4) (13.0 mg, 0.016 mmol, 14.3%) as an off-white solid LCMS (ES+): m/z 826 [M + H]+ ¾ NMR (400 MHz, DMSO-de) δ 11.00 (s, 1H), 9.27 (s, 1H), 8.21 (s, 1H), 7.98 - 7.83 (m, 2H), 7.79 - 7.69 (m, 1H), 7.63 - 7.55 (m, 2H), 7.53 - 7.40 (m, 3H), 7.36 - 7.22 (m, 1H), 6.97 (d, J = 13.2, 7.4 Hz, 1H), 6.83 (d, J = 8.4 Hz, 1H), 5.21 - 5.05 (m, 1H), 4.30 - 4.01 (m, 4H), 3.66 - 3.46 (m, 2H), 3.10 - 2.85 (m, 4H), 2.68 - 2.54 (m, 4H), 2.33 - 2.20 (m, 1H), 2.20 - 1.88 (m, 4H), 1.84 - 1.66 (m, 3H), 1.66 - 1.54 (m, 3H), 1.47 - 1.38 (m, 2H), 1.34 - 1.18 (m, 3H), 0.89 - 0.66 (m, 2H).

Example 7. Preparation of 4-(4-(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 5) Scheme 222. Brought 6-(4-chlorophenyl)- 1-methyl-8-(l -(7-(piperazin- 1-yl)heptyl)- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (80-1) (50.8 mg, 0.073 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-

3) (26.2 mg, 0.0949 mmol) up in MP (730 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (50.8 µ , 0.292 mmol) before heating to 90°C for 16 hours. Cooled reaction and injected crude mixture onto prep HPLC 5-95% ACN/water + 0.1% TFA to give 4-(4-(7-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- l-yl)heptyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione as the TFA salt(Degronimer 5) (20.0 mg, 0.02382 mmol, 32.6 %) as a yellow solid. LCMS (ES+): m/z 840 [M + H]+.

¾ MR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 9.47 (s, 1H), 8.23 (s, 1H), 8.01 - 7.86 (m, 2H), 7.86 - 7.70 (m, 2H), 7.58 (d, J = 8.6 Hz, 2H), 7.57 - 7.36 (m, 5H), 5.16 - 5.03 (m, 1H), 4.08 (t, J = 6.9 Hz, 2H), 3.91 - 3.80 (m, 4H), 3.31 - 3.08 (m, 6H), 2.94 - 2.78 (m, 1H), 2.62 - 2.55 (m, 4H), 2 .10 - 1.94 (m, 1H), 1.84 - 1.57 (m, 5H), 1.44 - 1.18 (m, 7H), 0.87 - 0.69 (m, 2H).

Example 8. Preparation of 4-(4-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 6) Scheme 223. Degronimer 6 To a solution of 6-(4-chlorophenyl)-l-methyl-8-(l-(6-(piperazin-l-yl)hexyl)-lH-pyrazol- 4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] hydrochloride (82-1) (150 mg, 247 µηιοΐ) inDMF(5 mL) was added N-ethyl-N-isopropylpropan-2-amine (191 mg, 1.48 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (88.6 mg, 321 µιηοΐ) . The mixture was heated to 80 °C overnight. The mixture was concentrated and purified with combi-flash to give 4-(4-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[fJ[l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperazin- 1-yl)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 6) (99.3 mg, 120 µπιοΐ , 48.9 %) LCMS (ES+): w/z=825 [M + H]+ 1H MR (500 MHz, DMSO) δ 11.09 (s, 1H), 8.24 (s, 1H), 7.95-7.90 (m, 2H), 7.75-7.67 (m, 2H), 7.60-7.58 (m, 2H), 7.49-7.45 (m, 3H), 7.36-7.31 (m, 2H), 5.07 (dd, J = 5, 12 Hz, 1H), 4.08 (t, J = 7 Hz, 2H), 3.31-3.26 (m, 4H), 2.88-2.84 (m, 1H), 2.58-2.55 (m, 4H), 2.5 (m, 4H, under DMSO peak), 2.28-2.27 (m, 2H), 2.02-1.99 (m, 1H), 1.78-1.70 (m, 3H), 1.43-1.23 (m, 8H), 0.80-0.73 (m, 2H). Example 9. Preparation of 4-(4-(5-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 7) Scheme 224.

Brought 6-(4-chlorophenyl)- 1-methyl-8-(l -(5-(piperazin- 1-yl)pentyl)- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (250mg, 450 µηιοΐ), 2-

(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (124 mg, 450 µηιοΐ) up in DMF (5 mL ) and added DIPEA (174 mg, 1.35 mmol) . Stirred reaction at r.t. for 4 hours. Concentrated and was purified by prep TLC (DCM/MeOH=10/l)to afford 4-(4-(5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentyl)piperazin- 1-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 7) (35.0 mg, 43.1 µιηοΐ , 9.58 %) as a yellow solid. LCMS (ES+): m/z= 8 11 [M + H]+ 1H MR (400 MHz, DMSO-de): δ 11.10 (s, 1H), 8.24 (s, 1H), 7.97-7.88 (m, 2H), 7.78-7.65 (m, 2H), 7.61-7.54 (m, 2H), 7.52-7.43 (m, 3H), 7.39-7.29 (m, 2H), 5.10-5.06 (m,lH), 4.14-4.07 (m, 3H), 3.26 (br, 4H), 2.92-2.80 (m, 2H), 2.59-2.58 (m,4H), 2.29 (br, 2H), 2.02-1.99 (m, 2H), 1.81- 1.70 (m, 4H), 1.48-1.37 (m, 4H), 1.24-1.20 (m, 2H), 0.79-0.72 (m, 2H).

Example 10. Preparation of 4-(4-(4-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 8) Scheme 225.

88-1

To a solution of 6-(4-chlorophenyl)-l-methyl-8-(l-(4-(piperazin-l-yl)butyl)-lH-pyrazol- 4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] hydrochloride (88-1) (220 mg, 380 µηιοΐ) in DMF(4 mL) was added 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline- 1,3-dione (1-3) (136 mg, 494 µιηοΐ) and N-ethyl-N-isopropylpropan-2-amine (244 mg, 1.89 mmol). The mixture was heated to 80 °C overnight. The mixture was concentrated and purified with combi-flash to give crude and the crude was purified with prep-TLC(DCM:MeOH=10: 1) to give 4-(4-(4-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)butyl)piperazin- 1-yl)-2-(2,6-dioxopiperidin-3 - yl)isoindoline-l,3-dione (Degronimer 8) (55.6 mg, 69.7 µηιοΐ , 18.4 %). LCMS (ES+): m/z= 797 [M + H ]+

1H M R (500 MHz, MeOD) δ 11.09 (s, 1H), 8.25 (s, 1H), Ί .96-1.92 (m, 2H), 7.76-7.69 (m, 2H), 7.67-7.57 (m, 2H), 7.50-7.45 (m, 3H), 7.36-7.31 (m, 2H), 5.09 (dd, J = 7, 16 Hz, 1H), 4.12 (t, J =

8.5 Hz, 2H), 3.31-3.26 (m, 4H), 2.86-2.55 (m, 6H), 2.34-2.31 (m, 3H), 2.04-1.97 (m, 2H), 1.90-

1.72 (m, 4H), 1.41-1.23 (m, 3H), 0.77-0.75 (m, 2H).

Example 11. Preparation of Synthesis of 4-(4-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 9) Scheme 226

Degronimer 9

Brought 6-(4-chlorophenyl)-l-methyl-8-(l-(3-(piperazin-l-yl)propyl)-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (88-1) (250 mg, 474 µηιοΐ) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (170 mg, 616 µ οΐ) up in DMF (5 mL ) and added DIPEA (183 mg, 1.42 mmol) . Stirred reaction at r.t. for 4 hours. The reaction and was purified by preparitive TLC (DCM/MeOH=10/l) to afford 4-(4-(3-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)- lH-pyrazol- 1-yl)propyl)piperazin- 1-yl)-2-(2,6-dioxopiperidin-3 -yl)isoindoline- 1,3-dione

(Degronimer 9) (60.0 mg, 76.6 µιηοΐ , 16. 1 %) as yellow solid. LCMS (ES+): m/z= 783 [M + H]+ 1H MR (400 MHz, DMSO-de): δ 11.10 (s, 1H), 8.24 (s, 1H), 7.97-7.93 (m, 2H), 7.76-7.68 (m, 2H), 7.59 (d, J=10.5Hz, 2H), 7.48 (d, J=10.5Hz, 2H), 7.45 (m, 1H), 7.37-7.30 (m, 2H), 5.1 1-5.07 (m, 1H), 4.17-4.14 (m, 2H), 3.27 (br, 4H), 2.91-2.82 (m,lH), 2.59-2.50 (m, 8H, 4 under DMSO), 2.3 (br, 2H), 2.03-1.96 (m, 3H), 1.73-1.71 (m, 1H), 1.40-1.37 (m, 1H), 0.81-0.77 (m, 2H).

Example 12. Preparation of 4-(4-(2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione Degronimer 10 Scheme 227

Degronimer 10 To a solution of 6-(4-chlorophenyl)-l-methyl-8-(l-(2-(piperazin-l-yl)ethyl)-lH-pyrazol- 4-yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (94-1) (100 mg, 194 µηιοΐ) in DMF (2.0 mL ) was added 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-

3) (107 mg, 388 µιηοΐ) and N-ethyl-N-isopropylpropan-2-amine (125 mg, 970 µιηοΐ) .The reaction mixture was heated to 80 °C and stirred for 16 h . After cooling to room temperature, the reaction solution was purified directly by reversed phase fish (ACN/H2O/0.01M NH4C03) to afford yellow solid (35 mg) which was further purified by prep-TLC (DCM/MeOH=10/l) to afford target compound 4-(4-(2-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)ethyl)piperazin- 1-yl)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 10) (26.0 mg, 33.9 µπιοΐ , 17.4 % ) as a yellow solid. LCMS (ES+): m/z= 769 [M + H ]+ 1H M R (400 MHz, DMSO-de): δ 11.08 (s, IH), 8.23 (s, IH), 7.95-7.92 (m, 2H), 7.75-7.69 (m, 2H), 7.58 (d, J=8.5Hz, 2H), 7.44 (d, J=9.0Hz, 3H), 7.36 (d, J=6.5Hz, IH), 7.31 (d, J=8.5Hz, IH), 5.10-5.06 (m, IH), 4.26 (t, J=6.5Hz, 2H), 3.34-3.24 (br, 4H), 2.87-2.84 (m,lH), 2.78 (t, J=6.5Hz,

2H), 2.63-2.57 (br, 8H), 2.03-2.00 (m, IH), 1.73-1.71 (m, IH), 1.40-1.37 (m, IH), 0.81-0.77 (m, 2H).

Example 13. Preparation of 4-(((l-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione Degronimer 11) Scheme 228 Degr r er 11 Brought 8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo [f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (95-1) (0.015 g, 0.03099 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (9.64 mg, 0.03099 mmol) and N-ethyl-N-isopropylpropan-2-amine (5.39 µ , 0.03099 mmol) up in THF (0.5 mL). Added copper(I) iodide (2.95 mg, 0.01549 mmol) and stirred at r.t. for 5 hrs. Complete by lc/ms. Concentrated, dissolved in ~ 1.0 mL DMSO and filtered. Purified by MS directed prep HPLC 1-99% CH3CN/water w TFA to give 4-(((l-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 11) (23.0 mg, 28.9 umol, 93.4%) as a yellow solid after lyophilization. LCMS

(ES+): m/z= 796 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.15 (s, 1H), 7.97 (s, 1H), 7.92 - 7.85 (m, 2H), 7.69 (d, J = 8.5 Hz, 1H), 7.61 - 7.50 (m, 2H), 7.50 - 7.38 (m, 3H), 7.19 - 7.04 (m, 1H), 7.04 - 6.87 (m, 2H), 5.09 - 4.93 (m, 1H), 4.51 (s, 2H), 4.26 (t, J = 7.0 Hz, 2H), 4.05 (t, J = 7.0 Hz, 2H), 2.90 - 2.69 (m, 1H), 2.55 (s, 4H), 2.50 - 2.46 (m, 2H), 2.34 - 2.19 (m, 2H), 2.07 - 1.90 (m, 1H), 1.73 - 1.60 (m, 1H), 1.40 - 1.28 (m, 1H), 0.84 - 0.64 (m, 2H). Example 14. Preparation of 4-(((l-(4-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 12) Scheme 229

Degroreimer 12

Brought 8-(l-(4-azidobutyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (96-1) (53 mg, 0.1064 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5- 1) (33.1 mg, 0.1064 mmol) up in THF (1.06 mL ) and added N-ethyl-N-isopropylpropan-2-amine (27.7 i , 0.1596 mmol) followed by copper(I) iodide (10.1 mg, 0.0532 mmol). Stirred at r.t. for one night and concentrated before redissolving in dmso and purifying by prep HPLC, 5-95% ACN/water + 0.1% TFA. Isolated 4-(((l-(4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 12) (80.0 mg, 0.09885 mmol, 92.9 % ) as a yellow solid. LCMS (ES+): m/z= 810 [M + H ]+ H MR (300 MHz, DMSO-de) δ 11.10 (s, 1H), 8.21 (s, 1H), 8.06 - 7.86 (m, 3H), 7.84 - 7.70 (m, 1H), 7.65 - 7.42 (m, 6H), 7.22 - 6.96 (m, 3H), 5.06 (d, J = 12.5 Hz, 1H), 4.58 (s, 2H), 4.10 (s, 2H), 2.99 - 2.81 (m, 1H), 2.68 - 2.55 (m, 5H), 2.14 - 1.93 (m, 1H), 1.83 - 1.62 (m, 6H), 1.54 - 1.35 (m, 1H), 0.95 - 0.64 (m, 2H).

Example 15. Preparation of 4-(((l-(5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 13) Scheme 230

Brought 8-(l-(5-azidopentyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo [f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (97-1) (60 mg, 0.1 171 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (36.4 mg, 0.1 171 mmol) and N-ethyl-N-isopropylpropan-2-amine (30.4 , 0.1756 mmol) up in

THF (1.17 mL ) and added copper(I) iodide ( 11.1 mg, 0.05855 mmol) under Ar. Stirred O/N at r.t. complete by uplc. concentrated and redissolved in DMSO ~ 1 mL for prep HPLC purification. 1- 99% ACN/Water w tfa to give 4-(((l-(5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentyl)-lH-l,2,34riazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione

(Degronimer 13) (71.0 mg, 0.086 mmol, 73.6%) as a yellow solid. LCMS (ES+): m/z= 824 [M + H]+ lH MR (400 MHz, DMSO-d 6) 11.10 (s, 1H), 8.20 (s, 1H), 8.02 - 7.86 (m, 3H), 7.75 (d, J = 8.5 Hz, 1H), 7.67 - 7.53 (m, 3H), 7.53 - 7.38 (m, 3H), 7.19 - 6.95 (m, 3H), 5.05 (dd, J = 12.8, 5.3 Hz, 1H), 4.55 (d, J = 4.1 Hz, 2H), 4.27 (t, J = 7.1 Hz, 2H), 4.04 (t, J = 6.9 Hz, 2H), 2.96 - 2.72 (m, 1H), 2.65 - 2.54 (m, 5H), 2.02 - 1.98 (m, 1H), 1.90 - 1.61 (m, 5H), 1.52 - 1.32 (m, 1H), 1.19 - 1.02 (m, 2H), 0.91 - 0.68 (m, 2H).

Example 16. Preparation of 4-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 14) Scheme 231 Brought 8-(l-(6-azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane (98-1) (0.080 g, 0.1520 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (56.7 mg, 0.1824 mmol) and N-ethyl-N-isopropylpropan-2-amine (39.6 , 0.228 mmol) up in THF. Copper(I) iodide (14.4 mg, 0.076 mmol) was added and the reaction was stirred O/N. The reaction was concentrated and the crude residue was dissolved in ~ 1.0 mL DMSO. It was filtered and purified by mass directed Prep HPLC 1-99% CftCN/Water w TFA. After lyophilization recovered 4-(((l-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[fJ[l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4- yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 14) (105 mg, 0.1253 mmol, 82.6 % ) as a yellow solid. LCMS (ES+): m/z= 836 [M + H ]+ l H M R (500 MHz, DMSO-de) δ 11.09 (s, 1H), 8.20 (s, 1H), 8.02 - 7.86 (m, 3H), 7.74 (d, J = 8.5 Hz, 1H), 7.65 - 7.53 (m, 3H), 7.53 - 7.40 (m, 3H), 7.13 (d, J = 8.7 Hz, 1H), 7.08 - 6.97 (m, 2H), 5.04 (dd, J = 12.9, 5.3 Hz, 1H), 4.57 (s, 2H), 4.27 (t, J = 7.0 Hz, 2H), 4.03 (t, J = 7.0 Hz, 2H), 2.95

- 2.79 (m, 1H), 2.64 - 2.58 (m, 4H), 2.06 - 1.93 (m, 1H), 1.82 - 1.63 (m, 5H), 1.45 - 1.33 (m, 1H), 1.24 - 1.14 (m, 5H), 0.87 - 0.69 (m, 2H).

Example 17. Preparation of 4-((2-(l-(5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)-lH-l,2,3-triazol-4-yl)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 15) Scheme 232 Deg i er Brought 8-(l-(5-azidopentyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (97-1) (60 mg, 0.1 171 mmol) , 4-(but-3-yn-l-ylamino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (8-2) (38.0 mg, 0.1 171 mmol) , N-ethyl-N-isopropylpropan-2-amine (40.7 µ , 0.2342 mmol) and copper(I) iodide ( 11.1 mg, 0.05855 mmol) up in THF (1.17 mL ) under Ar and stirred O/N at r.t. Concentrated off solvent dissolved in DMSO, filtered and purified by prep HPLC. 1-99% ACN/water w TFA. Isolated 4-((2-(l-(5-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)pentyl)-lH-l,2,3-triazol-4-yl)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 15) (62.0 mg, 0.07404 mmol, 63.2 %) as a yellow solid after lyophilization. LCMS (ES+): m/z= 838 [M + H]+ lHNMR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.20 (s, 1H), 7.99 - 7.82 (m, 3H), 7.74 (d, J = 8.4 Hz, 1H), 7.68 - 7.54 (m, 3H), 7.54 - 7.41 (m, 3H), 7.08 (d, J = 8.6 Hz, 1H), 7.01 (d, J = 7.0 Hz, 1H), 6.68 (s, 1H), 5.10 - 4.93 (m, 1H), 4.26 (t, J = 7.0 Hz, 2H), 4.05 (t, J = 6.9 Hz, 2H), 3.63 - 3.43 (m, 2H), 2.96 - 2.76 (m, 3H), 2.66 - 2.56 (m, 4H), 2.50 (2H, under DMSO), 2.07 - 1.87 (m, 1H), 1.85 - 1.64 (m, 4H), 1.52 - 1.32 (m, 1H), 1.25 - 1.05 (m, 2H), 0.92 - 0.65 (m, 2H). Example 18. Preparation of 4-((l-(l-(6-(4-(6-(4-Chlorophei methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)cyclopropyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 16) Scheme 233

Brought 2-(2,6-dioxopiperidin-3-yl)-4-((l-ethynylcyclopropyl)amino)isoindoline-l,3- dione 2,2,2-trifluoroacetate (98-1) (33 mg, 0.0731 1 mmol) , 8-(l-(6-azidohexyl)-lH-pyrazol-4- yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (7-2) (38.4 mg, 0.0731 1 mmol) and N-ethyl-N-isopropylpropan-2-amine (38.1 ,

0.2193 mmol) up in THF (731 µΙ_, ) . Added copper(I) iodide (6.96 mg, 0.03655 mmol) and stirred O/N at r.t.. Concentrated and purified by prep HPLC 5-99% ACNAVater w TFA to give 4-((l-(l- (6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)cyclopropyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 16) (44.0 mg, 0.0509 mmol, 69.7%) as a yellow solid. LCMS (ES+): m/z= 864 [M + H]+ lH MR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 8.20 (s, 1H), 7.98 - 7.85 (m, 3H), 7.74 (d, J = 8.4 Hz, 1H), 7.58 (d, J = 8.6 Hz, 2H), 7.55 - 7.40 (m, 4H), 7.24 (s, 1H), 7.16 - 6.97 (m, 2H), 5.06 (dd, J = 12.7, 5.5 Hz, 1H), 4.20 (t, J = 7.0 Hz, 2H), 4.02 (t, J = 6.9 Hz, 2H), 2.95 - 2.79 (m, 1H), 2.63 - 2.60 (m, 4H), 2.07 - 1.91 (m, 1H), 1.69 (s, 5H), 1.41 (s, 3H), 1.27 (d, J = 3.7 Hz, 2H), 1.23 - 1.09 (m, 4H), 0.79 (dd, J = 19.0, 8.5 Hz, 2H).

Example 19. Preparation of 4-(((l-(6-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4- yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 17) Scheme 234

Brought (S)-8-( 1-(6-azidohexyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (99-1) (20 mg, 0.03890 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (12.1 mg, 0.0389 mmol) up in thf (389 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (13.4 , 0.0778 mmol) followed by copper(I) iodide (3.70 mg, 0.01945 mmol). After stirring the reaction O/N, concentrated it and redissolved in DMSO to purifiy on prep HPLC 5-95% ACN/water + 0 .1% TFA to give 4-(((l-(6-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)hexyl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 17) (15.0 mg, 0.01817 mmol, 46.7 %) as a yellow solid. LCMS (ES+): m/z= 826 [M + H]+ 1H MR (500 MHz, DMSO-de) δ 11.07 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.96 - 7.86 (m, 2H), 7.77 (d, J = 8.5 Hz, 1H), 7.63 - 7.50 (m, 4H), 7.46 (d, J = 8.6 Hz, 2H), 7.12 (d, J = 8.6 Hz, 1H), 7 .10 - 6.97 (m, 2H), 5.03 (dd, J = 12.8, 5.4 Hz, 1H), 4.56 (d, J = 5.3 Hz, 2H), 4.33 - 4.19 (m, 3H), 4.03 (t, J = 7.0 Hz, 2H), 2.94 - 2.79 (m, 1H), 2.60 - 2.55 (m, 4H), 2.03 - 1.96 (m, 1H) 1.85 (d, J = 6.6 Hz, 3H), 1.79 - 1.65 (m, 5H), 1.27 - 1.12 (m, 4H). ok

Example 20. Preparation of 4-(((l-(6-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4- yl)methyl)(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 18) Scheme 235 Brought (S)-8-( 1-(6-azidohexyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (99-1) (20 mg, 0.03890 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(methyl(prop-2-yn-l-yl)amino)isoindoline-l,3-dione (6-1) (12.6 mg, 0.0389 mmol) up inthf(389 L ) and added N-ethyl-N-isopropylpropan-2-amine (13.4 L , 0.0778 mmol) followed by copper(I) iodide (3.70 mg, 0.01945 mmol). Stirred at r.t. O/N and concentrated before redissolving in DMSO and purifying by prep HPLC 5-95% ACN/water + 0 .1% TFA to give 4-(((l-(6-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)hexyl)- IH- 1,2,3 -triazol-4-yl)methyl)(methyl)amino)-2- (2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 18) (20.0 mg, 0.02382 mmol, 61.3 %) as a yellow solid.

LCMS (ES+): m/z= 840 [M + H]+ IH MR (400 MHz, DMSO-de) δ 11.09 (s, IH), 8.24 (s, IH), 8.04 - 7.85 (m, 2H), 7.85 - 7.72 (m, IH), 7.67 - 7.54 (m, 4H), 7.50 - 7.41 (m, 2H), 7.26 (dd, J = 11.0, 7.7 Hz, 2H), 5.10 (dd, J = 12.7, 5.4 Hz, IH), 4.82 - 4.65 (m, 2H), 4.33 - 4.19 (m, 3H), 4.03 (t, J = 7.0 Hz, 2H), 2.94 (s, 3H), 2.92 - 2.80 (m, IH), 2.69 - 2.54 (m, 4H), 2.10 - 1.95 (m, IH), 1.85 (d, J = 6.6 Hz, 3H), 1.79 - 1.64 (m, 5H), 1.30 - 1.05 (m, 4H).

Example 21. Preparation of 4-(((l-(10-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepin-8-yl)- IH-pyr azol-l-yl)decyl)- IH- 1,2,3-triazol-4- yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 19) Scheme 236 Brought (S)-8-( 1-(1 0-azidodecyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (100-1) (14 mg, 0.02455 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (7.64 mg, 0.02455 mmol) up in THF (491 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (4.27 µ , 0.02455 mmol) followed by copper(I) iodide (2.33 mg, 0.01227 mmol) and stirred O/N. In the morning concentrated and redissolved in dmso before purifying by prep HPLC 1-99% ACN/water + 0.1% TFA to give 4-(((l-(10-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)decyl)- 1H-1,2,3 -triazol-4-yl)methyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 19) (16.5 mg, 0.01871 mmol, 76.3 %) as a yellow solid. LCMS (ES+): m/z= 882 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.20 (s, 1H), 7.97 - 7.87 (m, 3H), 7.80 - 7.67 (m, 2H), 7.55 - 7.48 (m, 3H), 7.48 - 7.36 (m, 2H), 7.07 (d, J = 7.4 Hz, 1H), 7.06 - 6.93 (m, 2H), 4.99 (dd, J = 12.9, 5.4 Hz, 1H), 4.29 - 4.14 (m, 3H), 4.06 - 3.93 (m, 2H), 2.89 - 2.74 (m, 1H), 2.57 - 2.47 (m, 4H), 2.00 - 1.92 (m, 1H), 1.80 (d, J = 6.6 Hz, 3H), 1.75 - 1.58 (m, 4H), 1.26 - 1.03 (m, 14H). Example 22. Preparation of 4-((3-(l-(4-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepin-8-yl)- IH-pyr azol-l-yl)butyl)- 1H-1,2,3-triazol-4- yl)propyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 20) Scheme 237

Brought (S)-8-(l-(4-azidobutyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (101-1) (22 mg, 0.04527 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(pent-4-yn-l-ylamino)isoindoline-l,3-dione (9-2) (23.0 mg, 0.06790 mmol) up in thf (452 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (15.7 µ , 0.09054 mmol) followed by copper(I) iodide (4.30 mg, 0.02263 mmol). Stirred reaction O/N and concentrated before redissolving in DMSO and purifying on prep HPLC 1-99% ACN/water + 0.1% TFA. Isolated 4-((3-(l-(4-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)butyl)-lH-l,2,3-triazol-4- yl)propyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 20) (17.0 mg, 0.02059 mmol, 45.5 %) as a yellow solid after lyophilization of the fractions. LCMS (ES+): m/z= 826 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.26 (s, 1H), 8.00 - 7.91 (m, 2H), 7.91 - 7.85 (m, 1H), 7.84 - 7.75 (m, 2H), 7.62 - 7.53 (m, 3H), 7.53 - 7.44 (m, 2H), 7.12 - 7.05 (m, 1H), 7.05 - 6.96 (m, 1H), 6.64 (s, 1H), 5.04 (dd, J = 12.8, 5.5 Hz, 1H), 4.38 - 4.25 (m, 3H), 4.15 - 4.09 (m, 2H), 3.35 (br, 2H), 3.00 - 2.73 (m, 1H), 2.68 (t, J = 7.5 Hz, 2H), 2.64 - 2.57 (m, 4H), 2.07 - 2.00 (m, 1H), 1.96 - 1.82 (m, 5H), 1.82 - 1.67 (m, 4H).

Example 23. Preparation of 4-(((l-(3-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo [f] [1,2,4]triazolo [4,3-a] [1,4] diazepin-8-yl)- IH-pyr azol-l-yl)propyl)- 1H-1,2,3-triazol-4- yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 21) Scheme 238

Degronimer 2 1 Brought (S)-8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (102-1) (24 mg, 0.05085 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (15.8 mg, 0.05085 mmol) up in THF (1.0 mL). Added N-ethyl-N-isopropylpropan-2-amine (8.87 , 0.05085 mmol) followed by copper(I) iodide (4.84 mg, 0.02542 mmol) and stirred O/N at r.t. Concentrated off THF and brought up in 1mL DMSO. Filtered solution. Purified by prep HPLC 1-99% ACN/Water w tfa to give 4-(((l-(3-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H-benzo[fJ[l,2,4]triazolo[4,3- a][1,4]diazepin-8-yl)- lH-pyrazol- 1-yl)propyl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 21) (14.0 mg, 0.018 mmol, 35.0%) LCMS (ES+): m/z= 784 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.11 (s, 1H), 8.26 (d, J = 0.8 Hz, 1H), 8.04 (s, 1H), 8.02 - 7.92 (m, 2H), 7.80 (d, J = 8.5 Hz, 1H), 7.62 - 7.54 (m, 3H), 7.54 - 7.44 (m, 2H), 7.16 (d, J = 8.6 Hz, 1H), 7.10 - 6.96 (m, 2H), 5.06 (dd, J = 12.8, 5.4 Hz, 1H), 4.58 (d, J = 4.7 Hz, 2H), 4.34 (t, J = 7.0 Hz, 2H), 4.27 (q, J = 6.6 Hz, 1H), 4.12 (t, J = 6.8 Hz, 2H), 2.97 - 2.80 (m, 1H), 2.62 - 2.57 (m, 4H), 2.33 (m, 2H), 2.04 (m, 1H), 1.87 (d, J = 6.7 Hz, 3H).

Example 24. Preparation of 4-(((l-(2-(2-(2-(2-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l- yl)ethoxy)ethoxy)ethoxy)ethyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin- 3-yl)isoindoline-l,3-dione (Degronimer 22) Scheme 239

Brought (S)-8-(l-(2-(2-(2-(2-azidoethoxy)ethoxy)ethoxy)ethyl)-lH-pyrazol-4-yl)-6-(4- chlorophenyl)-l,4-dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (104-1) (14 mg, 0.02372 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione

(5-1) (7.38 mg, 0.02372 mmol) up in thf (237 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine

(4.12 µΐ , 0.02372 mmol) followed by copper(I) iodide (2.25 mg, 0.01 186 mmol) . Stirred reaction O/N and concentrated before dissolving in dmso and purifying by prep HPLC 1-99% ACN/water w TFA to give 4-(((l-(2-(2-(2-(2-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)ethoxy)ethoxy)ethoxy)ethyl)- lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 22) (3.5 mg, 16.4 %%) as a yellow solid. LCMS (ES+): m/z= 902 [M + H]+

1H MR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 8.22 (s, 1H), 7.99 - 7.91 (m, 3H), 7.88 - 7.71 (m, 2H), 7.62 - 7.53 (m, 3H), 7.53 - 7.42 (m, 2H) 7.16 - 7.13 (m, 1H), 7.06 - 7.10 (m, 2H), 5.05 (dd, J = 12.8, 5.4 Hz, 1H), 4.56 (d, J = 5.5 Hz, 2H), 4.44 (t, J = 5.2 Hz, 2H), 4.31 - 4.17 (m, 3H), 3.85 - 3.69 (m, 4H), 3.64 - 3.31 (m, 8H, under DMSO), 2.93 - 2.85 (m, 2H), 2.70 - 2.53 (m, 4H), 2.06 - 1.98 (m, 1H), 1.87 (dd, J = 6.7, 2.2 Hz, 3H).

Example 25. Preparation of 4-((l-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)-lH-l,2,3-triazol-4-yl)methoxy)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 23) Scheme 240 Degroriimer 23 Brought 8-(l-(3-azidopropyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropane] (95-1) (15.0 mg, 0.031 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-yloxy)isoindoline-l,3-dione (4-1) (9.7 mg, 0.031 mmol) and N-ethyl-N-isopropylpropan-2-amine (5.4 uL, 0.031 mmol) up in THF (0.5 mL). copper(I) iodide (3.0 mg, 0.015) and stirred at r.t. for 3-4 hrs. Added additional alkyne 7.0 mg. Stirred O/N. Concentrated, redissolved in DMSO (1.0 mL), filtered and purified by MS directed Prep HPLC 1-99% CH3CN/water w TFA. Isolated 4-((l-(3-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)- lH-pyrazol- 1-yl)propyl)- 1H- 1,2,3 -triazol-4-yl)methoxy)-2-(2,6-dioxopiperidin-3 - yl)isoindoline-l,3-dione (Degronimer 23) (18.0 mg, 0.023 mmol, 73.1%) as a whitish solid after lyophilization. LCMS (ES+): m/z= Ί 9Ί [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 8.22 (s, 1H), 8.18 (s, 1H), 7.95 - 7.85 (m, 2H),

7.77 (dd, J = 8.5, 7.2 Hz, 1H), 7.68 (dd, J = 11. 1, 8.5 Hz, 2H), 7.57 - 7.49 (m, 2H), 7.46 - 7.38 (m, 4H), 5.34 (s, 2H), 5.00 (dd, J = 12.7, 5.4 Hz, 1H), 4.33 (t, J = 7.0 Hz, 2H), 4.08 (t, J = 6.9 Hz, 2H), 2.90 - 2.74 (m, 1H), 2.59 - 2.51 (m, 4H), 2.38 - 2.23 (m, 2H), 2.02 - 1.88 (m, 1H), 1.73 - 1.60 (m, 1H), 1.33 - 1.29 (m, 1H), 0.84 - 0.65 (m, 2H). Example 26. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methoxy)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 24) Scheme 241

Brought 8-(l-(6-azidohexyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo [f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (98-1) (0.020 g, 0.03802 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-yloxy)isoindoline-l,3-dione (4-1)

( 11.8 mg, 0.03802 mmol) and N-ethyl-N-isopropylpropan-2-amine (6.61 µ , 0.03802 mmol) up in THF (0.5 mL) at r.t.. Added copper(I) iodide (3.62 mg, 0.01901 mmol) and stirred for 3 hours. Azide and product by lcms, no alkyne remaining. Added 7 mg more alkyne and stirred O/N. Now complete by lcms. Concnetrated reaction, dissolved in ~ 1.0 mL DMSO and filtered. Purified directly with Prep HPLC, 1-99% CftCN/Water w TFA to give 4-((l-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- l-yl)hexyl)-lH-l,2,34riazol-4-yl)methoxy)-^ (Degronimer 24) (26.0 mg, 0.038 mmol, 87.1%) as a white solid. LCMS (ES+): m/z= 839 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 8.18 (s, 1H), 8.15 (d, J = 0.8 Hz, 1H), 7.88 (dd, J = 8.5, 2.1 Hz, 1H), 7.83 (d, J = 0.8 Hz, 1H), 7.76 (dd, J = 8.5, 7.2 Hz, 1H), 7.67 (dd, J = 11.4, 8.5 Hz, 2H), 7.58 - 7.50 (m, 2H), 7.47 - 7.36 (m, 4H), 5.34 (s, 2H), 5.00 (dd, J = 12.7, 5.4 Hz, 1H), 4.28 (t, J = 7.0 Hz, 2H), 3.99 (t, J = 7.0 Hz, 2H), 2.89 - 2.73 (m, 1H), 2.62 - 2.52 (m, 4H), 1.95 - 1.87 (m, 1H), 1.82 - 1.59 (m, 5H), 1.40 - 1.29 (m, 1H), 1.23 - 1.13 (m, 4H), 0.83 - 0.65 (m, 2H).

Example 27. Preparation of 4-((3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)propyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 25) Scheme 242

Degror¾imer 25

Brought 3-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)propan-l-amine (105-1) (14 mg, 0.03057 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (12.6 mg, 0.04585 mmol) and N-ethyl-N-isopropylpropan-2-amine (10.6 , 0.061 14 mmol) up in DMACQ.O mL) and heated to 90°C O/N. Filtered solution and purified directly by Prep HPLC 1-99% CFBCN/water w TFA to give 4-((3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 25) ( 11.0 mg, 0.015 mmol, 50.4%) as a yellow solid. LCMS (ES+): m/z= 715 [M + H]+ lHNMR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 8.19 (s, 1H), 7.93 - 7.83 (m, 2H), 7.68 (d, J = 8.5 Hz, 1H), 7.59 - 7.49 (m, 3H), 7.49 - 7.37 (m, 4H), 6.96 (t, J = 7.8 Hz, 2H), 6.64 - 6.57 (m, 1H), 4.98 (dd, J = 12.7, 5.4 Hz, 2H), 4.13 (t, J = 6.8 Hz, 2H), 3.25 (d, J = 6.2 Hz, 2H), 2.93 - 2.75 (m, 1H), 2.54 (s, 3H), 2.49 (m, 1H), 2.1 1 - 1.84 (m, 3H), 1.70 - 1.63 (m, 1H), 1.41 - 1.25 (m, 1H), 0.82 - 0.64 (m, 2H).

Example 28. Preparation of 4-((6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione 1 (Degronimer 26) Scheme 243 Degronimer 26 Brought 6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexan-l-amine (106-1) (4 mg, 0.007999 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (3.75 mg, 0.01359 mmol) and N-ethyl-N-isopropylpropan-2-amine (2.77 µ , 0.01599 mmol) up in DMAC (0.5 mL) and heated to 95°C O/N. Filtered reaction and purified by prep HPLC 1-99% CFBCN/Water w TFA to give 4-((6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 26) (3.0 mg, 0.004 mmol, 50.0%) as a yellow solid. LCMS (ES+): m/z= Ί 5Ί [M + H]+ 1H NMR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 8.16 (d, J = 0.9 Hz, 1H), 7.87 (dd, J = 8.5, 2.1 Hz, 1H), 7.83 (d, J = 0.8 Hz, 1H), 7.68 (d, J = 8.5 Hz, 1H), 7.59 - 7.49 (m, 3H), 7.49 - 7.33 (m, 4H), 7.07 - 6.84 (m, 2H), 6.45 (s, 1H), 4.97 (dd, J = 12.8, 5.4 Hz, 1H), 4.02 (t, J = 7.0 Hz, 2H), 3.19 (d, J = 6.4 Hz, 2H), 2.85 - 2.78 (m, 1H), 2.58 - 2.51 (m, 4H), 2.09 - 1.86 (m, 1H), 1.78 - 1.62 (m, 3H), 1.57 - 1.44 (m, 2H), 1.44 - 1.24 (m, 3H), 1.24 - 1.13 (m, 3H), 0.85 - 0.61 (m, 2H). Example 29. Preparation of 4-((3-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)propyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 27) Scheme 244

Brought (S)-3-(4-(6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[fJ [1,2,4]triazolo[4,3 - a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)propan-l-amine (107-1) (0.02 g, 0.04484 mmol) , 2-(2,6- dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (18.5 mg, 0.06726 mmol) and N-ethyl- N-isopropylpropan-2-amine (7.82 µ , 0.04484 mmol) up in DMAC (500 uL) and heated vial to 95°C. In the morning purified mixture by prep HPLC 1-99% ACN/Water w TFA to give 4-((3- (4-((S)-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- lH-pyrazol-l-yl)propyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 27) (12.8 mg, 0.018 mmol, 40.7%) as a yellow solid. LCMS (ES+): m/z= 703 [M + H]+ 1HNMR (500 MHz, DMSO-de) δ 11.09 (s, 1H), 8.31 - 8.25 (m, 1H), 8.01 - 7.92 (m, 2H), 7.84 - 7.74 (m, 2H), 7.60 - 7.52 (m, 4H), 7.52 - 7.43 (m, 2H), 7.08 - 6.96 (m, 2H), 6.65 (s, 1H), 5.04 (dd, J = 12.9, 5.4 Hz, 1H), 4.27 (q, J = 6.9 Hz, 1H), 4.20 (t, J = 6.9 Hz, 2H), 3.33 (d, J = 6.2 Hz, 2H), 2.88 (s, 1H), 2.62 - 2.55 (m, 4H), 2.09 (dd, J = 13.2, 6.4 Hz, 3H), 2.03 - 1.97 (m, 1H), 1.87 (d, J = 6.6 Hz, 3H). Example 30. Preparation of 4-((6-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)hexyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 28) Scheme 245

Degronimer 28 Brought (S)-6-(4-(6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)hexan-l-amine (108-1) (14 mg, 0.02868 mmol) , 2-(2,6- dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) ( 11.8 mg, 0.04301 mmol) and N-ethyl- N-isopropylpropan-2-amine (10.0 µ , 0.05736 mmol) up in DMAC (300 uL) and heated O/N. Filtered mixture and purified by prep HPLC directly 1-99% ACNAVater w TFA. Isolated 4-((6- (4-((S)-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- lH-pyrazol-l-yl)hexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 28) (16.0 mg, 0.022 mmol, 75.1%) as a yellow solid. LCMS (ES+): m/z= 745 [M + H]+ 1H NMR (500 MHz, DMSO-de) δ 11.07 (s, 1H), 8.25 (s, 1H), 7.97 - 7.89 (m, 1H), 7.94 - 7.90 (m, 1H), 7.83 - 7.73 (m, 2H), 7.66 - 7.50 (m, 4H), 7.50 - 7.43 (m, 2H), 7.09 - 6.95 (m, 2H), 6.54 - 6.46 (br, 1H), 5.03 (dd, J = 12.8, 5.4 Hz, 1H), 4.25 (q, J = 6.6 Hz, 1H), 4.07 (t, J = 7.0 Hz, 2H), 3.24 (d, J = 6.4 Hz, 2H), 2.92 - 2.80 (m, 1H), 2.58 (s, 3H), 2.60 - 2.49 (m, 2H), 2.12 - 1.96 (m, 1H), 1.85 (d, J = 6.6 Hz, 3H), 1.82 - 1.72 (m, 2H), 1.58 - 1.48 (m, 2H), 1.39 - 1.29 (m, 2H), 1.29 - 1.21 (m, 2H).

Example 31. Preparation of 4-((10-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)decyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 29) Scheme 246

Brought (S)- 10-(4-(6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)decan-l-amine (109-1) (13 mg, 0.02389 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (9.89 mg, 0.03583 mmol) up in DMAC

(238 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (12.4 , 0.07167 mmol). Heated reaction to 90°C for 24 hours. Cooled reaction and loaded directly on to prep HPLC 5-95% ACN/water + 0.1% TFA to give 4-((10-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- IH-pyrazol- 1-yl)decyl)amino)-2-(2,6- dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 29) (7.00 mg, 0.008746 mmol, 36.6 %) as a yellow solid after lyophilization. LCMS (ES+): m/z= 801 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.20 (s, 1H), 7.94 - 7.83 (m, 1H), 7.79 - 7.67 (m, 1H), 7.56 - 7.47 (m, 4H), 7.47 - 7.38 (m, 2H), 7.01 (d, J = 8.7 Hz, 1H), 6.95 (dd, J = 7.0, 2.8 Hz, 1H), 6.45 (s, 1H), 4.98 (dd, J = 12.9, 5.3 Hz, 1H), 4.19 (q, J = 6.6 Hz, 1H), 4.00 (t, J = 6.9 Hz, 2H), 3.27 - 3.15 (m, 2H), 2.90 - 2.74 (m, 1H), 2.52 (s, 3H), 2.50 - 2.45 (m, 2H), 2.02 - 1.91 (m, 1H), 1.79 (d, J = 6.7 Hz, 3H), 1.73 - 1.66 (m, 2H), 1.53 - 1.43 (m, 2H), 1.30 - 1.07 (m, 10H).

Example 32. Preparation of 4-((2-(2-(2-(2-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l- yl)ethoxy)ethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 30) Scheme 247 Brought (S)-2-(2-(2-(2-(4-(6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin-8-yl)- IH-pyrazol- 1- yl)ethoxy)ethoxy)ethoxy)ethanamine (110-1) (9 mg, 0.01595 mmol) and 2-(2,6-dioxopiperidin-3- yl)-4-fluoroisoindoline-l,3-dione (1-3) (6.60 mg, 0.02392 mmol) up in DMAC (159 ΐ ) and added N-ethyl-N-isopropylpropan-2-amine (5.55 , 0.0319 mmol). Heated to 90°C for 20 hours and cooled before loading directly onto prep HPLC 5-95% ACN/water +0.1% TFA to give 4-((2- (2-(2-(2-(4-((S)-6-(4-chlorophenyl)- 1,4-dimethyl-4H-benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepin- 8-yl)-lH-pyrazol-l-yl)ethoxy)ethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 30) (3.50 mg, 0.004266 mmol, 26.9 %) as a yellow solid. LCMS (ES+): m/z= 821 [M + H]+ 1H NMR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 8.23 (s, 1H), 8.03 - 7.88 (m, 2H), 7.85 - 7.70 (m, 2H), 7.66 - 7.54 (m, 4H), 7.54 - 7.41 (m, 2H), 7.10 (d, J = 8.5 Hz, 1H), 7.02 (d, J = 7.0 Hz, 1H), 6.56 (s, 1H), 5.1 1 - 4.94 (m, 1H), 4.34 - 4.14 (m, 2H), 3.76 (t, J = 5.3 Hz, 2H), 3.56 (t, J = 5.5 Hz, 2H), 3.50 (~ 10H under solvent peak), 2.88 (br, 1H), 2.67 - 2.56 (m, 3H), 2.55 - 2.50 (m, 2H), 2.12 - 1.99 (m, 2H), 1.86 (d, J = 6.5 Hz, 3H).

Example 33. Preparation of 4-((7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 31) Scheme 248

Brought 7-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)heptan- 1-amine (111-2) (5Omg, 0.09726 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (40.2 mg, 0.1458 mmol) and N-ethyl-N-isopropylpropan-2-amine (50.8 µ , 0.2917 mmol) up in MP (972 µΙ_, ) and heated O/N at 95°C in a sealed vial. In the morning filtered sample and purified by prep HPLC directly 1-99% ACN/Water w TFA. After lyophilization isolated 4-((7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 31) (28.0 mg, 0.03635 mmol, 37.3 %). LCMS (ES+): m/z= 111 [M + H]+ lH MR (400 MHz, DMSO-de) δ 11.09 (s, 1H), 8.22 (s, 1H), 7.99 - 7.83 (m, 2H), 7.74 (d, J = 8.5 Hz, 1H), 7.65 - 7.54 (m, 3H), 7.54 - 7.38 (m, 4H), 7.03 (dd, J = 24.9, 7.8 Hz, 2H), 6.51 (s, 1H), 5.03 (dd, J = 12.8, 5.4 Hz, 1H), 4.07 (t, J = 7.0 Hz, 2H), 3.25 (s, 2H), 2.96 - 2.71 (m, 1H), 2.60 (s, 3H), 2.57 - 2.49 (m, 2H), 2.03 - 1.98 (m, 1H), 1.84 - 1.67 (m, 3H), 1.57 - 1.48 (m, 2H), 1.46 - 1.35 (m, 1H), 1.30 (s, 4H), 1.25 - 1.12 (m, 2H), 0.93 - 0.64 (m, 2H).

Example 34. Preparation of 4-((4-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)butyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione Degronimer 32, Scheme 249

Brought 4-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)butan-l-amine (112-1) (157 mg, 332 µηιοΐ) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (137 mg, 498 µ οΐ) and Diisopropylethylamine (128 mg, 996 µιτιοΐ) up in 2-Pyrrolidinone (2 mL ) and heated O/N at 95°C in a sealed vial. In the morning filtered sample and purified by reverse phase flash to give 4-((4- (4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)butyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3- dione (Degronimer 32) (50.0 mg, 68.6 µιηοΐ , 20.7 %) as a yellow solid. LCMS (ES+): m/z= 784 [M + H]+ 1H MR (500 MHz, CDCh) : δ 8.26 (s, 1H), 7.74-7.72 (m, 2H), 7.61-7.55 (m, 3H), 7.48-7.45 (m, 1H), 7.42-7.33 (m, 4H), 7.09 (d, J=7.5Hz, 1H), 6.86 (d, J=8.5,1H), 6.23-6.21 (m, 1H), 4.92- 4.89 (m, 1H), 4.21-4.18 (m, 2H), 3.31-3.27 (m, 2H), 2.90-2.86 (m, 1H), 2.80-2.72 (m, 2H), 2.67 (s, 3H), 2.13-2.1 1 (m, 1H), 2.05-2.00 (m, 3H), 1.70-1.62 (m, 2H), 1.60-1.51 (m, 1H) , 0.88-0.75 (m, 2H).

Example 35. Preparation of 4-((9-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)nonyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 33) Scheme 250

Degronimer 33 To a solution of 9-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[fJ[l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)nonan-l-amine (114-1) (75 mg, 138 µηιοΐ) in M P(3 mL) was added Diisopropylethylamine (35.6 mg, 276 µηιοΐ) and 2-(2,6- dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (53.3 mg, 193 µιηοΐ) . The mixture was stirred at 90 °C overnight. The mixture was purified with combi-flash to give 4-((9-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)nonyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 33) (20.0 mg, 25.0 µιηοΐ , 18.1 %). LCMS (ES+): m/z= 798 [M + H]+ 1H MR (500 MHz, MeOD) δ 8.19 (s, IH), 7.75-7.73 (m, 2H), 7.58-7.56 (m, 3H), 7.50-7.47 (m, IH), 7.41-7.34 (m, 4H), 7.08 (d, J= 7.0Hz, IH), 7.08 (d, J= 7.0Hz, IH), 6.86 (d, J= 8.5Hz, IH), 4.91-4.88 (m, IH), 4.14-4.1 1 (m, 2H), 3.25-3.23 (m, 2H), 2.9-2.71 (m, 3H), 2.67 (s, 3H), 2.1 1- 1.86 (m, 4H), 1.4-1.25 (m, 13H), 0.91-0.87 (m, 2H).

Example 36. Preparation of 4-((8-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)octyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 34) Scheme 251

Brought 8-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)octan-l-amine (118-1) (100 mg, 189 µηιοΐ) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (78.1 mg, 283 µηιοΐ) and Diisopropylethylamine (73.2 mg, 567 µ οΐ) up in MP (2 ml) and heated O/N at 95°C in a sealed vial. In the morning filtered sample and purified by reverse phase flash to give 4-((8-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)octyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 34) (31.0 mg, 39.5 µιηοΐ , 20.9 %) as a yellow solid. LCMS (ES+): m/z= 784 [M + H]+ IH MR (400 MHz, DMSO-de) δ 11.09 (s, IH), 8.22 (s, IH), 7.94 (dd, J=2.0Hz, 6.0 Hz, IH), 7.89 (s, IH), 7.74 (d, J=8.5Hz, IH), 7.59-7.54 (m, 3H), 7.49-7.44 (m, 2H), 7.07 (d, J=8.5Hz, IH), 7.00 (d, J=7.5Hz, IH), 6.53-6.50 (m, IH), 5.06 (dd, J=5.5Hz, 7.0 Hz, IH), 4.08 (t, J=7Hz, 2H), 3.30-3.24 (m, 2H), 2.91-2.84 (m, IH), 2.59 (s, 3H), 2.58-2.56 (br,lH), 2.03-2.0 (m, IH), 1.78-1.70 (m, 3H), 1.56-1.53 (m, 2H), 1.41-1.36 (m, IH), 1.34-1.14 (m, 10H), 0.85-0.72 (m, 2H).

Example 37. Preparation of 4-((5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 35) Scheme 252 Brought 5-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentan-l-amine (119-1) ( 110 mg, 226

µηιοΐ) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (88.44 mg, 320 µηιοΐ) and

Diisopropylethylamine (82.9 mg, 0.64174 mmol) up in MP (200 µΙ_, ) and heated O/N at 95°C in a sealed vial. Then the mixture was purified by prep-HPLC to afford 4-((5-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)pentyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 35) (30mg 17.9 %) as yellow solid. LCMS (ES+): m/z= 742 [M + H]+ 1H MR (500 MHz, DMSO-de) δ 11.09 (s, 1H), 8.29 (s, 1H), 7.94-7.90 (m, 2H), 7.75-7.73 (d, J=10Hz ,1H), 7.60-7.53 (m,3H), 7.49-7.48 (m,2H), 7.44 (d, J=1.5Hz, 1H), 7.07-6.99 (m, 2H), 6.54- 6.51 (m,lH), 5.75(s, 1H), 5.05-5.01 (m, 1H), 4.1 1 (t, J=6.5Hz, 2H), 3.32-3.24 (m, 2H), 2.90-2.83 (m, 1H), 2.59 (s,3H), 2.03-1.99 (m, 1H), 1.83-1.78 (m,2H), 1.75-1.70(m,lH), 1.59-1.55 (m,2H), 1.41-1.37 (m,lH), 1.37-1.23 (m,3H), 0.82-0.74 (m, 2H).

Example 38. Preparation of 4-((2-(2-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 36) Scheme 253 Degronimer 36 ' C Brought 2-(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)ethoxy)ethanamine 2,2,2- trifluoroacetate (121-1) (100 mg, 0.1661 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4- fluoroisoindoline-l,3-dione (1-3) (68.8 mg, 0.2491 mmol) up in DMSO (1.66 mL ) and added N- ethyl-N-isopropylpropan-2-amine (86.7 L, 0.4983 mmol). Heated reaction to 95°C O/N. cooled reaction and loaded directly onto RP isco 10-100% ACNAVater w TFA to give 4-((2-(2-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 36) (20.4 mg, 0.02741 mmol, 16.5 %) as a yellow solid after lyophilization. LCMS (ES+): m/z= 746 [M + H]+ 1H NMR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.17 (s, 1H), 7.88 (d, J = 7.3 Hz, 2H), 7.71 (d, J = 8.5 Hz, 1H), 7.57 (d, J = 8.6 Hz, 2H), 7.54 - 7.36 (m, 4H), 7.06 (d, J = 8.7 Hz, 1H), 6.97 (d, J = 7.0 Hz, 1H), 6.55 (s, 1H), 5.02 (dd, J = 12.9, 5.4 Hz, 1H), 4.24 (t, J = 5.4 Hz, 2H), 3.79 (t, J = 5.4 Hz, 2H), 3.55 (t, J = 5.4 Hz, 1H), 3.55 (t, J = 5.4 Hz, 1H), 3.41 (br, 2H), 2.87 (m, 1H), 2.60 (s, 4H), 2.08 - 1.88 (m, 1H), 1.79 - 1.65 (m, 1H), 1.51 - 1.30 (m, 1H), 0.90 - 0.65 (m, 2H). Example 39. Preparation of 4-((2-(2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethoxy)ethyl)(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 37) Scheme 254

Brought 2-(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4 ,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethoxy)-N-methylethanamine 2,2,2- trifluoroacetate (124-1) (76 mg, 0.1233 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4- fluoroisoindoline-l,3-dione (1-3) (51.0 mg, 0.1849 mmol) up in DMSO (1.23 mL ) and added N - ethyl-N-isopropylpropan-2-amine (64.4 , 0.3699 mmol) . Heated reaction to 95°C O/N and then loaded fractions directly onto 50g RP isco column 10-100% CH3CN/Water w TFA to give 4-((2-

(2-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)ethoxy)ethyl)(methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 37) (27.8 mg, 0.03666 mmol, 29.7 % ) after lyophilization as a yellow solid. LCMS (ES+): m/z= 759 [M + H ]+ l H M R (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.07 (s, 1H), 7.98 - 7.83 (m, 2H), 7.74 (d, J = 8.5

Hz, 1H), 7.66 - 7.53 (m, 2H), 7.53 - 7.36 (m, 4H), 7.13 (dd, J = 9.6, 7.8 Hz, 2H), 5.04 (dd, J = 12.9, 5.4 Hz, 1H), 4.16 (t, J = 5.3 Hz, 2H), 3.69 (t, J = 5.2 Hz, 1H), 2.90 (s, 3H), 2.89 - 2.86 (m, 1H), 2.62 (s, 3H), 2.60 - 2.55 (m, 2H),1.99 - 1.96 (m, 1H), 1.78 - 1.65 (m, 1H), 1.52 - 1.33 (m, 1H), 0.98 - 0.59 (m, 2H).

Example 40. Preparation of 4-((2-(4-(2-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)ethyl)piperazin-l-yl)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3- dione (Degronimer 38) Scheme 255

To a solution of 2-(4-(2-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)ethyl)piperazin- 1-yl)ethanamine with TFA salt (128-1) (150 mg, 223 µιηοΐ) in DMSO (6.0 mL ) in a seal tube, DIPEA (270 , 1.56 mmol) was added followed by the addition of 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3- dione (1-3) (36.7 mg, 133 µιηοΐ) at rt. The reaction was heated to 90 °C for 16h. The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was quenched with water(20.0 mL) and extracted with ethyl acetate(20x3 mL).The organic layers were dried over sodium sulfate,filtered and concentrated to afford crude.The crude product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-10% ) to give 4-((2-(4-(2-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)ethyl)piperazin-l-yl)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 38) (45.0 mg, 55.3 µηιοΐ , 24.8 %) as light yellow solid. LCMS (ES+): m/z= 2 [M + H]+ IH MR (400 MHz, DMSO- d6 + drops of TFA-d) δ 0.78-0.80 (m, 2H), 0.80-0.88 (m, 1H), 0.96- 1.06 (m, 2H), 1.18-1.26 (m, 6H), 1.42-1.58 (m, 2H), 1.73-1.81 (m, 1H), 1.98-2.06 (m, 1H), 2.81 (s, 3H), 2.82-2.90 (m, 1H), 3.40-3.50 (m, 2H), 3.68-3.79 (m, 4H), 4.55-4.62 (m, 2H), 5.04 (dd, J = 12.8 Hz & 5.2 Hz, 1H), 7.09 (d, J = 6.8 Hz, 1H), 7.16 (d, J = 8.4 Hz, 1H), 7.45 (d, J = 8.8 Hz, 2H), 7.51 (d, J = 1.6 Hz, 1H), 7.55-7.62 (m, 3H), 7.84 (d, J = 8.8 Hz, 1H), 8.01-8.05 (m, 2H), 8.26 (s, 1H).

Example 41. Preparation of 3-(4-((4-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)methyl)-l-oxo-l,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2- yl)-3-methylpiperidine-2,6-dione (Degronimer 39) Scheme 256 Brought 3-methyl-3-(l-oxo-4-(piperazin-l-ylmethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (40-1) (125 mg, 0.2651 mmol) and 6-(4- chlorophenyl)-8-(l-(6-iodohexyl)-lH-pyrazol-4-yl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropane] (131-1) (115 mg, 0.1893 mmol) up in DMF (946 µΐ ) and added N-ethyl-N-isopropylpropan-2-amine (131 L, 0.7574 mmol) and heated overnight at 60°C. No RI remaining in morning. Loaded crude reaction directly onto a lOOg reversed phase isco and purified with a 10-100% ACN/water w TFA gradient. Isolated 3-(4-((4-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperazin- 1-yl)methyl)- 1-oxo- lH-pyrrolo[3 ,4-c]pyridin-2(3H)-yl)-3 - methylpiperidine-2,6-dione (Degronimer 39) (55.0 mg, 0.06544 mmol, 34.5 %) as an off-white solid. LCMS (ES+): m/z= 841 [M + H]+ lHNMR (300 MHz, DMSO-de) δ 10.96 (s, 1H), 8.68 (d, J = 5.0 Hz, 1H), 8.23 (d, J = 2.3 Hz, 1H), 8.04 - 7.87 (m, 2H), 7.75 (d, J = 8.1 Hz, 1H), 7.66 - 7.55 (m, 3H), 7.55 - 7.41 (m, 3H), 4.86 - 4.78 (m, 1H), 4.15 - 4.04 (m, 2H), 4.03 - 3.93 (m, 2H), 3.56 - 3.30 (m, 1H), 2.87 - 2.66 (m, OH), 2.65 - 2.54 (m, 3H), 2.00 - 1.85 (m, 1H), 1.74 (s, 4H), 1.65 - 1.54 (m, 2H), 1.48 - 1.37 (m, 1H), 1.34 - 1.18 (m, 6H), 0.90 - 0.69 (m, 2H).

Example 42. Preparation of 3-(4-(2-(l-(5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)piperidin-4-yl)ethyl)-l-oxo-l,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2- yl)piperidine-2,6-dione (Degronimer 40) heme 257

Degronimer 40 Brought 3-(l-oxo-4-(2-(piperidin-4-yl)ethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (90 mg, 0.1913 mmol) and 6-(4-chlorophenyl)-8- (l-(5-iodopentyl)-lH-pyrazol-4-yl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-

4,l'-cyclopropane] (136 mg, 0.2295 mmol) up in DMF (956 µL· ) and added N-ethyl-N- isopropylpropan-2-amine (133 , 0.7652 mmol) . Heated the reaction to 60°C O/N. Purified directly by reversed phase Isco 10-100% ACN/water with 0.1% TFA to give 3-(4-(2-(l-(5-(4-(6- (4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]- 8-yl)-lH-pyrazol-l-yl)pentyl)piperidin-4-yl)ethyl)-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione (Degronimer 40) (85.0 mg, 0.1029 mmol, 54.1 %) as its TFA salt. LCMS (ES+): m/z= 826 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.02 (s, 1H), 9.09 - 8.90 (m, 1H), 8.66 (d, J = 5.0 Hz, 1H), 8.24 (s, 1H), 7.97 - 7.84 (m, 2H), 7.73 (d, J = 8.5 Hz, 1H), 7.57 (td, J = 5.3, 4.1, 1.8 Hz, 3H), 7.53 - 7.41 (m, 3H), 5.16 (dd, J = 13.3, 5.1 Hz, 1H), 4.66 - 4.51 (m, 1H), 4.42 (d, J = 17.7 Hz, 1H), 4.09 (t, J = 6.9 Hz, 2H), 3.43 (d, J = 11.9 Hz, 2H), 3.01 - 2.91 (m, 2H), 2.91 - 2.75 (m, 4H), 2.69 - 2.56 (m, 4H), 2.47 - 2.27 (m, 1H), 2.03 (dd, J = 12.2, 5.9 Hz, 1H), 1.97 - 1.88 (m, 2H), 1.88 - 1.46 (m, 8H), 1.44 - 1.30 (m, 2H), 1.30 - 1.13 (m, 2H), 0.93 - 0.63 (m, 2H).

Example 43. Preparation of 3-(4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperidin-4-yl)methyl)-l-oxo-l,3-dihydro-2H-pyrrolo[3,4-c]pyridin-2- yl)piperidine-2,6-dione (Degronimer 41) Scheme 258 Brought 6-(4-chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (131.1) (66.8 mg, 0.1095 mmol) and 3-(l-oxo-4-(piperidin-4-ylmethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (36-1) (25 mg, 0.05477 mmol) up in DMF (547 µL· ) before adding N-ethyl-N-isopropylpropan-2-amine (38.1 , 0.2190 mmol). Heated reaction to

50°C O/N. After cooling reaction to r.t. loaded mixture directly onto reversed phase Isco 10-100%

ACN/Water w 0.1% TFA. After lyophilization gave 3-(4-((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperidin-4-yl)methyl)-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (Degronimer 41) (30.0 mg, 0.03634 mmol, 66.3 % ) as a white solid. LCMS (ES+): m/z= 826 [M + H ]+

1H NMR (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.68 (s, 1H), 8.20 (s, 1H), 7.96 - 7.81 (m, 2H),

7.72 (d, J = 8.5 Hz, 1H), 7.63 - 7.53 (m, 2H), 7.52 - 7.35 (m, 3H), 5.17 (dd, J = 13.2, 5.1 Hz, 1H),

4.55 (d, J = 17.8 Hz, 1H), 4.40 (d, J = 17.7 Hz, 1H), 4.07 (t, J = 6.8 Hz, 2H), 3.00 - 2.75 (m, 6H),

2.70 - 2.61 (m, 2H), 2.58 (s, 3H), 2.40 - 2.37 (m, 1H), 2.18 - 2.10 (m, 1H), 2.08 - 2.01 (m, 1H),

1.88 - 1.64 (m, 5H), 1.60 - 1.49 (m, 2H), 1.47 - 1.32 (m, 3H), 1.32 - 1.10 (m, 5H), 0.84 - 0.62

(m, 2H).

Example 44. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f] [l,2,4]triazolo[4,3-a] [l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)azetidin-3-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 42) Scheme 259 Brought 6-(4-chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (131-1) (180 mg, 0.2946 mmol) and 4-(azetidin-3-ylamino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione 2,2,2- trifluoroacetate (16-2) (143 mg, 0.3240 mmol) up in M P (982 ) and added N-ethyl-N- isopropylpropan-2-amine (153 , 0.8838 mmol) before heating reaction to 60°C. Stirred reaction at elevated temp for 24 hrs before cooling to r.t. and purified directly by reversed phase isco 10- 100% ACN/water + 0.1% TFA. Lyophilization of the pure fractions gave the TFA salt of 4-((l-

(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)azetidin-3-yl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 42) (100 mg, 0.1232 mmol, 41.8 % ) as a yellow solid. LCMS (ES+): m/z= 8 11 [M + H ]+

1H NMR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 9.75 (br, 1H), 8.21 (d, J = 1.5 Hz, 1H), 8.01 -

7.86 (m, 2H), 7.74 (d, J = 8.5 Hz, 1H), 7.68 - 7.57 (m, 2H), 7.53 - 7.41 (m, 2H), 7.21 - 7.13 (m,

1.5H), 7.10 (t, J = 7.9 Hz, 1H), 6.97 - 6.93 (d, 0.5H), 5.14 - 5.00 (m, 1H), 4.62 (m, 1H), 4.49 (t, J

= 8.3 Hz, 2H), 4.38 - 4.18 (m, 2H), 4.10 - 4.06 (m, 4H), 3.17 (d, J = 6.3 Hz, 2H), 2.99 - 2.81 (m, lH), 2.59 (m, 4H), 2.13 - 1.97 (m, 1H), 1.84 - 1.68 (m, 3H), 1.42 (s, 3H), 1.24 (s, 3H), 0.89 - 0.64 (m, 2H).

Example 45. Preparation of 3-(4-((4-(6-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)methyl)-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione (Degronimer 43) heme 260

Brought 3-(l-oxo-4-(piperazin-l-ylmethyl)-lH-pyrrolo[3,4-c]pyridin-2(3H)- yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (40-1) (86.4 mg, 0.189 mmol) , 6-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)hexanal (136-1) (78.5 mg, 0.1575 mmol) , sodium acetate (25.8 mg, 0.315 mmol) and acetic acid (0.315 mmol) up in DCM (1.57 mL ) . Added 4A mol sieves and stirrred at r.t. for 3 hours at which time added sodium triacetoxy borohydride (43.3 mg, 0.2047 mmol). Continued stirring o/n. Quenched with sodium bicarbonate solution and extracted into DCM 2x. dried over sodium sulfate and concentrated to an oil. Purified by reversed phase isco 10-100% ACN/water with TFA. After lyophilization, the TFA salt of 3-(4-((4-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperazin-l-yl)methyl)-l-oxo-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione (Degronimer 43) (60.0 mg, 0.07260 mmol, 46.1 % ) was isolated as as an off-white solid. LCMS (ES+): m/z= 827 [M + H ]+ 1H M R (400 MHz, CD30D) δ 8.72 - 8.71 (m, 1H), 8.04 (s, 1H), 8.00 - 7.97 (m,lH), 7.85 (s,

1H), 7.78 - 7.72 (m, 2H), 7.60 (dd, J = 8.7, 2.2 Hz, 2H), 7.52 (s, 1H), 7.45 (dd, J = 8.6, 2.1 Hz,

2H), 5.24 - 5.20 (m, 1H) 4.76 - 4.66 (m, 1H), 4.17 (t, J = 7.0 Hz, 2H), 4.02 - 3.85 (m, 2H), 3.42

(m, 2H), 3.14 - 3.09 (m, 3H) 2.99 - 2.80 (m, 3H), 2.76 (s, 3H), 2.60 - 2.56 (m, 3H), 1.91 - 1.83

(m, 3H), 1.77 - 1.49 (m, 3H), 1.39 (s, 4H), 0.91 - 0.87 (m, 2H).

Example 46. Preparation of 3-(4-(5-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)pentyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 44) Scheme 261

Brought 3-(piperazin-l-yl)piperidine-2,6-dione hydrochloride (49-2) (36.1 mg, 0.1546 mmol) and 5-(4-(6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentanal (137-1) (0.075 g, 0.1546 mmol) up in DCM (1.54 mL ) and added sodium acetate (19.0 mg, 0.2319 mmol)/ acetic acid (13.9 mg, 0.2319 mmol) . Stirred at r.t. for 20 minutes and then added sodium triacetoxyborohydride (49.1 mg, 0.2319 mmol) . Continued stirring O/N at r.t., complete by LCMS in AM. Quenched reaction with sat. soldium bicarbonate solution and extracted 3 x with DCM. dried combined organic layer over sodium sulfate and concentrated purified by reversed phase isco 10-100% ACN/water w TFA to give 3-(4-(5-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine- 4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 44) (35.9 mg, 0.05277 mmol, 34.1 %) as a white solid after lyophilization. LCMS (ES+): m/z= 681 [M + H]+ lHNMR (400 MHz, DMSO-de) δ 10.96 (s, 1H), 8.22 (s, 1H), 7.99 - 7.88 (m, 2H), 7.74 (d, J = 8.4 Hz, 1H), 7.59 (d, J = 8.6 Hz, 2H), 7.56 - 7.48 (m, 2H), 7.44 (d, J = 2.1 Hz, 1H), 5.03 (s, 1H), 4.1 1 (t, J = 6.8 Hz, 2H), 4.05 - 3.82 (m, 2H), 3.80 - 3.24 (m, 5H), 3.20 - 3.06 (m, 2H), 2.88 - 2.72 (m, 1H), 2.60 (s, 4H), 2.40 - 2.23 (m, 1H), 1.93 - 1.57 (m, 5H), 1.48 - 1.33 (m, 1H), 1.30 - 1.16 (m, 2H), 0.92 - 0.63 (m, 2H).

Example 47. Preparation of 3-(4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 45) Scheme 262 Brought 1-(5-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)pentyl)-lH-l,2,3-triazole-4- carbaldehyde (139-1) (65.3 mg, 0.1 155 mmol), 3-(piperazin-l-yl)piperidine-2,6-dione hydrochloride (49-2) (27 mg, 0.1 155 mmol) and sodium acetate (14.2 mg, 0.1732 mmol) up in DCM (1.15 mL )/acetic acid (10.4 mg, 0.1732 mmol) and stirred for an hour before adding sodium triacetoxyborohydride (36.7 mg, 0.1732 mmol) and continuing stirring at r.t. overnight. Diluted with DCM and extracted with sodium bicarbonate. Dried the organic layer over sodium sulfate and concentrated. Dissolved the crude material in DMSO and purified by reversed phase isco 10- 100% ACN/water + 0 .1% TFA and isolated the TFA salt of 3-(4-((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 45) (61.5 mg, 0.08078 mmol, 69.9 %) after lyophilization. LCMS (ES+): m/z= 775 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 10.95 (s, 1H), 8.23 (d, J = 5.0 Hz, 2H), 8.04 - 7.83 (m, 2H), 7.74 (d, J = 8.5 Hz, 1H), 7.66 - 7.55 (m, 2H), 7.55 - 7.38 (m, 3H), 4.99 (s, 1H), 4.36 (t, J = 7.1 Hz, 4H), 4.06 (t, J = 7.0 Hz, 2H), 3.92 - 3.65 (m, 1H), 3.47 - 3.30 (m, 4H), 2.86 - 2.69 (m, 1H), 2.60 (s, 4H), 2.30 - 2.25 (m, 1H), 1.88 - 1.67 (m, 6H), 1.50 - 1.35 (m, 1H), 1.35 - 1.16 (m, 4H), 0.93 - 0.65 (m, 2H). Example 49. Preparation of 3-(4-(6-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione(Degronimer 47) Scheme 264

Brought 3-(piperazin-l-yl)piperidine-2,6-dione hydrochloride (23.3 mg, 0.1001 mmol) and 6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexanal (0.05 g, 0.1001 mmol) up in DCM (1.00 mL ) and added sodium acetate (12.3 mg, 0.1501 mmol) / acetic acid (9.01 mg, 0.1501 mmol) . Stirred at r.t. for 20 minutes and then added sodium triacetoxyborohydride (31.8 mg, 0.1501 mmol) . Continued stirring O/N at r.t., complete by LCMS in AM. Quenched reaction with sat. soldium bicarbonate solution and extracted 3 x with DCM. Dried combined organic layer over sodium sulfate and concentrated. Purified by reversed phase isco 10-100% ACN/water w TFA to give 3- (4-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 47) (35.9 mg, 0.05277 mmol, 52.7 %) as a white solid TFA salt. LCMS (ES+): m/z= 694 [M + H]+ l H MR (400 MHz, DMSO-de) δ 10.85 (s, 1H), 8.23 (s, 1H), 7.99 - 7.86 (m, 2H), 7.74 (d, J = 8.5 Hz, 1H), 7.67 - 7.56 (m, 2H), 7.54 - 7.39 (m, 3H), 4.91 (s, 1H), 4.07 (t, J = 6.9 Hz, 2H), 3.28 - 3.09 (m, 2H), 2.99 (q, J = 16.5 Hz, 2H), 2.85 - 2.62 (m, 1H), 2.58 (s, 2H), 2.37 - 2.21 (m, 3H), 1.91 - 1.61 (m, 4H), 1.50 - 1.34 (m, 3H), 1.34 - 1.13 (m, 4H), 0.97 - 0.60 (m, 2H).

Example 50. Preparation of 3-(7-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (Degronimer 48) Scheme 265 Brought 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (44 mg, 0.076 mmol ) and 3-(7-amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (28-3) (28.2 mg, 0.09860 mmol) up in DCE (758 µL· ) and added acetic acid (5 uL , 0.07585 mmol) followed by 4 A MS and sodium triacetoxy borohydride (16.0 mg, 0.07585 mmol) after 20 minutes. Stirred at r.t. O/N. Quenched with sat. NaHC03 solution and extracted into CHC13. concentrated and purified by RP Isco 10-100 % ACN/water + TFA, loaded in DMSO. After lyophilization recovered 3-(7-(((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine- 2,6-dione (Degronimer 48) (17.5 mg, 0.021 mmol, 27.1 %) as an off-white solid. LCMS (ES+): m/z= 5 \ [M + H]+ 1H MR (400 MHz, DMSO-de) δ 10.91 (s, 1H), 8.19 (s, 1H), 7.97 (s, 1H), 7.96 - 7.93 (m, 1H), 7.88 (s, 1H), 7.73 (d, J = 8.8 Hz, 1H), 7.66 (d, J = 8.8 Hz, 1H), 7.61 - 7.55 (m, 2H), 7.47 (d, J = 8.2 Hz, 2H), 7.44 (s, 1H), 7.1 1 (t, J = 5.7 Hz, 1H), 6.79 (dd, J = 8.8, 2.2 Hz, 1H), 6.55 (d, J = 2.3 Hz, 1H), 5.10 (dd, J = 11.5, 5.7 Hz, 1H), 4.37 (d, J = 5.6 Hz, 2H), 4.27 (t, J = 6.9 Hz, 2H), 4.03 (t, J = 6.9 Hz, 2H), 2.82 - 2.61 (m, 1H), 2.58 (s, 3H), 2.51 (s, 3H), 2.31 (br, 1H), 2.09 - 2.06 (m, 2H), 1.77 - 1.45 (m, 4H), 1.41 - 1.35 (m, 1H), 1.26 - 1.16 (m, 4H), 0.87 - 0.66 (m, 2H).

Example 51. Preparation of 3-(6-((( l-(6 -(4-(6-(4-Chlorophenyl)-l - methylspiro[benzo[f][l,2 ,4]triazolo[4,3-a][l ,4]diazepine-4,l'-cyclopropan]-8-yl )-l H- pyrazol-l-yl)hexyl )-l H-l,2,3-triazol -4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (Degronimer 49) Scheme 266 Brought 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (0.06 g, 0.1034 mmol) and 3-(6-amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (27-3) (44.4 mg, 0.1551 mmol) up in dee (1.03 mL ) / acetic acid (24.8 , 0.4136 mmol) and added 4 A MS. After 20-40 minutes at r.t. added sodium triacetoxy borohydride (28.4 mg, 0.1344 mmol) and stirred at r.t. O/N. Quenched reaction with saturated bicarbonate solution and extracted with DCM 3x. dried combined organic over sodium sulfate and concentrated. Purified by reversed phase isco 10-100% ACN/water w tfa to give 3-(6-(((l-(6-(4- (6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4- oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (Degronimer 49) (40.0 mg, 0.047 mmol, 45.5%) as an off-white solid after lyophilization. LCMS (ES+): m/z= 851 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 10.97 (s, 1H), 8.19 (s, 1H), 7.93 (d, J = 6.4 Hz, 2H), 7.88 (s, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.63 - 7.55 (m, 2H), 7.51 - 7.41 (m, 3H), 7.37 (dd, J = 8.8, 2.2 Hz, 2H), 7.19 (dd, J = 8.8, 2.7 Hz, 1H), 7.03 (d, J = 2.7 Hz, 1H), 5.18 (dd, J = 11.5, 5.7 Hz, 1H), 4.40 (br, 2H), 4.27 (t, J = 7.0 Hz, 2H), 4.03 (t, J = 7.1 Hz, 2H), 2.85 - 2.77 (m, 1H), 2.60 (s, 4H), 2.55 (s, 4H), 2.19 - 2.00 (m, 1H), 1.81 - 1.62 (m, 5H), 1.42 - 1.36 (m, 1H), 1.27 - 1.10 (m, 4H), 0.91 - 0.68 (m, 2H).

Example 52. Preparation of 3-(5-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-l-oxoisoindolin-2-yl)piperidine- 2,6-dione 1091-58, Degronimer 50 Scheme 267 Degr i i er 50 Brought 3-(5-amino-l-oxoisoindolin-2-yl)piperidine-2,6-dione (23-3) (33.4 mg, 0.1292 mmol) and 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (50 mg, 0.08619 mmol) up in DCE (861 µΐ ) . Added acetic acid (9.80 µΐ , 0.1723 mmol) and 4A MS and stirred for 1 hour before adding (acetyloxy)(sodio)boranyl acetate; acetic acid (23.7 mg, 0.1 120 mmol) . Stirred at r.t. O/N and then quenched with saturated sodium bicarbonate before extracting with DCM 3x. Dried organic layers over sodium sulfate and concentrated. Purified by isco MeOH/DCM 1-10% to give 3-(5-(((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione

(Degronimer 50) (13.5 mg, 0.01639 mmol, 19.0 %) as a whitish solid. LCMS (ES+): m/z= 823 [M + H]+ lH MR (400 MHz, DMSO-de) δ 10.92 (s, 1H), 8.21 (s, 1H), 8.07 - 7.83 (m, 3H), 7.74 (d, J = 8.5 Hz, 1H), 7.63 - 7.55 (m, 2H), 7.51 - 7.43 (m, 3H), 7.40 - 7.30 (m, 1H), 6.71 (s, 2H), 5.00 (dd, J = 13.3, 5.1 Hz, 1H), 4.41 - 4.18 (m, 5H), 4.18 - 3.96 (m, 3H), 3.03 - 2.78 (m, 1H), 2.61 (s, 4H), 2.38 - 2.25 (m, 1H), 1.99 - 1.86 (m, OH), 1.74 - 1.61 (m, 4H), 1.49 - 1.31 (m, 1H), 1.31 - 1.12 (m, 4H), 0.91 - 0.66 (m, 2H).

Example 53. Preparation of 3-(8-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (Degronimer 51) Scheme 268

Brought 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (55 mg, 0.09481 mmol) and 3-(8-amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (26-3) (37.9 mg, 0.1327 mmol) up in DCE (948 µL· ) and added acetic acid (70 , 1.16 mmol) . Also added a scoop of activated 4A MS. Stirred at r.t. for 2 hours. Added sodium triacetoxy borohydride (30.1 mg, 0.1422 mmol) and stirred O/N. Almost complete by lcms. worked up anyway partitioned between DCM and sat bicarb. Washed aq with 2x DCM. Dried combined organics over sodium sulfate and then concentrated. Purified by reversed phase isco 5-100% ACN/Water w TFA. Isolated 3-(8-(((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)-yl)piperidine- 2,6-dione (Degronimer 51) (33.0 mg, 0.03880 mmol, 40.9 %) as a whitish solid. LCMS (ES+): m/z= 850 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.01 (s, 1H), 8.20 (s, 1H), 7.98 - 7.91 (m, 2H), 7.89 (s, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.62 - 7.55 (m, 2H), 7.53 - 7.43 (m, 3H), 7.19 (t, J = 7.8 Hz, 1H), 7.12 (d, J = 7.7 Hz, 1H), 6.86 (d, J = 7.5 Hz, 1H), 5.22 (dd, J = 11.5, 5.5 Hz, 1H), 4.48 (s, 2H), 4.25 (t, J = 6.9 Hz, 2H), 4.02 (t, J = 7.1 Hz, 2H), 2.89 - 2.74 (m, 1H), 2.63 (s, 4H), 2.60 (s, 4H), 2.21 - 2.04 (m, 1H), 1.79 - 1.64 (m, 5H), 1.48 - 1.34 (m, 1H), 1.27 - 1.10 (m, 4H), 0.87 - 0.69 (m, 2H).

Example 54. Preparation of 3-(5-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (Degronimer 52) Scheme 269 Brought 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (0.060 g, 0.1034 mmol) , 3-(5-amino-2-methyl-4-oxoquinazolin-3(4H)- yl)piperidine-2,6-dione (25-4) (38.4 mg, 0.1344 mmol) up in DCE (1.03 mL ) / acetic acid (0.1034 mmol) and added 4A mol. sieves. Stirred at r.t. for 1 hour and then added sodium triacetoxy borohydride (30.6 mg, 0.1447 mmol) continued stirring at r.t. o/n. Quenched reaction with sodium bicarb solution and extracted into DCM. Purified by prep hplc 5-95% ACN/water w tfa to give 3- (5-(((l -(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,1 '-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)-2-methyl- 4-oxoquinazolin-3(4H)-yl)piperidine-2,6-dione (Degronimer 52) (21.5 mg, 0.02528 mmol, 24.4 %) after lyophilization as a white solid. LCMS (ES+): m/z= 850 [M + H]+ 1H NMR (300 MHz, DMSO-de) δ 11.01 (s, 1H), 8.22 (d, J = 2.0 Hz, 1H), 8.08 - 7.87 (m, 3H), 7.76 (d, J = 8.1 Hz, 1H), 7.67 - 7.56 (m, 2H), 7.55 - 7.42 (m, 4H), 6.67 (t, J = 9.3 Hz, 2H), 5.28 - 5.17 (m, 1H), 4.56 - 4.41 (m, 2H), 4.37 - 4.23 (m, 3H), 4.12 - 3.99 (m, 2H), 2.98 - 2.76 (m, 1H), 2.71 - 2.56 (m, 8H), 2.23 - 2.10 (m, 1H), 1.87 - 1.66 (m, 6H), 1.52 - 1.35 (m, 1H), 0.93 - 0.71 (m, 2H). Example 55. Preparation of 3-(4-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)-l-oxoisoindolin-2-yl)piperidine- 2,6-dione (Degronimer 53) heme 270

Brought 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l (138-2) (35 mg, 0.06033 mmol) and 3-(4-amino-l-oxoisoindolin-2- yl)piperidine-2,6-dione (29-1) (23.4 mg, 0.09049 mmol) up in DCE (603 ΐ )/ acetic acid (60 uL, 0.9991 mmol) and added a small scoop of powdered 4A activated sieves. Stirred at r.t. for 1-2 hours and added sodium triacetoxyborohydride (25.5 mg, 0.1206 mmol) . Stirred o/n at r.t. and quenched w sat sodium bicarb solution. Extracted w DCM and dried combined organic over sodium sulfate. Purified by prep HPLC 5-95% ACN/water w 0.1% TFA to give 3-(4-(((l-(6-(4- (6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)- 1- oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 53) (20.0 mg, 0.02429 mmol, 40.3 %) as an off-white solid after lyophilization. LCMS (ES+): m/z= 823 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.00 (s, 1H), 8.21 (d, J = 2.4 Hz, 1H), 7.96 - 7.91 (m, 2H), 7.89 (s, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.63 - 7.54 (m, 3H), 7.54 - 7.41 (m, 4H), 7.23 (t, J = 7.7 Hz, 1H), 6.92 (d, J = 7.4 Hz, 1H), 6.78 (d, J = 8.1 Hz, 1H), 5.09 (dd, J = 13.4, 5.1 Hz, 1H), 4.45 - 4.35 (m, 2H), 4.30 - 4.19 (m, 3H), 4.18 - 3.99 (m, 3H), 3.01 - 2.82 (m, 1H), 2.70 - 2.56 (m, 4H),

2.34 - 2.23 (m, 1H), 2.07 - 1.95 (m, OH), 1.88 - 1.78 (m, 1H), 1.76 - 1.65 (m, 5H), 1.47 - 1.34

(m, 1H), 1.30 - 1.1 1 (m, 5H), 0.91 - 0.70 (m, 2H).

Example 56. Preparation of N-(2-((7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)amino)ethyl)-l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxamide (Degronimer 54) Scheme 271

Degronimer 54 Brought 8-(l-(7-chloroheptyl)-lH-pyrazol-4-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (142-2) (122 mg,

0.2300 mmol), potassium iodide (172 mg, 1.04 mmol) and N-(2-aminoethyl)-l-(2,6- dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3-carboxamide 2,2,2-trifluoroacetate (140-2)

(0.085 g, 0.2091 mmol) up in CftCN (836 ΐ ). Added N-ethyl-N-isopropylpropan-2-amine (145

, 0.8364 mmol) and began heating at 70°C. Stirred at elevated t em p for 2 days before cooling to r.t. and purifying by reversed phase isco 10-100% ACN/water + 0.1% TFA. Lyophilized the pure fractions to givethe TFA salt of N-(2-((7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl)amino)ethyl)- 1-(2,6-dioxopiperidin-3 -yl)-6-oxo- 1,6-dihydropyridine-3 -carboxamide

(Degronimer 54) (25.0 mg, 0.03167 mmol, 15.1 % ) a s a whitish solid. LCMS (ES+): m/z= 790

[M + Na]+

1H M R (400 MHz, DMSO-de) δ 11.09 (s, 1H), 8.53 - 8.40 (m, 1H), 8.36 - 8.25 (m, 2H), 8.21

(s, 1H), 7.96 - 7.84 (m, 3H), 7.74 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.6 Hz, 2H), 7.52 - 7.46 (m,

2H), 7.43 (d, J = 2.0 Hz, 1H), 6.50 (d, J = 9.6 Hz, 1H), 4.07 (t, J = 7.0 Hz, 2H), 3.48 (d, J = 5.9 Hz,

1H), 3.12 - 2.97 (m, 1H), 2.95 - 2.72 (m, 1H), 2.59 (s, 4H), 2.12 - 1.97 (m, 1H), 1.84 - 1.67 (m,

3H), 1.65 - 1.46 (m, 1H), 1.44 - 1.35 (m, 1H), 1.32 - 1.15 (m, 6H), 0.91 - 0.69 (m, 2H) *missing

4 protons under large water peak.

Example 57. Preparation of 3-(4-(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 55) Scheme 272 Brought 6-(4-chlorophenyl)-8-(l-(7-iodoheptyl)-lH-pyrazol-4-yl)-l- methylspiro[benzo[fJ[l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (131-1) (120 mg, 0.1920 mmol) , 3-(2-oxopiperazin-l-yl)piperidine-2,6-dione hydrochloride (49-2) (66.5 mg, 0.2688 mmol) and N-ethyl-N-isopropylpropan-2-amine (100 µ , 0.576 mmol) up in DMF (640 ) and heated to 60°C overnight. Cooled to r.t. and purified directly by reversed phase isco 10 - 100% ACN/Water + 0.1% TFA to give the TFA salt of 3-(4-(7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl)-2-oxopiperazin-l-yl)piperidine-2,6-dione (Degronimer 55) (30.0 mg, 0.04235 mmol, 22.2 %) as a white solid after lyophilization. LCMS (ES+): m/z= 708 [M + H]+ 1HNMR (400 MHz, DMSO-de) δ 10.94 (s, 1H), 8.20 (s, 1H), 7.92 (dd, J = 8.5, 2.1 Hz, 1H), 7.88 (s, 1H), 7.73 (d, J = 8.5 Hz, 1H), 7.61 - 7.54 (m, 2H), 7.51 - 7.45 (m, 2H), 7.43 (d, J = 2.1 Hz, 1H), 5.02 (s, 1H), 4.07 (t, J = 7.0 Hz, 2H), 3.57 - 3.46 (m, 1H), 3.17 - 3.05 (m, 2H), 2.87 - 2.71 (m, 1H), 2.59 (s, 4H), 2.43 - 2.24 (m, 1H), 1.88 - 1.81 (m, 1H), 1.78 - 1.70 (m, 3H), 1.66 - 1.57 (m, 3H), 1.44 - 1.35 (m, 1H), 1.35 - 1.14 (m, 10H), 0.86 - 0.66 (m, 2H). Example 58. Preparation of 4-(((l-(8-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct-7-yn-l- yl)-lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 56) Scheme 273

145-1 5-1 Degronimer 56

Brought 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (12.8 mg, 0.04121 mmol) and 8-(8-azidooct-l-yn-l-yl)-6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (145-1) (19 mg,

0.03925 mmol) up in THF (392 µΙ_, ) and added N-ethyl-N-isopropylpropan-2-amine (13.6 µ ,

0.0785 mmol) followed by copper(I) iodide (3.73 mg, 0.01962 mmol) . Stirred under A r O/N at r.t. Product and two high M W byproducts seen in UPLC. Concentrated and purified by prep HPLC

(1-99% ACN/water w TFA) to give 4-(((l-(8-(6-(4-chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct-7-yn-l-yl)-

lH-l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 56) (7.0 mg, 0.009 mmol, 22.4 % ) as a yellow solid after lyophilization. LCMS

(ES+): m/z= 796 [M + H ]+

l H M R (400 MHz, DMSO-de) δ 11.10 (s, 1H), 7.99 (s, 1H), 7.72 (d, J = 1.7 Hz, 2H), 7.60 - 7.42

(m, 5H), 7.21 (s, 1H), 7.13 (d, J = 8.6 Hz, 1H), 7.10 - 6.97 (m, 2H), 5.05 (dd, J = 12.9, 5.3 Hz,

1H), 4.57 (s, 2H), 4.42 (t, J = 7.0 Hz, 1H), 4.28 (t, J = 7.0 Hz, 2H), 2.94 - 2.80 (m, 1H), 2.58 (s, 4H), 2.36 (q, J = 7.0, 5.9 Hz, 2H), 2.07 - 1.93 (m, 1H), 1.82 - 1.63 (m, 3H), 1.52 - 1.30 (m, 5H), 1.25 - 1.13 (m, 2H), 0.79 (t, J = 8.8 Hz, 2H).

Example 59. Preparation of 3-(4-((4-(6-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)methyl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 57) Scheme 274

To a solution of 6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl methanesulfonate (147-1) (140 mg, 241 µηιοΐ), 3-(l-oxo-4-(piperazin-l-ylmethyl)isoindolin-2-yl)piperidine-2,6-dione trifluoroacetate (14-3) (98.9 mg, 289 µιηοΐ) in ACN (3 mL ) under nitrogen atmosphere, added DIPEA (38.9 mg, 301 µιηοΐ) and sealed the cap. stirred the reaction at 80°C for 18 h.The reaction progress was monitored by TLC and LCMS.The RM was quenched with water (lOmL) and extracted with ethyl acetate (10mLx3) .Combined organic layers and dried over anhydrous sodium sulfate.The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-15%) to give 3-(4-((4-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperazin- 1-yl)methyl)- 1- oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 57) (45.0 mg, 54.5 µη οΐ , 22.7 %) as white solid. LCMS (ES+): m/z= 825 [M + H]+ ¾ MR (400 MHz, DMSO- d6) δ 0.73-0.78 (m, 2H), 1.22-1.27 (m, 7H), 1.37-1.40 (m, 2H), 1.55- 2.00 (m, 6H), 2.35-2.42 (m, 2H), 2.52-2.55 (m, 2H), 2.58 (s, 3H), 2.92-3.33 (m, 5H), 3.61 (bs, 2H), 4.06-4.07(m, 2H), 4.39 (d, J = 18.2 Hz, 1H), 4.46 (d, J = 18.2 Hz, 1H), 5.15 (dd, J = 13.6 Hz & 3.6 Hz, 1H), 7.47-7.57 (m, 5H), 7.58 (d, J= 8.0 Hz, 2H), 7.63-7.74 (m, 2H), 7.90-7.94 (m, 2H), 8.22 (s, 1H), 11.01(s, 1H).

Example 60. Preparation of 3-(4-((4-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)propyl)piperazin-l-yl)methyl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione Degronimer 58 Scheme 275 To a solution of 3-(l-oxo-4-(piperazin-l-ylmethyl)isoindolin-2-yl)piperidine-2,6- dione(with TFA salt) (14-3) (171 mg, 502 µηιοΐ) in ACN (3 mL ) at RT under nitrogen atmosphere added 3-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)propyl methanesulfonate (135-1) (180 mg, 335 µιηοΐ) followed by the addition of DIPEA (302 mg, 2.34 mmol) .Heated and stirred the RM at 90°C.The reaction progress was monitored by TLC and LCMS. After consumption of starting material, distilled the reaction mixture directly under reduced pressure. The product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-15%) to give 3-(4-((4-(3-(4- (6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)- lH-pyrazol- 1-yl)propyl)piperazin- 1-yl)methyl)- 1-oxoisoindolin-2- yl)piperidine-2,6-dione (21.0 mg, 26.8 µιηοΐ , 8.01 %) as off white solid. LC/MS (ES+): m/z 783 [M + H]+ H MR (400 MHz, DMSO- d6) δ 0.72-0.83 (m, 2H), 1.15-1.46 (m, 4H), 1.66-1.88 (m, 2H), 1.90-2.07 (m, 2H), 2.57 (s, 3H), 2.58-2.70 (m, 4H), 2.81-3.10 (m, 4H), 3.10-3.21 (m, 2H), 3.58-3.77 (m, 2H), 4.08-4.16 (m, 2H), 4.35 (d, J = 17.2 Hz, 1H), 4.50 (d, J = 16.0 Hz, 1H), 5.13 (d, J = 10.8 Hz, 1H), 7.37-7.55 (m, 4H), 7.55-7.63 (m, 3H), 7.64-7.70 (m, 1H), 7.70-7.78 (m, 1H), 7.88-8.02 (m, 2H), 8.22 (bs, 1H), 11.01 (s, 1H).

Example 61. Preparation of 3-(4-(l-(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)-l,2,3,6-tetrahydropyridin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6- dione (Degronimer 59) Scheme 276 To a solution of 3-(l-oxo-4-(2-oxopiperazin-l-yl)isoindolin-2-yl)piperidine-2,6-dione (12-3) (82.8 mg, 242 µηιοΐ) in Acetonitrile (5 mL ), Ν ,Ν -diisopropylethyl amine (142 mg, 1.41 mmol) was added and stirred for 15 minute at room temperature followed by addition of 7-(4-(6- (4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-

8-yl)-lH-pyrazol-l-yl)heptyl methanesulfonate (146-2) (120 mg, 202 µιηοΐ) and heated the reaction mixture at 80 °C for 16 h, After TLC showed completion , the resulting reaction mixture was quenched into ice water and extracted with EA (2x20 mL) and resulting organic phase was dried over sodium sulfate and concentrate under reduced pressure, the resulting crude material was purified by column chromatography by using 10% MeOH in DCM as eluent to give 3-(4-(l-(7-(4- (6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptyl)-l,2,3,6-tetrahydropyridin-4-yl)-l-oxoisoindolin-2- yl)piperidine-2,6-dione (Degronimer 59) (53.9 mg, 65.5 µπιοΐ , 32.4 %) as white solid. LCMS (ES-): m/z= 820 [M - H]- ¾ MR (400 MHz, DMSO- d6) δ 0.75-0.78 (m, 2H), 1.22-1.28 (m, 10H), 1.31-1.58 (m, 4H), 1.69-1.82 (m, 4H), 1.99-2.0 (m, 1H), 2.36-2.47 (m, 1H), 2.58 (s, 3H), 2.59-2.68 (m, 2H), 2.89- 2.95 (m, 2H), 4.08 (t, J= 8.0 Hz, 2H), 4.37 (d, J = 17.6, Hz, 1H), 4.54 (d, J = 17.2 Hz, 1H), 5.17 (dd, J = 13.6 Hz & 3.6 Hz, 1H), 6.01 (bs, 1H), 7.44 (s, 1H), 7.46-7.58 (m, 3H), 7.59 (d, J = 8.0 Hz, 2H), 7.62-7.67 (m, 1H), 7.74 (d, J= 8.0 Hz, 1H), 7.90 (s, 1H), 7.93 (d, J= 8.0 Hz, 2H), 8.22 (s, 1H), 10.99 (s, 1H). Example 62. Preparation of 3-(6-(((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a ][l,4]diazepine-4 ,l'-cyclopropan]-8-yl )-l H- pyrazol-l-yl)hexyl )-l H-l,2,3-triazol-4-yl)methyl)amino )-l-oxoisoindolin-2-yl)piperidine- 2,6-dione (Degronimer 60) Scheme 277

Degronimer 60 To a solution of l-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)-lH-l,2,3-triazole-4- carbaldehyde (138-2) (60 mg, 103 µηιοΐ) , 3-(6-amino-l-oxoisoindolin-2-yl)piperidine-2,6-dione

(24-3) (39.9 mg, 154 µιηοΐ) in 1,2-Dichloroethane (1.2 mL ) , Acetic acid (98.4 mg, 1.64 mmol) was added. Then the reaction mixture was stirred at rt for 2 h . Then charged Sodium triacetoxyborohydride (43.6 mg, 206 µιηοΐ) at 0 °C and the reaction mixture was stirred at rt for another 16 h . The reaction progress was monitored by TLC and LCMS. The Reaction mixture was quenched with ice water(10.0 mL) and extracted with DCM(20.0 mLx2). The combined organic layers were dried over sodium sulfate,filtered and concentrated to afford crude.The crude compound was purified by reverse phase prep HPLC and dried under Ihypholization to get 3-(6- (((1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)- 1- oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 60) (24.0 mg, 29.1 µηιοΐ , 28.3 %). LCMS (ES+): m/z= 824 [M + H]+

¾ MR (400 MHz, DMSO - ) 0.76-0.81 (m, 2H), 1.20-1.23 (m, 4H), 1.25-1.39 (m, 1H), 1.72- 1.73 (m, 6H), 1.98-2.0 (m, 1H), 2.32-2.33 (m, 1H), 2.59 (s, 3H), 2.65-2.98 (m, 1H), 4.08 (t, J= 8.0Hz, 2H), 4.10-4.14 (m, 2H), 4.26-4.29 (m, 2H), 4.31-4.33 (m, 2H), 5.17 (dd, J = 13.6 Hz & 3.6 Hz, 1H), 6.37 (s, 1H), 6.86-6.90 (m, 1H), 6.91 (d, J= 8.0 Hz, 1H), 7.25 (d, J= 8.0 Hz, 1H), 7.44- 7.45 (m, 1H), 7.48 (d, J = 8.0 Hz, 2H), 7.58 (d, J = 8.0 Hz, 2H), 7.73 (d ,J= 8.0 Η ζ,ΙΗ ), 7.89-7.93 (m, 3H), 8.20 (s, 1H), 10.99 (bs, 1H).

Example 63. Preparation of 3-(4-(l-(7-(4-(6-(4-Chlorophenyl)- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)-4-hydroxypiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 61) Scheme 278

Degronimer 6 1

To a solution of 3-(4-(4-hydroxypiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione with TFA salt (11-1) (91.9 mg, 201 µηιοΐ) in Acetonitrile (4 mL ), DIPEA (233 , 1.34 mmol) was added followed by the addition of 7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl methanesulfonate (146-1) (100.0 mg, 168 µιτιοΐ) at rt. The reaction was heated to 80 °C for 16h. The reaction progress was monitored by TLC and LCMS analysis. The reaction mixture was quenched with water(15.0 mL) and extracted with ethyl acetate(25x3 mL). The organic layers were dried over sodium sulfate,filtered and concentrated to afford crude.The crude product was purified by silica gel flash chromatography (12 g Isco gold, DCM/MeOH 0-15% with 10% methanolic ammonia) to give 3-(4-(l-(7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl)-4-hydroxypiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 61) (30.0 mg, 35.6 µιηοΐ , 21.2 %) as off white solid. LCMS (ES+): m/z= 841 [M + H]+ lHNMR(400 MHz, DMSO- d6) δ 0.73-0.76 (m, 2H), 1.12-1.30 (m, 9H), 1.33-1.42 (m, 4H), 1.68- 1.76 (m, 6H), 1.77-2.21 (m, 4H), 2.28-2.49 (m, 2H), 2.58 (s, 3H), 2.85-2.95 (m, 1H), 4.09-4.12 (m, 2H), 4.58 (d, J = 18.2 Hz, 1H), 4.67 (d, J = 18.2 Hz, 1H), 5.01 (bs, 1H), 5.15 (dd, J = 13.6 Hz & 3.6 Hz, 1H), 7.44-7.50 (m, 4H), 7.55-7.62 (m, 4H), 7.74 (d, J = 8.4 Hz, 1H), 7.90 (s, 1H), 7.96 (dd, J = 8.4 Hz & 2.0 Hz, 1H), 8.23 (s, 1H), 10.99 (s, 1H). ES-MS (m/z): 840.70 (M+H+)

Example 64. Preparation of 3-(4-(l-(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)piperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 62) Scheme 279 O Degronimer 62

To a solution of 3-(l-oxo-4-(piperidin-4-yl)isoindolin-2-yl)piperidine-2,6-dione (13-1) (98.8 mg, 302 µηιοΐ) Acetonitrile (10 mL ), Ν ,Ν -diisopropylethyl amine (227 mg, 1.76 mmol) was added and stirred for 15 minute at room temperature followed by addition of 7-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)heptyl methanesulfonate (146-2) (150 mg, 252 µιηοΐ) and heated the reaction mixture at 80 °C for 16 h, After showed TLC completion , the resulting reaction mixture was quiench into ice water and extract with EA (2x20 mL) and resulting organic phase was dried over sodium sulfate and concentrate under reduced pressure, the resulting crude material was purified by column chromatography by using 10% MeOH in DCM as eluent to give 3-(4-(l-(7-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)heptyl)piperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione

(Degronimer 62) (84.6 mg, 102 µιηοΐ, 40.8 %) as a white solid. LCMS (ES+): m/z= 825 [M + H]+ 1H MR (400 MHz, DMSO- d6) δ 0.74-0.81 (m, 2H), 1.19-1.30 (m, 10H), 1.35-1.42 (m, 2H), 1.68-1.78 (m, 6H), 1.91-2.09 (m, 4H), 2.59 (s, 3H), 2.58-2.64 (m, 2H), 2.85-2.95 (m, 2H), 3.51- 3.61 (m, 1H), 4.09-4.12 (m, 2H), 4.35 (d, J = 18.4 Hz, 1H), 5.05 (d, J = 18.2 Hz, 1H), 5.15 (dd, J = 13.6 Hz & 3.4 Hz, 1H), 7.45 (d, J = 2.0 Hz, 1H), 7.48-7.50 (m, 4H), 7.56-7.63 (m, 3H), 7.74 (d, J = 8.4 Hz, 1H), 7.90 (s, 1H), 7.94 (dd, J = 8.4 Hz & 2.0 Hz, 1H), 8.23 (s, 1H), 11.01 (s, 1H). Example 65. Preparation of 3-(4-((l -(6-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperidin-4-yl)methyl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 63) Scheme 280

Degronimer 63 To a solution of 6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl methanesulfonate (147-1) (0.100 g, 172 µηιοΐ), 3-(l-oxo-4-(piperidin-4-ylmethyl)isoindolin-2-yl)piperidine-2,6-dione with TFA salt (13-1) (70.3 mg, 206 µιηοΐ) in ACN (2 mL ) under nitrogen atmosphere, added DIPEA (22. 1 mg, 172 µ οΐ) and sealed the cap. stirred the reaction at 80°C for 18 h.The reaction progress was monitored by TLC and LCMS.The RM was quenched with water (lOmL) and extracted with ethyl acetate (10mLx3) .Combined organic layers and dried over anhydrous sodium sulfate.The product was purified by silica gel flash chromatography (12 g , DCM/MeOH 0-15%) with 10% methanolic ammonia solution to give 3-(4-((l-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperidin-4-yl)methyl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 63)

(23.0 mg, 27.8 µιηοΐ , 16.3 %) as a white solid. LCMS (ES+): m/z= 824 [M + H]+ lHNMR(400 MHz, DMSO- d6) δ 0.72-0.85 (m, 2H), 1.1 1-1.28 (m, 8H), 1.36-1.40 (m, 4H), 1.40- 1.65 (m, 4H), 1.66-1.83 (m, 4H), 1.95-2.05 (m, 2H), 2.59 (s, 3H), 2.58-2.64 (m, 2H), 2.85-2.95 (m, 2H), 3.48-3.52 (m, 1H), 4.05-4.15 (m, 2H), 4.29 (d, J = 18.2 Hz, 1H), 4.46 (d, J = 18.2 Hz, 1H), 5.15 (dd, J = 13.6 Hz & 3.8 Hz, 1H), 7.41-7.50 (m, 5H), 7.55-7.64 (m, 3H), 7.74 (d, J = 8.4 Hz, 1H), 7.90 (s, 1H), 7.93 (dd, J = 8.4 Hz & 2.0 Hz, 1H), 8.22 (s, 1H), 11.01 (s, 1H).

Example 66. Preparation of 3-(4-(4 -(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)-2-oxopiperazin-l-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 64) Scheme 281

Deg i ei- 64 To a solution of 7-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)heptyl methanesulfonate (146-2)

(0.130 g, 219 µηιοΐ) , 3-(l-oxo-4-(2-oxopiperazin-l-yl)isoindolin-2-yl)piperidine-2,6-dione with TFA salt (49-2) (89.6 mg, 262 µιηοΐ) in ACN (2 mL ) under nitrogen atmosphere, added DIPEA

(197 mg, 1.53 mmol) and sealed the cap. stirred the reaction at 80°C for 18 h.The reaction progress was monitored by TLC and LCMS.The RM was quenched with water (lOmL) and extracted with ethyl acetate (10mLx3) .Combined organic layers and dried over anhydrous sodium sulfate. The product was purified by silica gel flash chromatography (12 g , DCM/MeOH 0-15%) to give 3-(4- (4-(7-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)- lH-pyrazol- 1-yl)heptyl)-2-oxopiperazin- 1-yl)- 1-oxoisoindolin-2- yl)piperidine-2,6-dione (Degronimer 64) (14.0 mg, 16.6 µπιοΐ , 7.65 %) as a white solid. LCMS (ES+): m/z= 839 [M + H]+ lHNMR(400 MHz, DMSO- d6) δ 0.73-0.85 (m, 2H), 1.15-1.27 (m, 8H), 1.28-1. 50 (m, 2H), 1.68- 1.81 (m, 4H), 1.91-2.07 (m, 2H), 2.59 (s, 3H), 2.68-2.80 (m, 2H), 2.82-2.97 (m, 2H), 3.1 1-3.25 (m, 2H), 3.58-3.72 (m, 2H), 4.05-4.12 (m, 2H), 4.14-4.40 (m, 2H), 5.08-5.19 (m, 1H), 7.45 (d, J = 1.6 Hz, 1H), 7.49 (d, J = 8.4 Hz, 2H), 7.56-7.64 (m, 4H), 7.68-7.77 (m, 2H), 7.90 (s, 1H), 7.94 (dd, J = 8.4 Hz & 2.0 Hz, 1H), 8.23 (s, 1H), 11.01 (s, 1H).

Example 67. Preparation of 3-(2-((4-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)methyl)-5-oxo-5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6- yl)piperidine-2,6-dione (Degronimer 65) Scheme 282 Degronimer 85 To a stirred solution of 3-(5-oxo-2-(piperazin-l-ylmethyl)-5H-pyrrolo[3,4-b]pyridin-

6(7H)-yl)piperidine-2,6-dione with TFA salt (51-3) (177 mg, 388 µιηοΐ) in Acetonitrile (10 mL ) drop wise DIEA (138 mg, 1.07 mmol) was added to the reaction mixture after that 6-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)-lH-pyrazol-l-yl)hexyl methanesulfonate (147-1) (148 mg, 257 µιηοΐ) was added and reaction stirred for 16hr at 90°C. The progress of reaction monitored by TLC. The reaction mixture was directly concentrated. The product was purified by using hillic coloumn (4gm, DCM/MEOH 0- 35%) to give 3-(2-((4-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperazin- 1-yl)methyl)-5-oxo-

5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6-dione (Degronimer 65)(32.0 mg, 38.7 µπιοΐ , 14.9 %) as off white solid. LCMS (ES+): m/z= 827 [M + H]+ lHNMR(400 MHz, DMSO- d6) δ 0.78-0.91 (m, 2H), 1.18-1.25 (m, 6H), 1.26-1.37 (m, 3H), 1.73- 1.74 (m, 3H), 1.90-2.02(m,lH),2.18-2.23(m,2H), 2.40-2.50(m, 4H),2.59(s, 3H), 2.88-2.95(m, lH),3.68(br,s, 2H),4.00-4.07(m, 2H), 4.31 (d, J = 17.2 Hz, 1H), 4,47 (d, J = 17.6 Hz, 1H), 5.15 (dd, J = 13.2 Hz & 5.2 Hz, 1H), 7.44-7.49(m, 3H), 7.58(d, J=8.0 Hz,3H),7.73-7.75(d, J=8.0Hz,lH),

7.89(s,lH),7.93(d,J=8.0 Hz,lH),8.10(d,J=8.0 Hz,lH), 8.22(s,lH),11.01( br, s, 1H).

Example 68. Preparation of 3-(2-((4-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperazin-l-yl)methyl)-7-oxo-5,7-dihydro-6H-pyrrolo[3,4-b]pyridin-6- yl)piperidine-2,6-dione (Degronimer 66) Scheme 283

To a solution of 3-(7-oxo-2-(piperazin-l-ylmethyl)-5H-pyrrolo[3,4-b]pyridin-6(7H)- yl)piperidine-2,6-dione(with TFA salt) (52-3) (151 mg, 341 µιηοΐ) , 6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl methanesulfonate (147-1) (180 mg, 310 µιηοΐ) in ACN (2 mL ) at RT under nitrogen atmosphere added DIEA (280 mg, 2.17 mmol) .Heated and stirred the RM at 90°C for 16h.The reaction progress was monitored by TLC and LCMS.After consumption of SM purified it by reverse phase column chromatography using HILLIC 12g ,using 0-70% ACN:0.02%ammonium acetate water as mobile phase to obtain (Degronimer 66) 3-(2-((4-(6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)piperazin-l-yl)methyl)-7-oxo-5H-pyrrolo[3,4-b]pyridin-6(7H)-yl)piperidine-2,6-dione

(15.0 mg, 18.1 µιηοΐ , 5.85 %) as white solid. LCMS (ES+): m/z= 827 [M + H]+ 1H MR (400 MHz, DMSO- d6) δ 0.75 (m, 2H), 1.22-1.37 (m, 8H), 1.73-1.74 (m, 3H), 2.04 (m, 1H), 2.19 (m, 2H), 2.40(m, 5H),2.59(s, 3H),2.6 (m, 4H) ,2.91(m, lH),3.66(s, 2H),4.06(m, 2H),4.31-4.47( dd, 2H), 5.14 (m, 1H), 7.44-7.49(m, 3H), 7.57-7.63(d,3H),7.73-7.75(d, J=8.0Hz,lH),7.89(s,lH),7.93(d,J=8.0 Hz,lH),8.02(d,J=8.0 Hz,lH ), 8.23(s,lH),l 1.02( br, s, 1H).

Example 69. Preparation of 3-(4-(l-(7-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)-4-fluoropiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 67) Scheme 284

In 30 ml sealed vial 3-(4-(4-fluoropiperidin-4-yl)-l-oxoisoindolin-2-yl)piperidine-2,6- dione TFA (50-3) (174 mg, 505 µιηοΐ) add ACN (8 mL ) add N,N-Diisopropylethylamine (261 mg, 2.02 mmol) dropwise then add 7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl methanesulfonate (146-2) (200 mg, 337 µιηοΐ) reflux this stirred solution at 90°C for 16h. Reaction monitored by TLC. After consumption of starting material rm was quenched in water (~5ml) and extract using DCM (~5ml x 3) combined organic layer was dry using anhydrous sodium sulfate, filter it and dry under reduced pressure. The product was purified by silica gel flash chromatography (12 g redisep, DCM/MeOH 0-10%) to give3-(4-(l-(7-(4-(6-(4- chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8- yl)- lH-pyrazol- 1-yl)heptyl)-4-fluoropiperidin-4-yl)- 1-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 67) (7.54 mg, 8.95 µιηοΐ , 2.66 %) as white solid. LCMS (ES+): m/z= 842 [M + H]+ ¾ MR (400 MHz, DMSO- d6) δ 0.73-0.77 (m, 2H), 1.22-1.28 (m, 6H), 1.38-1.42 (m, 3H), 1.43- 1.77 (m, 3H), 1.89-2.01 (m, 4H), 2.01-2.07 (m, 2H), 2.14-2.20(m,3H),2.28-2.32(m,2H),2.58 (s, 3H), 2.78-2.82 (m, 2H), 2.86-2.93(m,lH),4.08 (t,J=8.0Hz, 2H), 4.46 (d, J = 16.8, Hz, 1H), 4.58 (d, J = 18.0 Hz, 1H), 5.12 (dd, J = 12.8 Hz & 5.2 Hz, 1H), 7.44(br,s,lH),7.48(d,J=8.0Hz,2H),7.53(d,J=8.0Hz,lH),7.58(d,J=8.0Hz,3H),7.68(d,J=7.2,lH),7.7 4(d,J=8.4Hz,lH), 7.90 (s, 1H), 7.94 (dd, J = 8.4 Hz & 2.0 Hz, 1H), 8.23 (s, 1H), 10.98 (s, 1H). ES- MS (m/z): 842.40 (M+H +)

Example 70. Preparation of 4-(((l-(7-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-y^ yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 68) Scheme 285 Deg r n me r 68 Brought (S)-8-( 1-(7-azidoheptyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (111-1) (14 mg, 0.02651 mmol) and 2-(2,6- dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (8.25 mg, 0.02651 mmol) up in dioxane (600 uL) and added Cp*RuCl(PPh3)2 (2.1 1 mg, 0.002651 mmol). Heated vial to 60°C O/N. Added 2 mg more Ru and heated for a second night. Concentrated and purified by prep hplc 1-99% ACN/Water w TFA to give 4-(((l-(7-(4-((S)-6-(4-chlorophenyl)-l,4- dimethyl-4H-benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)heptyl)-lH- l,2,3-triazol-5-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 68) (3.80 mg, 0.004527 mmol, 17. 1 %) as a yellow solid after lyophilization. LCMS (ES+): m/z= 840 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.23 (s, 1H), 7.98 - 7.91 (m, 2H), 7.83 - 7.74 (m, 1H), 7.68 - 7.53 (m, 5H), 7.51 - 7.43 (m, 2H), 7.18 (t, J = 6.2 Hz, 1H), 7.13 - 7.04 (m, 2H), 5.05 (dd, J = 12.9, 5.5 Hz, 1H), 4.69 (d, J = 6.1 Hz, 2H), 4.35 (t, J = 7.2 Hz, 2H), 4.25 (q, J = 6.5 Hz, 1H), 4.1 1 - 4.02 (m, 2H), 2.87 (t, J = 15.8 Hz, 1H), 2.57 (s, 4H), 2.05 - 1.97 (m, 1H), 1.85 (d,

J = 6.6 Hz, 3H), 1.80 - 1.63 (m, 5H), 1.31 - 1.1 1 (m, 6H). Example 71. Preparation of 5-(l-(7-(4-((S)-6-(4-Chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-y^ yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 69) Scheme 286

Degronimer 69

Brought (S)-8-( 1-(7-azidoheptyl)- lH-pyrazol-4-yl)-6-(4-chlorophenyl)- 1,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine (111-1) (14 mg, 0.02651 mmol) and 2-(2,6- dioxopiperidin-3-yl)-5-ethynylisoindoline-l,3-dione (3-2) (7.48 mg, 0.02651 mmol) up in dioxane (600 uL). Added N-ethyl-N-isopropylpropan-2-amine (4.60 , 0.02651 mmol) followed by copper(I) iodide (2.52 mg, 0.01325 mmol) and stirred O/N at r.t. Purified reaction directly by prep HPLC 1-99% ACN/Water w TFA to give 5-(l-(7-(4-((S)-6-(4-chlorophenyl)-l,4-dimethyl-4H- benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepin-8-yl)-lH-pyrazol-l-yl)heptyl)-lH-l,2,3-triazol-4-yl)- 2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 69) (5.70 mg, 0.007034 mmol, 26.6 %) as an off-white solid. LCMS (ES+): m/z= 8 11 [M + H]+ 1H MR (400 MHz, DMSO-de) δ 11.15 (s, 1H), 8.93 (s, 1H), 8.39 - 8.31 (m, 2H), 8.25 (s, 1H), 8.05 - 7.88 (m, 3H), 7.76 (d, J = 8.4 Hz, 1H), 7.61 - 7.51 (m, 3H), 7.50 - 7.41 (m, 2H), 5.17 (dd, J = 13.0, 5.4 Hz, 1H), 4.40 (t, J = 7.0 Hz, 2H), 4.24 (q, J = 6.6 Hz, 1H), 4.06 (t, J = 6.9 Hz, 2H), 3.21 - 3.09 (m, 2H), 3.00 - 2.82 (m, 1H), 2.69 - 2.62 (m, 1H), 2.56 (s, 3H), 2.14 - 2.02 (m, OH), 1.85 (d, J = 6.6 Hz, 4H), 1.80 - 1.71 (m, 1H), 1.61 - 1.50 (m, 2H), 1.36 - 1.16 (m, 2H), 0.92 (t, J 7.3 Hz, 3H).

Example 72. Preparation of 5-(4-(7-(4-(6-(4-Chloroph methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)heptyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 70) heme 287

Brought 6-(4-chlorophenyl)- 1-methyl-8-(l -(7-(piperazin- 1-yl)heptyl)- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (80-1) (70 mg, 0.1004 mmol) , 2-(2,6-dioxopiperidin-3-yl)-5-fluoroisoindoline-l,3-dione (2-2) (41.5 mg, 0.1506 mmol) up in DMSO (1.0 mL) and added N-ethyl-N-isopropylpropan-2-amine (52.4 , 0.3012 mmol) . Stirred in a sealed vial O/N and then concentrated. Dissolved crude material in DMSO and purified by reversed phase isco to give 5-(4-(7-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)heptyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 70) (50.0 mg, 0.060 mmol, 59.3 %) as a yellow solid after lyophilization. LCMS (ES+): m/z= 840 [M + H]+

Example 73. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(6-((4-(l-methyl-8-(l-methyl- lH-pyrazol-4-yl)spiro [benzo [f] [l,2,4]triazolo [4,3-a] [1,4]diazepine-4,l '-cyclopropan]-6- yl)phenyl)amino)hexyl)-lH-l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 71) heme 288

Brought N-(6-azidohexyl)-4-(l -methyl-8-(l -methyl- lH-pyrazol -4- yl)spiro[benzo[fJ[l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)aniline (149-1) (0.039 g, 0.07490 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3- dione (5-1) (25.6 mg, 0.08239 mmol) and N-ethyl-N-isopropylpropan-2-amine (26.0 µ , 0.1498 mmol) up in thf (748 µΙ_, ) and then added copper(I) iodide (7.13 mg, 0.03745 mmol) . Purged vial with Ar and stirred at r.t. O/N. complete by uplc. Concentrated solvent and dissolved in DMSO for purification by prep HPLC 5-95% ACN/water + 0.1% TFA. Isolated 2-(2,6-dioxopiperidin-3- yl)-4-((( 1-(6-((4-(l -methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)amino)hexyl)- 1H- 1,2,3 -triazol-4- yl)methyl)amino)isoindoline-l,3-dione (Degronimer 71) (30.0 mg, 0.03606 mmol, 48.1 %) as a yellowish solid. LCMS (ES+): m/z= 833 [M + H]+ lHNMR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.26 (s, 1H), 8.12 (d, J = 8.1 Hz, 1H), 8.06 - 7.90 (m, 2H), 7.83 (d, J = 8.5 Hz, 1H), 7.63 (d, J = 2.1 Hz, 1H), 7.61 - 7.37 (m, 2H), 7.13 (d, J = 8.7 Hz, 1H), 7.08 - 6.99 (m, 2H), 6.69 (d, J = 8.8 Hz, 2H), 5.04 (dd, J = 12.6, 5.4 Hz, 1H), 4.57 (d, J = 5.0 Hz, 2H), 4.30 (t, J = 7.0 Hz, 2H), 3.85 (s, 3H), 3.22 - 3.09 (m, 2H), 3.00 - 2.75 (m, 1H), 2.61 (s, 3H), 2.10 - 1.92 (m, 1H), 1.89 - 1.66 (m, 4H), 1.57 - 1.44 (m, 2H), 1.36 - 1.28 (m, 2H), 1.28 - 1.13 (m, 2H), 1.06 (s, 1H), 0.88 (s, 1H).

Example 74. Preparation of 4-(((l-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)but-3-yn-l- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 72) Scheme 289 8-(4-Azidobut- 1-yn- 1-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[/] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropane] (151-2) (.083 g, .1939 µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-

(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (60.3 mg, .1939 µηιοΐ) and N-ethyl-N- isopropylpropan-2-amine (367.5 , .3878 µηιοΐ) were dissolved in THF (1.93 mL ) . Copper iodide (18.4 mg, .09695 µιηοΐ) was added, the atmosphere purged with argon and the reaction was stirred for 16 hours, diluted with DMSO and purified by reverse phase HPLC to give 4-(((l-(4-(6- (4-chlorophenyl)- 1-methylspiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]- 8-yl)but-3-yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 72) (73.0mg, 51.0 %) as a yellow powder. LC/MS (ES+): m/z 740.5 [M + H]+ H MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.09 (s, 1H), 7.73 (d, J = 8.4 Hz, 1H), 7.61 (dd, J = 8.4, 1.9 Hz, 1H), 7.57 - 7.38 (m, 5H), 7.19 (d, J = 1.9 Hz, 1H), 7.07 (dd, J = 37.3, 7.8 Hz, 2H), 5.05 (dd, J = 12.9, 5.3 Hz, 1H), 4.60 - 4.46 (m, 4H), 2.99 (t, J = 6.6 Hz, 2H), 2.93 - 2.80 (m, 1H), 2.60 (s, 3H), 2.57 - 2.43 (m, 4H), 2.09 - 1.94 (m, 1H), 1.78 - 1.66 (m, 1H), 1.49 - 1.33 (m, 1H), 0.95 - 0.69 (m, 1H). Example 75. Preparation of 4-(((l-(10-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)dec-9-yn-l- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 73) Scheme 290

8-(l 0-Azidodec- 1-yn- 1-yl)-6-(4-chlorophenyl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4, 1'-cyclopropane] (153-1) (.150 g, 0.2929 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (91.1 mg, 0.2929 mmol) , N-ethyl-N-isopropylpropan-2-amine (102 , 0.5858 mmol) were taken up in THF (2.92 mL ) . Copper(I) iodide (27.8 mg, 0 .1464 mmol) was added, the atmosphere purged with argon and the reaction stirred for 16 hours. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(((l-(10-(6-(4-chlorophenyl)-l- methylspiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)dec-9-yn- 1-yl)- lH -l,2,34riazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 73) (185 mg, 76.7 %) as a yellow solid. LC/MS (ES+): m/z 824.7 [M + H]+ H MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.00 (s, 1H), 7.79 - 7.66 (m, 2H), 7.60 - 7.44 (m, 5H), 7.21 (d, J = 1.6 Hz, 1H), 7.08 (dd, J = 39.8, 7.8 Hz, 2H), 5.05 (dd, J = 12.9, 5.4 Hz, 1H), 4.57 (s, 2H), 4.28 (t, J = 7.0 Hz, 2H), 2.97 - 2.75 (m, 1H), 2.61 (s, 3H), 2.56 - 2.46 (m, 6H), 2.37 (t, J = 7.0 Hz, 2H), 2.09 - 1.93 (m, 1H), 1.80 - 1.64 (m, 3H), 1.53 - 1.36 (m, 3H), 1.34 - 1.26 (m, 2H), 1.24 - 1.13 (m, 3H), 0.85 - 0.76 (m, 2H).

Example 76. Preparation of 4-(4-(3-(3-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)propoxy)propyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3- dione (Degronimer 74) Scheme 291 2-(2,6-Dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (7.03 mg, 0.02548 mmol) 6-(4-chlorophenyl)-l-methyl-8-(l-(3-(3-(piperazin-l-yl)propoxy)propyl)-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (155-1) (16.2 mg, 0.02317 mmol) and N-ethyl-N-isopropylpropan-2-amine (16.0 , 0.09268 mmol) were heated to 90 C for 16 hours in DMAC (231 µΙ_, ) . The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(4-(3-(3-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)propoxy)propyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 74) (10.0 mg, 51.5 %) as a yellow solid. LC/MS (ES+): m/z 841.4 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 9.78 (s, 1H), 8.23 (s, 1H), 7.98 - 7.85 (m, 2H), 7.82 - 7.70 (m, 2H), 7.66 - 7.55 (m, 2H), 7.50 - 7.39 (m, 5H), 5.10 (dd, J = 12.7, 5.4 Hz, 1H), 4.12 (t, J = 6.9 Hz, 3H), 3.89 - 3.79 (m, 1H), 3.75 - 3.68 (m, 2H), 3.65 - 3.55 (m, 2H), 3.50 - 3.43 (m, 2H), 3.43 - 3.35 (m, 2H), 3.32 - 3.17 (m, 4H), 2.95 - 2.80 (m, 1H), 2.62 - 2.56 (m, 3H), 2.13 - 1.92 (m, 1H), 1.91 - 1.80 (m, 2H), 1.80 - 1.60 (m, 1H), 1.58 - 1.45 (m, 2H), 1.45 - 1.34 (m, 1H), 0.91 - 0.57 (m, 3H).

Example 77. Preparation of 4-(4-(2-(4-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)butoxy)ethyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 75) Scheme 292

6-(4-Chlorophenyl)-l-methyl-8-(l-(4-(2-(piperazin-l-yl)ethoxy)butyl)-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (157-1) (29 mg, 0.04147 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (12.5 mg, 0.04561 mmol) and N-ethyl-N-isopropylpropan-2-amine (28.8 , 0.1658 mmol) were heated to 90 °C in DMAC (414 ) for 12 hours. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(4-(2-(4-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)butoxy)ethyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 75) (18.0 mg, 51.7 %) as a yellow solid. LC/MS (ES+): m/z 841.5 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.07 (s, 1H), 9.87 (s, 1H), 8.21 (s, 1H), 8.00 - 7.82 (m, 2H), 7.79 - 7.65 (m, 2H), 7.65 - 7.53 (m, 2H), 7.55 - 7.30 (m, 6H), 5.09 (dd, J = 12.7, 5.5 Hz, 1H), 4.16 - 4.06 (m, 2H), 3.88 - 3.75 (m, 2H), 3.75 - 3.64 (m, 3H), 3.64 - 3.52 (m, 2H), 3.45 (t, J = 6.4 Hz, 2H), 3.42 - 3.33 (m, 1H), 3.33 - 3.20 (m, 4H), 2.58 (s, 3H), 2.10 - 1.92 (m, 1H), 1.89 - 1.77 (m, 2H), 1.77 - 1.64 (m, 1H), 1.58 - 1.43 (m, 2H), 1.43 - 1.34 (m, 1H), 0.89 - 0.66 (m, 3H).

Example 78. Preparation of 4-(4-((6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)(methyl)amino)piperidin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 76) heme 293

Degronimer 76

6-(4-Chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo [fJ[l,2,4]triazolo[4,3-a][l,4]diazepine-4,r-cyclopropane] (131-1) (63.0 mg, 0.1032 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-(4-(methylamino)piperidin-l-yl)isoindoline-l,3- dione 2,2,2-trifluoroacetate (21-2) (.05 g, 0.1032 mmol) and N-ethyl-N-isopropylpropan-2-amine (53.9 , 0.3096 mmol) were stirred for 16 hours in DMF (1.03 mL ) at 60 °C under argon. The reaction was diluted with DMSO and purified by reverse phase HPLC followed by preparative tic (15% MeOH in DCM) to give 4-(4-((6-(4-(6-(4-chlorophenyl)-l- methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)(methyl)amino)piperidin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 76) (3.00 mg, 3.40 %) as a thin film. LC/MS (ES+): m/z 853.6 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 9.25 (s, 1H), 8.21 (s, 1H), 7.99 - 7.83 (m, 2H), 7.71 (dd, J = 14.4, 8.1 Hz, 2H), 7.65 - 7.52 (m, 2H), 7.51 - 7.23 (m, 5H), 5.07 (dd, J = 12.7, 5.5 Hz, 1H), 4.09 (t, J = 6.9 Hz, 2H), 3.80 (s, 3H), 3.14 (s, 1H), 3.08 - 2.79 (m, 4H), 2.73 (d, J = 4.8 Hz, 3H), 2.58 (s, 3H), 2.04 (t, J = 16.3 Hz, 3H), 1.89 - 1.53 (m, 4H), 1.45 - 1.08 (m, 7H), 0.94 - 0.54 (m, 3H).

Example 79. Preparation of 4-((2-((6-(4-(6-(4-Chloroph methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)(methyl)amino)ethyl)(methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 77) Scheme 294 Degroriimer 77 6-(4-Chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] (131-1) (41 1 mg, 0.6732 mmol) 2-(2,6-dioxopiperidin-3-yl)-4-(methyl(2-(methylamino)ethyl)amino)isoindoline-

1,3-dione 2,2,2-trifluoroacetate (22-2) (.376 g, 0.6732 mmol) were dissolved in DMF (2.24 mL ). N-ethyl-N-isopropylpropan-2-amine (467 , 2.69 mmol) was added and the reaction heated to 50 °C for 16 hrs. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-((2-((6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1- yl)hexyl)(methyl)amino)ethyl)(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 77) (45.0 mg, 8.09 %) as a yellow solid. LC/MS (ES+): m/z 827.5 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.13 - 10.96 (m, 1H), 9.27 (s, 1H), 8.19 (s, 1H), 7.99 - 7.81 (m, 2H), 7.81 - 7.64 (m, 2H), 7.64 - 7.53 (m, 2H), 7.52 - 7.28 (m, 5H), 5.07 (dd, J = 12.8, 5.4 Hz, 1H), 4.07 (t, J = 7.0 Hz, 2H), 3.73 - 3.62 (m, 4H), 3.48 - 3.26 (m, 2H), 2.98 (s, 3H), 2.76 (dd, J = 5.0, 2.6 Hz, 3H), 2.58 (s, 3H), 2.09 - 1.90 (m, 1H), 1.81 - 1.67 (m, 3H), 1.67 - 1.45 (m, 2H), 1.32 (s, 1H), 1.29 - 1.15 (m, 6H), 0.91 - 0.63 (m, 2H). Example 80. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-lH- pyrazol-l-yl)hexyl)piperidin-4-yl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 78) Scheme 295

Degronimer 78

6-(4-Chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (131-1) (401 mg, 0.6568 mmol) and 2-(2,6-dioxopiperidin-3-yl)-4-(piperidin-4-ylamino)isoindoline-l,3-dione 2,2,2-trifluoroacetate (19-2) (.309 g, 0.6568 mmol) were dissolved in DMF (2.18 mL ) . N-ethyl-

N-isopropylpropan-2-amine (455 , 2.62 mmol) was added and the reaction stirred for 16 hours at 60 °C. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(( 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,l'-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)piperidin-4-yl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 78) (60.0 mg, 10.8 % ) as a yellow solid. LC/MS (ES+): m/z 840.6 [M + H ]+ ¾ M R (400 MHz, DMSO-de) δ 11.07 (s, 1H), 9.1 1 (s, 1H), 8.21 (s, 1H), 8.02 - 7.85 (m, 2H), 7.73 (d, J = 8.5 Hz, 1H), 7.68 - 7.54 (m, 3H), 7.54 - 7.37 (m, 3H), 7 .19 (dd, J = 14.2, 8.7 Hz, 1H), 7.09 (d, J = 7.0 Hz, 1H), 6.25 (d, J = 8.1 Hz, 1H), 5.04 (dd , J = 12.7, 5.5 Hz, 1H), 4.09 (t, J = 7.0 Hz, 2H), 3.80 (d, J = 7.9 Hz, 1H), 3.53 (d, J = 12.7 Hz, 3H), 3.01 (d, J = 11.2 Hz, 3H), 2.94 - 2.79 (m, 1H), 2.58 (s, 3H), 2.16 (d, J = 13.3 Hz, 1H), 2.07 - 1.87 (m, 1H), 1.81 - 1.52 (m, 5H), 1.27 (d, J = 39.2 Hz, 4H), 0.90 - 0.65 (m, 2H).

Example 81. Preparation of 3-(4-(4-(6-(4-(6-(4-Chloroph methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)piperazin-l-yl)-l-oxo-l,3-dihydro-2 H -pyrrolo[3,4-c]pyridin-2- yl)piperidine-2,6-dione (Degronimer 79) Scheme 296

Degrorsimer 79

6-(4-(6-(4-Chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexanal (137-1) (0.06 g, 0.1202 mmol), 3-(l-oxo-4- (piperazin-l-yl)-lH-pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine -(289-l) 2,6-dione 2,2,2- trifluoroacetate (69.2 mg, 0.1562 mmol) sodium acetate (14.7 mg, 0.1803 mmol) and acetic acid

(10.2 , 0.1803 mmol) were dissolved in DCE (1.20 mL ) and stirred for 1 hour. Sodium triacetoxy borohydride (33.1 mg, 0.1562 mmol) was added and the reaction stirred for 16 hours. Sat NaHCCb was added and the layers separated. The aqueous layer was extracted with DCM (2 x 5 mL) the organics combined and concentrated. The residue was purified by reverse phase HPLC to give 3-(4-(4-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperazin- 1-yl)- 1-oxo- 1H- pyrrolo[3,4-c]pyridin-2(3H)-yl)piperidine-2,6-dione isolated as the TFA salt (Degronimer 79) (24.0 mg, 24.5 %). LC/MS (ES+): m/z 812.6 [M + H]+ ¾ NMR (400 MHz, DMSO-de) δ 11.05 (s, 1H), 9.66 (s, 1H), 8.32 (d, J = 5.0 Hz, 1H), 8.22 (s, 1H), 7.97 - 7.88 (m, 2H), 7.74 (d, J = 8.4 Hz, 1H), 7.63 - 7.54 (m, 2H), 7.53 - 7.41 (m, 3H), 7.15 (d, J = 5.0 Hz, 1H), 5.16 (dd, J = 13.3, 5.1 Hz, 1H), 4.65 (d, J = 17.5 Hz, 1H), 4.48 (d, J = 17.5 Hz, 1H), 4.28 (d, J = 14.0 Hz, 2H), 4.09 (t, J = 7.0 Hz, 2H), 3.52 (d, J = 11.9 Hz, 2H), 3.28 (t, J = 13.1 Hz, 2H), 3.14 - 3.06 (m, 5H), 3.01 - 2.87 (m, 1H), 2.63 (d, J = 3.6 Hz, 1H), 2.60 (s, 3H), 2.47 - 2.32 (m, 1H), 2.06 - 1.96 (m, 1H), 1.84 - 1.68 (m, 2H), 1.45 - 1.35 (m, 1H), 1.34 - 1.20 (m, 5H), 0.87 - 0.69 (m, 2H).

Example 82. Preparation of 4-(4-(8-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)oct-7-yn-l- yl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 80) Scheme 297 6-(4-Chlorophenyl)- 1-methyl-8-(8-(piperazin- 1-yl)oct- 1-yn- 1- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropane] 2,2,2-trifluoroacetate (161-1) (.06 g, 0.09358 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (28.4 mg, 0.1029 mmol) and N-ethyl-N-isopropylpropan-2-amine (65.0 , 0.3743 mmol) were stirred for 16 hours in DMSO (935 µΙ_, ) at 90 °C. The reaction mixture was purified by reverse phase HPLC to give 4-(4-(8-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)oct-7-yn- 1-yl)piperazin- 1-yl)-2-(2,6-dioxopiperidin-3 - yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 80) (44.0 mg, 60.0 %) as a yellow solid. LC/MS (ES+): m/z 783.6 [M + H]+ H MR (400 MHz, DMSO-de) δ 11.09 (s, 1H), 9.54 (s, 1H), 7.81 - 7.67 (m, 3H), 7.59 - 7.44 (m, 5H), 7.41 (d, J = 8.4 Hz, 1H), 7.20 (d, J = 1.5 Hz, 1H), 5.10 (dd, J = 12.7, 5.5 Hz, 1H), 3.84 (d, J = 10.4 Hz, 2H), 3.62 (d, J = 9.0 Hz, 2H), 3.39 - 3.04 (m, 1H), 2.98 - 2.76 (m, 1H), 2.57 (s, 3H), 2.42 (t, J = 7.0 Hz, 2H), 2.18 - 1.89 (m, OH), 1.79 - 1.61 (m, 2H), 1.53 (q, J = 7.2 Hz, 2H), 1.48 - 1.16 (m, 5H), 0.90 - 0.65 (m, 2H) (Some peaks are obscured by the water peak).

Example 83. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/] [l ,2,4]triazolo[4,3-a] [l ,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)azetidin-3-yl)(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3- dione (Degronimer 81) Scheme 298 6-(4-(6-(4-Chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4,r-cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexanal (137-1) (0.07 g, 0.1402 mmol), (16-2) 4- (azetidin-3-yl(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione 2,2,2- trifluoroacetate (83.1 mg, .1822 mmol) sodium acetate (17.2 mg, 0.2103 mmol) and acetic acid

(12 µΐ 0.2103 mmol) were dissolved in DCE ( 1.40 mL ) and stirred for 1 hour. Sodium triacetoxy borohydride (38.6 mg, 0 .1822 mmol) was added and the reaction stirred for 16 hours. SatNaHCCb was added and the layers separated. The aqueous layer was extracted with DCM (2 x 5 mL) the organics combined and concentrated. The residue was purified by reverse phase HPLC to give 4- ((1 -(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)azetidin-3-yl)(methyl)amino)-2-(2,6-dioxopiperidin-

3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 81) (15.0 mg, 13.0 %). LC/MS (ES+): m/z 825.5 [M + H ]+ ¾ M R (400 MHz, DMSO-de) δ 11.07 (s, 1H), 9.64 (d, J = 36.5 Hz, 1H), 8.20 (s, 1H), 7.97 - 7.84 (m, 2H), 7.70 (dd, J = 20.7, 8.1 Hz, 2H), 7.58 (d, J = 8.5 Hz, 2H), 7.51 - 7.35 (m, 4H), 7.26

(d, J = 8.5 Hz, 1H), 5.09 (dd, J = 12.7, 5.4 Hz, 1H), 4.72 - 4.59 (m, 1H), 4.36 (d, J = 8.5 Hz, 2H), 4.22 (q, J = 9.1, 8.6 Hz, 2H), 4.07 (t, J = 6.9 Hz, 2H), 3.14 (s, 2H), 3.06 (d, J = 8.1 Hz, 3H), 2.58 (s, 3H), 2.00 (dd , J = 13.3, 6.7 Hz, 1H), 1.74 (dt , J = 12.2, 6.3 Hz, 3H), 1.53 - 1.33 (m, 2H), 1.22 (s, 6H), 0.77 (ddd, J = 29.8, 11.8, 7.4 Hz, 2H).

Example 84. Preparation of 4-(4-((6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)amino)piperidin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 82) Scheme 299 Degronimer 82

6-(4-(6-(4-Chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine- 4, -cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexanal (137-1) (.07 g, 0.1402 mmol) 4-(4- aminopiperidin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione 2,2,2-trifluoroacetate (17- 2) (85.7 mg, .1822 mmol) sodium acetate (17.2 mg, 0.2103 mmol) and acetic acid (12 , 0.2103 mmol) were dissolved in DCE (1.40 mL ) and stirred for 1 hour. Sodium triacetoxy borohydride (38.6 mg, 0.1822 mmol) was added and the reaction stirred for 16 hours. SatNaHCCb was added and the layers separated. The aqueous layer was extracted with DCM (2 x 5 mL) the organics combined and concentrated. The residue was purified by reverse phase HPLC to give 4-(4-((6-(4- (6-(4-chlorophenyl)- 1-methyl spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)amino)piperidin-l-yl)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 82) (30.0 mg, 25.6 %). LC/MS (ES+): m/z 839.4 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.03 (s, 1H), 8.21 (d, J = 0.8 Hz, 1H), 7.92 (dd, J = 8.4, 2.1 Hz, 1H), 7.88 (s, 1H), 7.72 (d, J = 8.4 Hz, 1H), 7.69 - 7.60 (m, 1H), 7.60 - 7.51 (m, 2H), 7.51 - 7.41 (m, 4H), 7.34 - 7.21 (m, 2H), 5.06 (dd, J = 12.9, 5.4 Hz, 1H), 4.07 (t, J = 6.9 Hz, 2H), 3.60 (d, J = 11.9 Hz, 2H), 2.97 - 2.81 (m, 3H), 2.57 (s, 4H), 2.06 - 1.94 (m, 1H), 1.86 (d, J = 12.3 Hz, 2H), 1.82 - 1.64 (m, 3H), 1.48 - 1.15 (m, 7H), 0.86 - 0.65 (m, 2H). Example 85. Preparation of 3-(4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)piperidin-4-yl)(methyl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione (Degronimer 83) heme 300

6-(4-Chlorophenyl)-8-(l-(6-iodohexyl)-lH-pyrazol-4-yl)-l- methylspiro[benzo [f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 , -cyclopropane] (131-1) (105 mg, 0.1721 mmol) and 3-(4-(methyl(piperidin-4-yl)amino)-l-oxoisoindolin-2-yl)piperidine-2,6-dione 2,2,2-trifluoroacetate (215-2) (.081 g, 0 .1721 mmol) were dissolved in DMF (0.573 mL ) . N-ethyl-

N-isopropylpropan-2-amine ( 119 , .6884 mmol) was added and the reaction stirred for 16 hours at 60 °C. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 3-(4-(( 1-(6-(4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperidin-4-yl)(methyl)amino)- l-oxoisoindolin-2-yl)piperidine-2,6-dione isolated as the TFA salt (Degronimer 83) (4.0 mg, 3 %) as a yellow solid. LC/MS (ES+): m/z 839.4 [M + H]+ ¾ M R (400 MHz, DMSO-de) δ 10.96 (s, 1H), 9.14 (s, 1H), 8.20 (s, 1H), 7.98 - 7.84 (m, 2H),

7.72 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.5 Hz, 2H), 7.52 - 7.37 (m, 3H), 7.27 (d, J = 7.4 Hz, 1H),

7.18 (d, J = 8.0 Hz, 1H), 5.09 (dd, J = 13.2, 5.2 Hz, 1H), 4.59 - 4.21 (m, 2H), 4.15 - 4.00 (m, 3H),

3.08 - 2.83 (m, 7H), 2.71 (s, 4H), 2.58 (s, 4H), 2.1 1 - 1.88 (m, 4H), 1.88 - 1.68 (m, 4H), 1.68 -

1.50 (m, 2H), 1.50 - 1.33 (m, 1H), 1.33 - 1.09 (m, 3H), 0.93 - 0.61 (m, 4H).

Example 86. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)piperidin-4-yl)(methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 84) Scheme 301

6-(4-Chlorophenyl)-8-( 1-(6-iodohexyl)- lH-pyrazol-4-yl)- 1- methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (131-1) (.2 g, 0.3273 mmol) and2-(2,6-dioxopiperidin-3-yl)-4-(methyl(piperidin-4-yl)amino)isoindoline-l,3-dione 2,2,2-trifluoroacetate (20-2) ( 206 mg, 0.4254 mmol) were taken up in DMF (1.63 mL ) and N - ethyl-N-isopropylpropan-2-amine (283 µ , 1.63 mmol) was added. The reaction was stirred for 16 hours at 70 °C, concentrated and the residue purified by reverse phase HPLC to give 4-((l-(6- (4-(6-(4-chlorophenyl)- 1-methylspiro[benzo[fJ [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)hexyl)piperidin-4-yl)(methyl)amino)-2-(2,6-dioxopiperidin- 3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 84) (40.0 mg, 14.3 %) as a yellow solid. LC/MS (ES+): m/z 853.6 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.08 (s, 1H), 9.18 (s, 1H), 8.21 (s, 1H), 7.94 (dd, = 8.5, 2.0 Hz, 1H), 7.89 (s, 1H), 7.74 (d, 7 = 8.4 Hz, 1H), 7.72 - 7.62 (m, 1H), 7.58 (d, 7 = 8.6 Hz, 2H), 7.53 - 7.39 (m, 3H), 7.32 (dd, 7 = 17.8, 7.8 Hz, 2H), 5.07 (dd, 7 = 12.8, 5.4 Hz, 1H), 4.08 (t, 7 = 6.9 Hz, 3H), 4.04 - 3.87 (m, 1H), 3.04 - 2.85 (m, 5H), 2.81 (s, 3H), 2.61 (s, 4H), 2.59 - 2.53 (m, 2H), 2.08 - 1.89 (m, 4H), 1.83 - 1.66 (m, 3H), 1.64 - 1.52 (m, 2H), 1.48 - 1.36 (m, 1H), 1.37 - 1.14 (m, 4H), 0.92 - 0.67 (m, 1H).

Example 87. Preparation of 4-((l-(6-(4-(6-(4-Chloroph methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)piperidin-4-yl)oxy)-2-(2,6-dioxopiperidin-3-yl)-l H -pyrrolo[3,4- c]pyridine-l,3(2 H )-dione (Degronimer 85) Scheme 302 2-(2,6-Dioxopiperidin-3-yl)-4-(piperidin-4-yloxy)-lH-pyrrolo[3,4-c]pyridine-l,3(2H)- dione 2,2,2-trifluoroacetate (43-1) (0.096 g, 0.2032 mmol), 6-(4-chlorophenyl)-8-(l-(6- iodohexyl)- lH-pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (131-1) (173 mg, 0.2844 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.6096 mmol) were dissolved in DMF (2.03 mL ) and heated for 16 hours at 50°C. After cooling the reaction was directly loaded onto reverse phase silica gel column (0-100% ACN in water) and purified to give 4-((l-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperidin-4-yl)oxy)-2-(2,6- dioxopiperidin-3-yl)-lH-pyrrolo[3,4-c]pyridine-l,3(2H)-dione (Degronimer 85) (55.0 mg, 32.3 %) LC/MS (ES+): m/z 841 .9 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.12 (s, 1H), 9.27 (d, J = 34.7 Hz, 1H), 8.72 (t, J = 4.8 Hz, 1H), 8.21 (s, 1H), 7.98 - 7.85 (m, 2H), 7.73 (d, J = 8.4 Hz, 1H), 7.61 - 7.39 (m, 7H), 5.1 1 (dd, J = 12.8, 4.7 Hz, 1H), 4.13 - 3.99 (m, 2H), 3.58 - 3.39 (m, 2H), 3.19 - 2.95 (m, 4H), 2.95 - 2.77 (m, 1H), 2.58 (s, 3H), 2.39 - 2.28 (m, 1H), 2.24 - 1.97 (m, 2H), 1.84 - 1.68 (m, 3H), 1.68 - 1.55 (m, 1H), 1.45 - 1.14 (m, 6H), 0.85 - 0.66 (m, 2H).

Example 88. Preparation of 4-((l-(6-(4-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)-l H - pyrazol-l-yl)hexyl)piperidin-4-yl)amino)-2-(2,6-dioxopiperidin-3-yl)-l H -pyrrolo[3,4- c]pyridine-l,3(2 H )-dione (Degronimer 86) Scheme 303 2-(2,6-Dioxopiperidin-3-yl)-4-(piperidin-4-ylamino)-lH-pyiTolo[3,4-c]pyridine-l,3(2H)- dione 2,2,2-trifluoroacetate (46-l) (0.096 g, 0.2036 mmol), 6-(4-chlorophenyl)-8-(l-(6- iodohexyl)- lH-pyrazol-4-yl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (131-1) (174 mg, 0.2850 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.8144 mmol) were dissolved in DMF (2.03 mL ) and stirred for 16 hours at 50°C. After cooling the reaction was directly loaded onto reverse phase silica gel column (0-100% ACN in water) and purified to give 4-((l-(6-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)hexyl)piperidin-4-yl)amino)-2-(2,6- dioxopiperidin-3-yl)-lH-pyrrolo[3,4-c]pyridine-l,3(2H)-dione (Degronimer 86) (55.0 mg, 0.06544 mmol, 32.1 %) as a yellow solid. LC/MS (ES+): m/z 840.6 [M + H]+

¾ NMR (400 MHz, DMSO-de) δ 11.1 1 (s, 1H), 9.12 (s, 1H), 8.59 (d, J = 4.8 Hz, 1H), 8.21 (s, 1H), 7.97 - 7.85 (m, 2H), 7.73 (dd, J = 8.5, 2.5 Hz, 1H), 7.65 - 7.54 (m, 2H), 7.54 - 7.38 (m, 4H), 7.07 (d, J = 4.8 Hz, 1H), 6.80 (d, J = 7.7 Hz, 1H), 5.09 (dd, J = 12.9, 5.4 Hz, 1H), 4.38 - 4.22 (m, 1H), 4.08 (t, J = 7.0 Hz, 2H), 3.51 (d, J = 12.0 Hz, 2H), 3.42 - 3.12 (m, 1H), 3.12 - 2.94 (m, 3H), 2.94 - 2.78 (m, 1H), 2.59 (s, 4H), 2.18 - 1.97 (m, 2H), 1.96 - 1.67 (m, 3H), 1.67 - 1.50 (m, 2H), 1.44 - 1.36 (m, 1H), 1.36 - 1.14 (m, 6H), 0.92 - 0.64 (m, 2H). Example 89. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(8-((4-(l-methyl-8-(l-methyl- l H -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropan]-6- yl)phenyl)amino)octyl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 87) Scheme 304

N-(8-Azidooctyl)-4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4, Γ-cyclopropan] (212-1) (0.005 g, 9.1 1

µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (3.67 mg, 11.8 µηιοΐ) and N-ethyl-N-isopropylpropan-2-amine (3.35 L, 18.2 µηιοΐ) were dissolved in THF

(91.1 µΙ_, ). Copper iodide (866 µg, 4.55 µηιοΐ) was added, the atmosphere purged with argon and the solution stirred for 16 hours. Purification by reverse phase HPLC to give 2-(2,6- dioxopiperidin-3-yl)-4-(((l-(8-((4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)amino)octyl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 87) (6.00 mg, 76.6 %) as a yellow powder. LC/MS (ES+): m/z 860.7 [M + H]+ H MR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.16 (s, 1H), 7.98 (s, 1H), 7.86 (s, 1H), 7.68 (d, J = 8.2 Hz, 1H), 7.59 - 7.47 (m, 2H), 7.31 (d, J = 8.2 Hz, 2H), 7.13 (d, J = 8.6 Hz, 1H), 7.02 (d, J = 6.9 Hz, 1H), 6.87 (s, 1H), 6.67 (s, 1H), 5.03 (dd, J = 12.9, 5.3 Hz, 1H), 4.56 (d, J = 6.1 Hz, 2H), 4.27 (t, J = 7.0 Hz, 1H), 3.97 (s, 1H), 3.82 (s, 2H), 3.06 - 2.91 (m, 6H), 2.74 - 2.62 (m, 3H), 2.56 (s, 2H), 2.33 - 2.29 (m, 1H), 2.05 - 1.95 (m, 2H), 1.74 (t, J = 7.3 Hz, 1H), 1.48 (s, 2H), 1.32 (s, 9H), 0.84 (t, J = 6.4 Hz, 2H).

Example 90. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(6-(4-(l-methyl-8-(l-methyl- l H -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropan]-6- yl)phenyl)hex-5-yn-l-yl)-l H-l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 88) Scheme 305

6-(4-(6-Azidohex- 1-yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (168-1) (0.066 g, 131 µ οΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (48.8 mg,

157 µ οΐ) and N-ethyl-N-isopropylpropan-2-amine (34.0 , 196 µ οΐ) were dissolved in THF

(1.30 mL ). Copper(I) iodide (12.4 mg, 65.5 µιηοΐ) was added, the atmosphere purged with argon and the reaction stirred for 16 hours. Purification by reverse phase HPLC to give 2-(2,6- dioxopiperidin-3-yl)-4-(((l-(6-(4-(l-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)hex-5-yn- 1- yl)-lH-l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 88) (66.0 mg, 62.2 % ) as a yellow powder. LC/MS (ES+): m/z 813.5 [M + H ]+

¾ NMR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.15 (s, 1H), 8.03 (s, 1H), 7.92 (dd, J = 8.5, 2.0 Hz, 1H), 7.87 (s, 1H), 7.75 (d, J = 8.4 Hz, 1H), 7.59 - 7.47 (m, 3H), 7.45 - 7.33 (m, 3H), 7.13 (d, J = 8.6 Hz, 1H), 7.02 (d, J = 7.1 Hz, 1H), 5.10 - 4.98 (m, 1H), 4.58 (s, 2H), 4.36 (t, J = 6.9 Hz, 2H), 3.81 (s, 3H), 2.93 - 2.78 (m, 1H), 2.61 (s, 3H), 2.53 (d, J = 10.7 Hz, 1H), 2.09 - 1.83 (m,

4H), 1.77 - 1.66 (m, 1H), 1.52 - 1.32 (m, 2H), 1.23 (s, 1H), 0.88 - 0.68 (m, 2H).

Example 91. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(8-(4-(l-methyl-8-(l-methyl- l H -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1'-cyclopropan]-6- yl)phenyl)oct -7-yn-l-yl)-l H-l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 89) heme 306

6-(4-(8-Azidooct- 1-yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (168-1) (0.065 g, 122 µιηοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (45.4 mg,

146 µιηοΐ) and N-ethyl-N-isopropylpropan-2-amine (29.5 170 µιηοΐ) were dissolved in THF

(1.22 mL ) . Copper(I) iodide ( 11.6 mg, 61.0 µιηοΐ), was added, the atmosphere purged with argon and the reaction was stirred for 16 hours, diluted with DMSO and purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-(((l-(8-(4-(l-methyl-8-(l -methyl- lH -pyrazol -4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)oct-7-yn- 1- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 89) (56.0 mg, 54.9 %) as a yellow powder. LC/MS (ES+): m/z 841 .6 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.14 (s, 1H), 8.00 (s, 1H), 7.92 (dd, J = 8.5, 2.1 Hz, 1H), 7.86 (s, 1H), 7.74 (d, J = 8.5 Hz, 1H), 7.61 - 7.48 (m, 3H), 7.45 - 7.30 (m, 3H), 7.13 (d, J = 8.6 Hz, 1H), 7.03 (d, J = 7.1 Hz, 1H), 5.03 (dd, J = 12.8, 5.4 Hz, 1H), 4.56 (s, 2H), 4.30 (t, J = 7.0 Hz, 2H), 3.81 (s, 3H), 2.96 - 2.77 (m, 1H), 2.60 (s, 3H), 2.38 (t, J = 6.8 Hz, 2H), 2.07 - 1.93 (m, 2H), 1.90 - 1.64 (m, 3H), 1.53 - 1.33 (m, 5H), 1.29 - 1.14 (m, 3H), 0.92 - 0.67 (m, 2H).

Example 93. Preparation of N-(10-Azidodecyl)-4-(l-methyl-8-(l-methyl-l H-pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)aniline (Degronimer 90) Scheme 308 N-(l 0-Azidodecyl)-4-( 1-methyl-8-( 1-methyl- lH-pyrazol -4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (171-1) (0.021 g, 36.4

µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (12.4 mg, 40.0 µηιοΐ) and N-ethyl-N-isopropylpropan-2-amine (12.6 µ , 72.8 µηιοΐ) were dissolved in

THF (364 µΙ_, ) . Copper(I) iodide (3.46 mg, 18.2 µιηοΐ) was added, the atmosphere purged with argon and the reaction was stirred for 16 hours, diluted with DMSO and purified by reverse phase HPLC to give N -(10-azidodecyl)-4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)aniline (Degronimer 90) (14.0 mg, 43.3 %) as an orange powder. LC/MS (ES+): m/z 888.7 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.23 (s, 1H), 7.99 (s, 1H), 7.92 (s, 1H), 7.78 (d, J = 8.3 Hz, 1H), 7.72 - 7.51 (m, 2H), 7.41 (s, 1H), 7.22 - 6.88 (m, 3H), 6.64 (s, 2H), 5.05 (dd, J = 12.9, 5.3 Hz, 1H), 4.57 (d, J = 5.8 Hz, 2H), 4.28 (t, J = 7.0 Hz, 2H), 3.84 (s, 3H), 3.14 (s, 2H), 2.97 - 2.78 (m, 1H), 2.59 (s, 3H), 2.1 1 - 1.93 (m, 1H), 1.86 - 1.66 (m, 2H), 1.60 - 1.44 (m, 2H), 1.34 - 1.06 (m, 9H), 0.91 - 0.67 (m, 2H). (Some peaks are obscured by the water peak).

Example 94. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(10-(4-(l-methyl-8-(l-methyl- l H -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropan]-6- yl)phenyl)dec-9-yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione Degronimer 91 Scheme 309 6-(4-( 10-Azidodec- 1-yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol -4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (173-1) (0.05 g, 89.6 µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (30.6 mg, 98.5 µηιοΐ) and N-ethyl-N-isopropylpropan-2-amine (31.1 µ , 179 µηιοΐ) were dissolved in

THF (895 µL ) . Copper(I) iodide (8.53 mg, 44.8 µιηοΐ) was added, the atmosphere purged with argon and the reaction was stirred for 16 hours, diluted with DMSO and purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-(((l-(10-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)dec-9-yn- 1- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 91) (53.0 mg, 68.1 %) as a yellow powder. LC/MS (ES+): m/z 870.7 [M + H]+ ¾ NMR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.13 (s, 1H), 7.98 (s, 1H), 7.90 (dd, J = 8.5, 2.1 Hz, 1H), 7.85 (s, 1H), 7.72 (d, J = 8.5 Hz, 1H), 7.60 - 7.48 (m, 3H), 7.42 - 7.32 (m, 3H), 7.13 (d, J = 8.6 Hz, 1H), 7.03 (d, J = 7.1 Hz, 1H), 5.03 (dd, J = 12.9, 5.4 Hz, 1H), 4.56 (d, J = 5.8 Hz, 2H), 4.28 (t, J = 7.0 Hz, 2H), 3.81 (s, 3H), 2.97 - 2.75 (m, 1H), 2.57 (s, 3H), 2.40 (t, J = 6.9 Hz, 2H),

2.12 - 1.91 (m, 1H), 1.84 - 1.66 (m, 2H), 1.57 - 1.46 (m, 2H), 1.41 - 1.29 (m, 3H), 1.29 - 1.1 1 (m, 7H), 0.87 - 0.67 (m, 1H). Example 95. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(4-(4-(l-methyl-8-(l-methyl- l H -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1'-cyclopropan]-6- yl)phenyl)but-3-yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 92) heme 310

6-(4-(4-Azidobut- 1-yn- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (175-1) (0.012 g, 25.3

µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (8.65 mg, 27.8 µηιοΐ) and N-ethyl-N-isopropylpropan-2-amine (8.80 L, 50.6 µηιοΐ) were dissolved in

THF (252 µΙ_, ) . Copper(I) iodide (2.39 mg, 12.6 µιηοΐ) was added, the atmosphere purged with argon and the reaction stirred for 16 hrs. The product was purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-(((l-(4-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)but-3 -yn- 1- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 92) (13.0 mg, 65.6 %) as a yellow powder. LC/MS (ES+): m/z 785.6 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.15 (s, 1H), 8.10 (d, J = 4.7 Hz, 1H), 7.93 (d, J = 8.5 Hz, 1H), 7.87 (s, 1H), 7.75 (d, J = 8.5 Hz, 1H), 7.48 (d, J = 8.1 Hz, 2H), 7.40 (s, 1H), 7.26 (d, J = 8.0 Hz, 1H), 7.21 - 7.07 (m, 1H), 7.05 - 6.90 (m, 1H), 5.08 - 4.89 (m, 1H), 4.70 - 4.40 (m, 3H), 3.81 (s, 3H), 3.01 (t, 7 = 6.6 Hz, 1H), 2.86 (td, 7 = 13.1, 6.8 Hz, 1H), 2.60 (s, 3H), 2.07 - 1.90 (m, 1H), 1.86 - 1.61 (m, 1H), 1.53 - 1.33 (m, 1H), 0.96 - 0.65 (m, 2H). (Some peaks are obscured by the water peak).

Example 96. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-((10-(4-(l-methyl-8-(l-methyl- lH -pyrazol-4-yl)spiro [benzo [ ][1,2,4]triazolo [4,3-a] [1,4] diazepine-4,1 '-cyclopropan]-6- yl)phenyl)dec-9-yn-l-yl)amino)isoindoline-l,3-dione (Degronimer 93) Scheme 311

10-(4-( 1-Methyl-8-( 1-methyl- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)dec-9-yn-l -amine (176-1) (0.03 g, 56.4 µ οΐ), 2-

(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (17.1 mg, 62.0 µιηοΐ) andN-ethyl-

N-isopropylpropan-2-amine (19.4 , 112 µ οΐ) were dissolved in DMAC (282 µΙ_, ) and heated to 90°C for 16 hrs. After cooling, the reaction was diluted with DMSO and purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-((10-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)dec-9-yn- 1- yl)amino)isoindoline-l,3-dione (Degronimer 93) (17.0 mg, 38.2 %) as a yellow powder. LC/MS (ES+): m/z 788.6 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.16 (s, 1H), 7.93 (dd, J = 8.4, 2.1 Hz, 1H), 7.88 (s, 1H), 7.75 (d, J = 8.4 Hz, 1H), 7.61 - 7.50 (m, 3H), 7.48 - 7.36 (m, 3H), 7.07 (d, J = 8.7 Hz, 1H), 6.99 (d, J = 7.0 Hz, 1H), 6.52 (s, 1H), 5.03 (dd, J = 12.9, 5.4 Hz, 1H), 3.81 (s, 3H), 3.27 (t, J = 7.1 Hz, 2H), 2.86 (ddd, J = 17.4, 14.0, 5.4 Hz, 1H), 2.62 (s, 3H), 2.60 - 2.52 (m, 1H), 2.42 (t, J = 7.0 Hz, 2H), 2.09 - 1.93 (m, 1H), 1.78 - 1.68 (m, 1H), 1.63 - 1.47 (m, 4H), 1.45 - 1.14 (m, 9H), 0.89 - 0.68 (m, 2H).

Example 97. Preparation of 4-(4-((l-(2,2-Dimethyl-4-(4-(l-methyl-8-(l-methyl-l H -pyrazol- 4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3- yn-l-yl)-l H-l,2,3-triazol-4-yl)methyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3-yl)isoindoline- 1,3-dione (Degronimer 94) heme 312 6-(4-(4-Azido-3 ,3-dimethylbut- 1-yn- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (179-1) (0.008 g, 0.0 1594 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-(4-(prop-2-yn- 1-yl)piperazin- 1-yl)isoindoline- 1,3- dione (213-2) (7.27 mg, 0.01912 mmol) and N-ethyl-N-isopropylpropan-2-amine (5.55 µ , 0.03188 mmol) were dissolved in THF (159 L ), copper(I) iodide (1.51 mg, 0.00797 mmol) was added, the atmosphere purged with argon and stirred for 16 hours. The reaction was diluted with DMSO and purified by reverse phase HPLC to give 4-(4-((l-(2,2-dimethyl-4-(4-(l-methyl-8-(l- methyl- lH -pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)but-3-yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)piperazin-l-yl)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 94) (7.00 mg, 50.0 %) as a yellow solid. LC/MS (ES+): m/z 882.6 [M + H]+ H MR (500 MHz, DMSO-de) δ 11.09 (s, 1H), 10.22 (s, 1H), 8.38 (s, 1H), 8.12 (s, 1H), 7.90 (s, 1H), 7.83 (d, J = 2.7 Hz, 1H), 7.80 - 7.64 (m, 2H), 7.54 (d, J = 8.2 Hz, 2H), 7.48 - 7.23 (m, 4H), 5.12 - 5.03 (m, 1H), 4.59 (s, 4H), 3.82 (s, 3H), 3.35 - 3.07 (m, 4H), 2.95 - 2.79 (m, 1H), 2.62 (s, 1H), 2.58 (s, 4H), 2.35 (s, 1H), 2.07 - 1.94 (m, 3H), 1.79 - 1.65 (m, 1H), 1.33 (s, 6H), 0.91 - 0.65 (m, 3H).

Example 98. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(2-(4-(4-(l-methyl-8-(l- methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropan]-6-yl)phenyl)piperazin-l-yl)ethyl)-l H -1,2,3-triazol-4- yl)methyl)amino)isoindoline-l,3-dione (Degronimer 95) Scheme 313 6-(4-(4-(2- Azidoethyl)piperazin- 1-yl)phenyl)- 1-methyl-8-( 1-methyl- lH -pyrazol-4- yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (183-1) (0.019 g, 35.6 µ οΐ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (12.1 mg, 39.1 µ οΐ) and N-ethyl-N-isopropylpropan-2-amine (12.3 µ , 71.2 mol) were dissolved in

THF (356 µΙ_, ), copper(I) iodide (3.39 mg, 17.8 µιηοΐ) was added and the reaction stirred for 16 hours at room temperature. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-(((l-(2-(4-(4-(l-methyl-8-(l -methyl- 1H - pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)piperazin-l-yl)ethyl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 95) (15.0 mg, 50.0 % ) as a yellow solid. LC/MS (ES+): m/z 845.5 [M + H ]+ ¾ M R (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.18 (s, 1H), 8.10 (s, 1H), 7.95 (d, J = 8.4, 2.1 Hz, 1H), 7.86 (s, 1H), 7.76 (d, J = 8.5 Hz, 1H), 7.57 (t, J = 8.5, 7.1 Hz, 1H), 7.52 - 7.38 (m, 3H),

7.21 - 7.10 (m, 2H), 7.09 - 6.93 (m, 3H), 5.05 (dd, J = 12.7, 5.5, 2.4 Hz, 1H), 4.80 (t, J = 6.6 Hz, 2H), 4.63 (d, J = 5.5 Hz, 2H), 3.83 (s, 3H), 3.66 (s, 1H), 3.56 - 2.98 (m, 7H), 2.96 - 2.79 (m, 1H), 2.60 (s, 3H), 2.08 - 1.94 (m, 1H), 1.77 - 1.65 (m, 1H), 1.53 - 1.34 (m, 1H), 1.22 (s, 1H), 0.87 -

0.66 (m, 2H). Example 99. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(6-(4-(l-methyl-8-(l-methyl-l H - pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6- yl)phenyl)-2,6-diazaspiro[3.3]heptan-2-yl)isoindoline-l,3-dione (Degronimer 96) Scheme 314

6-(4-(2,6-Diazaspiro[3.3]heptan-2-yl)phenyl)-l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4 ,r-cyclopropane] (185-1) (0.012 g, 0.02517 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (7.64 mg, 0.02768 mmol) and N-ethyl-N-isopropylpropan-2-amine (8.76 , 0.05034 mmol) were dissolved in N-Methyl-2-pyrrolido (25 1 µΙ_, ) . The reaction was heated for 16 hours at 90°C and cooled. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give2-(2,6- dioxopiperidin-3-yl)-4-(6-(4-(l-methyl-8-(l-methyl-lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)-2,6- diazaspiro[3.3]heptan-2-yl)isoindoline-l,3-dione (Degronimer 96) ( 11.0 mg, 59.7 %). LC/MS (ES+): m/z 733.5 [M + H ]+

¾ M R (400 MHz, DMSO-de) δ 11.08 (s, 1H), 8.21 (s, 1H), 8.10 - 8.00 (m, 1H), 7.90 (s, 1H), 7.79 (d, J = 8.3 Hz, 1H), 7.63 - 7.55 (m, 1H), 7.55 - 7.44 (m, 2H), 7.14 (d, J = 7.0 Hz, 1H), 6.81 (d, J = 8.6 Hz, 1H), 6.49 (d, J = 8.8 Hz, 2H), 5.05 (dd, J = 12.7, 5.5 Hz, 1H), 4.41 (s, 4H), 4.34 -

4.05 (m, 4H), 3.85 (d, J = 5.0 Hz, 3H), 3.01 - 2.74 (m, 1H), 2.61 (s, 2H), 2.40 - 2.16 (m, 1H), 2.07 - 1.91 (m, 2H), 1.85 - 1.72 (m, 1H), 1.68 - 1.40 (m, 1H), 1.23 (s, 1H), 1.00 - 0.74 (m, 1H).

Example 100. Preparation of 4-(((l-(2,2-Dimethyl-4-(4-(l-methyl-8-(l-methyl-li/-pyrazol- 4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-6-yl)phenyl)but-3- yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3- dione (Degronimer 97) Scheme 315

6-(4-(4-Azido-3 ,3-dimethylbut- 1-yn- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (179-1) (0.013 g, 25.9

µηιοΐ ), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (8.84 mg, 28.4 µηιοΐ ) and N-ethyl-N-isopropylpropan-2-amine (9.01 , 51.8 µηιοΐ ) were dissolved in

THF (258 µΙ_, ) . Copper(I) iodide (2.45 mg, 12.9 µιηοΐ ) was added, the atmosphere purged with argon and the reaction was stirred for 16 hours.. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(((l-(2,2-dimethyl-4-(4-(l-methyl-8-(l-methyl-l H - pyrazol-4-yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6- yl)phenyl)but-3-yn-l-yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 97) (12.0 mg, 57.1 %) as a yellow solid. LC/MS (ES+): m/z 813.6 [M + H]+ ¾NMR (400 MHz, DMSO-de) δ 8.14 (s, 1H), 8.05 (d, J = 11.5 Hz, 1H), 7.87 (s, 1H), 7.76 (dd, J = 8.4, 3.0 Hz, 1H), 7.64 (dd, J = 8.6, 7.1 Hz, 2H), 7.48 (d, J = 8.4 Hz, 2H), 7.28 (dd, J = 8.2, 5.1 Hz, 2H), 7.15 (d, J = 8.4 Hz, 2H), 7.01 - 6.84 (m, 2H), 5.06 (dd, J = 12.9, 5.4 Hz, 1H), 4.61 (d, J = 4.4 Hz, 2H), 4.46 (s, 2H), 4.26 - 4.08 (m, 2H), 3.81 (s, 3H), 3.27 - 3.09 (m, 1H), 2.87 (ddd, J = 17.2, 14.0, 5.5 Hz, 2H), 2.61 (d, J = 2.5 Hz, 4H), 2.06 - 1.93 (m, 3H), 1.82 - 1.67 (m, 1H), 1.26 (s, 3H), 0.79 (d, J = 23.5 Hz, 2H).

Example 116. Preparation of 2-(2,6-Dioxopiperidin-3-yl)-4-(((l-(6-(4-(4-(l-methyl-8-(l- methyl-l H -pyrazol-4-yl)spiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropan]-6-yl)phenyl)piper azin-l-yl)hexyl)- \H- 1,2,3-triazol-4- yl)methyl)amino)isoindoline-l,3-dione (Degronimer 98) Scheme 309 6-(4-(4-(6- Azidohexyl)piperazin- 1-yl)phenyl)- 1-methyl-8-(l -methyl- lH-pyrazol-4- yl)spiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l '-cyclopropane] (187-1) 0.02 g, 0.03391 mmol), 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) ( 11.6 mg, 0.03730 mmol)and N-ethyl-N-isopropylpropan-2-amine ( 11.8 , 0.06782 mmol) were dissolved in THF (339 µΙ_, ) copper(I) iodide (645 g, 0.003391 mmol) was added and the reaction stirred for 16 hours. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 2-(2,6-dioxopiperidin-3-yl)-4-(((l-(6-(4-(4-(l-methyl-8-(l-methyl-l H -pyrazol-4- yl)spiro[benzo[/] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'-cyclopropan]-6-yl)phenyl)piperazin- 1- yl)hexyl)-l H -l,2,3-triazol-4-yl)methyl)amino)isoindoline-l,3-dione (Degronimer 98) (0.022g, 72.1 %) as a bright yellow solid. LC/MS (ES+): m/z 901.6 [M + H]+ H MR (500 MHz, DMSO-de) δ 11.09 (s, 1H), 9.45 (s, 1H), 8.17 (s, 1H), 8.01 (s, 1H), 7.91 (d, J = 8.3 Hz, 1H), 7.85 (s, 1H), 7.74 (d, J = 8.4 Hz, 1H), 7.57 (t, J = 7.8 Hz, 1H), 7.52 - 7.40 (m, 3H), 7.15 (d, J = 8.6 Hz, 1H), 7.03 (dd, J = 26.9, 7.7 Hz, 4H), 5.05 (dd, J = 12.8, 5.4 Hz, 1H), 4.58 (s, 2H), 4.32 (t, J = 7.1 Hz, 2H), 3.96 (d, J = 11.9 Hz, 2H), 3.83 (s, 3H), 3.55 (d, J = 10.9 Hz, 2H), 3.08 (d, J = 14.6 Hz, 6H), 2.59 (s, 4H), 2.12 - 1.94 (m, 1H), 1.90 - 1.75 (m, 2H), 1.73 - 1.53 (m, 3H), 1.40 - 1.17 (m, 6H), 0.77 - 0.67 (m, 2H).

Example 117. Preparation of 4-(((l-(6-(6-(4-Chlorophenyl)-l- methylspiro[benzo[/][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'-cyclopropan]-8-yl)hex-5-yn-l- yl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 99) Scheme 310 8-(6-Azidohex- 1-yn- 1-yl)-6-(4-chlorophenyl)- 1-methylspiro[benzo[f] [1,2,4]triazolo[4,3 - a][l,4]diazepine-4,l '-cyclopropane] (163-1) (.1 g, 0.2193 mmol) , 2-(2,6-dioxopiperidin-3-yl)-4- (prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (68.2 mg, 0.2193 mmol) , N-ethyl-N- isopropylpropan-2-amine (76.2 L, 0.4386 mmol) were taken up in THF (2.19 mL ) . copper(I) iodide (20.8 mg, 0.1096 mmol) was added, the atmosphere purged with argon and the reaction stirred for 16 hours. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-(((l-(6-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)hex-5-yn- 1-yl)- 1H- 1,2,3 -triazol-4-yl)methyl)amino)-2- (2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 99) (40.0 mg, 23.8 %) as a yellow solid. LC/MS (ES+): m/z Ί 6Ί . [M + H]+ H MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.01 (s, 1H), 7.77 - 7.64 (m, 2H), 7.61 - 7.42 (m, 5H), 7.22 (s, 1H), 7.13 (d, J = 8.6 Hz, 1H), 7.02 (d, J = 7.1 Hz, 1H), 5.05 (dd, J = 12.8, 5.4 Hz, 1H), 4.57 (s, 2H), 4.34 (t, J = 6.9 Hz, 2H), 2.97 - 2.79 (m, 1H), 2.60 (s, 4H), 2.58 - 2.51 (m, 1H), 2.42 (t, J = 7.1 Hz, 2H), 2.08 - 1.94 (m, 1H), 1.94 - 1.82 (m, 2H), 1.79 - 1.60 (m, 1H), 1.50 - 1.35 (m, 2H), 0.88 - 0.67 (m, 1H). Example 118. Preparation of N-(2-(((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethyl)-2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamide (Degronimer 100) Scheme 311

2-((2-(2,6-Dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetic acid (311-1)(2.37 mg, 7.14 µιηοΐ) was dissolved in DMF (23.7 ΐ ) and HATU (2.71 mg, 7.14 µιηοΐ) was added. After stirring for 10 minutes a solution of (R)-2-((4-((2-aminoethyl)sulfonyl)-2-methylpiperazin- l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one 2,2,2-trifluoroacetate (189-1) (.003 g, 4.76 µιηοΐ) and diisopropylethylamine (3.24 µΐ , 19.0 µmol) in DMF (23.7 µΙ_, ) was added and the solution allowed to stir for 1 hour at which time the reaction was diluted with DMSO and purified by reverse phase HPLC to give N-(2-(((R)-4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazin-l-yl)sulfonyl)ethyl)-2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)acetamide isolated as the TFA salt (Degronimer 100) (2.00 mg, 50.6 %) as an off white powder. LC/MS (ES+): m/z 830.6 [M + H]+ Example 104. Preparation of N -(10-(((R )-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)decyl)-2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamide (Degronimer 101) Scheme 319

Degronimer 0 1

(R)-2-((4-((10-Aminodecyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)-7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (193-1) (0.013 g, 0.02070 mmol), 2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetic acid (311-1) (10.3 mg, 0.03105 mmol) and diisopropylethylamine (8.84 , 0.05175 mmol) were taken up in

DMF (200uL), cooled to 0 °C and stirred for five minutes. T3P (17.1 µΐ , 0.02691 mmol) was added and the ice bath removed. The reaction was stirred for 1 hour, diluted with DMSO and purified by reverse phase HPLC to give N-(10-(((R)-4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)- 5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)decyl)-2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamide isolated as the TFA salt (Degronimer 101) (4.90 mg, 25. 1 %) as a white solid. LC/MS (ES+): m/z 972.7 [M + H]+

Example 105. Preparation of 4-((12-((( R )-4-((7-(3-(Cyclopropylmethoxy)pyridin -4-yl)-5- methyl-4-oxo-4,5-dihydrofuro [3,2-c]pyridin -2-yl)methyl )-3-methylpiperazin-l- yl)sulfonyl)dodecyl)amino )-2-(2,6-dioxopiperidin -3-yl)isoindoline-l ,3-dione (Degronimer 102) Scheme 320

Degronimer 1 2 (R)-2-((4-(( 12-Aminododecyl)sulfonyl )-2-methylpiperazin- 1-yl)methyl)-7-(3- (cyclopropylmethoxy)pyridin -4-yl)-5-methylfuro [3,2-c]pyridin-4(5H)-one (197-1) (0.005 g, 7.62

µηιοΐ ), 2-(2,6-dioxopiperidin -3-yl)-4-fluoroisoindoline-l ,3-dione (1-3) (3.14 mg, 11.4 µ οΐ ) and

Diisopropylethylamine (2.64 L, 15.2 mol) were dissolved in DMAC (76.2 µΙ_, ), the atmosphere was purged with argon and the reaction heated to 90 °C for 4 hrs. The reaction was diluted with DMSO and purified by reverse phase HLPC to give 4-((12-(((R)-4-((7-(3- (cyclopropylmethoxy)pyridin -4-yl)-5-methyl-4-oxo-4,5-dihydrofuro [3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazin-l-yl)sulfonyl)dodecyl)amino )-2-(2,6-dioxopiperidin -3-yl)isoindoline-l ,3- dione isolated as the TFA salt (Degronimer 102) (2.40 mg, 34.5 %) as a yellow powder. LC/MS (ES+): m/z 457.2 [M + H]+. ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.48 (s, 1H), 8.32 (d, J = 4.9 Hz, 1H), 8.07 (s, 1H), 7.65 - 7.49 (m, 2H), 7.26 (s, 4H), 7.13 (s, 4H), 7.07 (d, J = 8.6 Hz, 1H), 7.01 (d, J = 5.2 Hz, 5H), 6.52 (t, J = 6.0 Hz, 1H), 5.04 (dd, J = 12.8, 5.4 Hz, 1H), 4.01 (d, J = 6.8 Hz, 2H), 3.27 (q, J = 6.5 Hz, 2H), 3.04 (t, J = 7.7 Hz, 2H), 2.06 - 1.91 (m, 1H), 1.67 - 1.44 (m, 4H), 1.41 - 0.99 (m, 21H), 0.49 (dd, J = 8.0, 1.9 Hz, 2H), 0.26 (d, J = 4.9 Hz, 2H).

Example 106. Preparation of 4-((6-(((R )-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)hexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 103) heme 321

(R)-2-((4-((6-Aminohexyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)-7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (201-1) (.005 g, 8.74 µηιοΐ) and 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (3.61 mg, 13.1 µηιοΐ) were dissolved in DMAC (87.3 µΙ_, ). Diisopropylethylamine (3.01 , 17.4 µηιοΐ) was added, the atmosphere purged with argon and the reaction heated to 90 °C for 4 hrs. The reaction was diluted with DMSO and purified by reverse phase HPLC to give 4-((6-(((R)-4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazin-l-yl)sulfonyl)hexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione isoalated as the TFA salt (Degronimer 103) (4.80 mg, 66.3 %) as a yellow powder. LC/MS (ES+): m/z 848.7 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.45 (s, 1H), 8.28 (d, J = 5.0 Hz, 1H), 7.98 (s, 1H), 7.62 - 7.45 (m, 2H), 7.20 (s, 1H), 7.06 (d, J = 9.3 Hz, 2H), 7.00 (d, J = 7.1 Hz, 1H), 6.95 (s, 1H), 5.03 (dd, J = 12.9, 5.4 Hz, 1H), 4.00 (d, J = 6.8 Hz, 2H), 3.55 (s, 3H), 3.06 - 2.79 (m, 4H), 1.99 (t, J = 7.5 Hz, 1H), 1.74 - 1.45 (m, 4H), 1.45 - 1.08 (m, 19H), 0.54 - 0.37 (m, 2H), 0.27 - 0.16 (m, 2H).

Example 107. Preparation of 4-((10-(((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)decyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 104) Scheme 322

Degronimer 4 (R)-2-((4-((10-Aminodecyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)-7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (193-1) (.013 g, 20.7

µιηοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (8.56 mg, 31.0 µιηοΐ) and diisopropylethylamine (7.21 , 41.4 µιηοΐ) were dissolved in DMAC (206 µΙ_, ) . The atmosphere was purged with argon and the reaction heated to 90°C for 16 hrs. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give 4-((10-(((R)-4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methy 3-methylpiperazin-l-yl)sulfonyl)decyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 104) (4.70 mg, 25.8 %) as a yellow solid. LC/MS (ES+): m/z 884.8 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.49 (s, 1H), 8.33 (d, J = 5.0 Hz, 1H), 8.10 (s, 1H), 7.72 - 7.43 (m, 2H), 7.04 (dd, J = 27.1, 7.8 Hz, 2H), 6.52 (s, 1H), 5.04 (dd, J = 12.8, 5.4 Hz, 1H), 4.02 (d, J = 6.9 Hz, 2H), 3.57 (s, 3H), 3.32 - 3.19 (m, 2H), 3.1 1 - 2.96 (m, 2H), 2.94 - 2.75 (m, 1H), 2.68 - 2.51 (m, 1H), 2.06 - 1.93 (m, 1H), 1.69 - 1.43 (m, 4H), 1.38 - 1.02 (m, 15H), 0.60 - 0.44 (m, 2H), 0.27 (dd, J = 4.7, 1.7 Hz, 2H). (Some peaks are obscured by the water peak)

Example 108. Preparation of 4-((l-(2-(2-(2 -(((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4- yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethoxy)ethoxy)ethyl)-l H -l,2,3-triazol-4-yl)methoxy)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione (Degronimer 105) Scheme 323 (R)-2-((4-((2-(2-(2-Azidoethoxy)ethoxy)ethyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)- 7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (202-1) (.002 g,

3.17 µηιοΐ) and 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-yloxy)isoindoline-l,3-dione (4-1)

(988 g, 3.17 µηιοΐ) were dissolved in THF (31.7 L ). Diisopropylethylamine (1.64 µ , 9.51

µηιοΐ) was added followed by copper iodide (300 µg, 1.58 µιηοΐ) and the atmosphere was purged with argon. The reaction was stirred for 4 hours, diluted with DMSO and purified by reverse phase HPLC to give 4-((l-(2-(2-(2-(((R)-4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo- 4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethoxy)ethoxy)ethyl)-lH-l,2,3-triazol-4-yl)methoxy)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 105) (900 µg, 30.2 %) as a thin film. LCVMS (ES+): m/z 942 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.45 (s, 1H), 8.29 (d, J = 9.8 Hz, 1H), 8.20 (s, 1H), 8.00 - 7.93 (m, 1H), 7.85 - 7.76 (m, 1H), 7.71 (d, J = 8.6 Hz, 1H), 7.51 (d, J = 4.5 Hz, 1H), 7.45 (d, J = 7.2 Hz, 1H), 6.87 (s, 2H), 6.67 (s, 1H), 5.39 (s, 2H), 5.10 - 4.96 (m, 1H), 4.58 - 4.47 (m, 2H), 4.13 - 3.94 (m, 5H), 3.78 (t, J = 5.1 Hz, 2H), 3.73 - 3.50 (m, 6H), 2.82 - 2.58 (m, 2H), 2.08 - 1.91 (m, 2H), 1.27 - 1.05 (m, 9H), 0.47 (d, J = 8.1 Hz, 2H), 0.29 - 0.18 (m, 2H). (Some peaks obscured by the water peak).

Example 109. Preparation of 4-(((l-(2-(2-(2 -(((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4- yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethoxy)ethoxy)ethyl)-l H -l,2,3-triazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin- 3-yl)isoindoline-l,3-dione (Degronimer 106) Scheme 324 (R)-2-((4-((2-(2-(2-Azidoethoxy)ethoxy)ethyl)sulfonyl)-2-methylpiperazin-l-yl)methyl)- 7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (202-2) (.002 g, 3.17 µηιοΐ) and 2-(2,6-dioxopiperidin-3-yl)-4-(prop-2-yn-l-ylamino)isoindoline-l,3-dione (5-1) (985 µg, 3.17 µηιοΐ) were dissolved in THF (31.7 L ) Diisopropylethylamine (1.64 µ , 9.51

µηιοΐ) was added followed by copper iodide (300 µg, 1.58 µιηοΐ) and the atmosphere purged with argon. The reaction was stirred for 4 hours, diluted with DMSO and purified by reverse phase HPLC to give 4-(((l-(2-(2-(2-(((R)-4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo- 4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l- yl)sulfonyl)ethoxy)ethoxy)ethyl)-lH-l,2,34riazol-4-yl)methyl)amino)-2-(2,6-dioxopiperidin-3- yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 106) (900 µg, 30.2 %). LC/MS (ES+): m/z 941.7 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.06 (s, 1H), 8.45 (s, 1H), 7.96 (s, 1H), 7.53 (d, J = 8.4 Hz, 1H), 7.15 (d, J = 8.6 Hz, 1H), 7.06 - 6.93 (m, 2H), 6.49 (bs, 14H), 4.56 (d, J = 5.9 Hz, 1H), 4.45 (s, 1H), 3.99 (d, J = 6.9 Hz, 2H), 3.74 (t, J = 5.2 Hz, 1H), 3.68 - 3.51 (m, 4H), 2.65 (d, J = 2.0 Hz, 1H), 2.31 (d, J = 2.2 Hz, 2H), 1.99 (s, 3H), 1.33 - 1.02 (m, 7H), 0.55 - 0.39 (m, 1H), 0.29 - 0.16 (m, 2H). (Some peaks are obscured by the water peak). Example 110. Preparation of 4-((ll -((R)-4-((7-(3,4-Dimethoxyphenyl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-ll-oxoundecyl)amino)-2- (2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 107) Scheme 325

(R)-2-((4-( 11-Aminoundecanoyl)-2-methylpiperazin- 1-yl)methyl)-7-(3 ,4- dimethoxyphenyl)-5-methylfuro[3,2-c]pyridin-4(5H)-one (206-1) (.03 g, 51.6 µιηοΐ), 2-(2,5- dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (21.3 mg, 77.4 µηιοΐ) and

Diisopropylethylamine (28.4 µ , 154 µηιοΐ) were dissolved in DMF (258 µΙ_, ) . The atmosphere was purged with argon and the reaction heated for 16 hrs at 90 °C and cooled. Purification by reverse phase hplc to give 4-((l l-((R)-4-((7-(3,4-dimethoxyphenyl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-l l-oxoundecyl)amino)-2-(2,5- dioxopiperidin-3-yl)isoindoline-l,3-dione isolated as the TFA salt (Degronimer 107) (14.0 mg, 32.4 %) as a yellow powder. LC/MS (ES+): m/z 838.3 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 7.99 (s, 1H), 7.56 (dd, J = 8.5, 7.1 Hz, 1H), 7.31 - 7.19 (m, 3H), 7.13 - 6.87 (m, 3H), 6.51 (s, 1H), 5.04 (dd, J = 12.8, 5.4 Hz, 1H), 3.80 (d, J = 12.9 Hz, 7H), 3.58 (s, 3H), 3.26 (d, J = 6.8 Hz, 2H), 2.94 - 2.77 (m, 1H), 2.69 - 2.52 (m, 2H), 2.34 - 2.15 (m, 3H), 2.07 - 1.90 (m, 1H), 1.60 - 1.48 (m, 2H), 1.50 - 1.34 (m, 5H), 1.35 - 1.02 (m, 15H).

Example 111. Preparation of N -(ll -((R )-4-((7-(3,4-Dimethoxyphenyl)-5-methyl-4-oxo-4,5- dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-ll-oxoundecyl)-2-((2-(2,6- dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamide (Degronimer 108) Scheme 326

HATU (29.4 mg, 77.4 µιηοΐ ) was added to a solution of 2-((2-(2,5-dioxopiperidin-3-yl)- l,3-dioxoisoindolin-4-yl)oxy)acetic acid (311-1) (25.7 mg, 77.4 µιηοΐ ) and diisopropylethylamine

(28.4 , 154 µιηοΐ ) in DMF (258 µ ) . After stirring 30 min (R)-2-((4-(l l-aminoundecanoyl)-2- methylpiperazin-l-yl)methyl)-7-(3,4-dimethoxyphenyl)-5-methylfuro[3,2-c]pyridin-4(5H)-one

(206-1) (.03 g, 51.6 µιηοΐ ) was added and the solution stirred for 16 hrs. The reaction mixture was diluted with DMSO and purified by reverse phase HPLC to give N-(l l-((R)-4-((7-(3,4- dimethoxyphenyl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3- methylpiperazin-l-yl)-ll-oxoundecyl)-2-((2-(2,5-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4- yl)oxy)acetamide isolated as the TFA salt (Degronimer 108) (24.0 mg, 52.0 %) as a white powder LC/MS (ES+): m/z 895.8 [M + H]+ ¾ NMR (400 MHz, DMSO-de) δ 11.12 (s, 1H), 8.00 (s, 1H), 7.79 (t, J = 7.9 Hz, 1H), 7.48 (d, J = 7.2 Hz, 1H), 7.37 (d, J = 8.6 Hz, 1H), 7.35 - 7.21 (m, 3H), 7.05 (d, J = 8.3 Hz, 1H), 5.17 - 5.06 (m, 1H), 4.75 (s, 2H), 4.19 (s, 1H), 3.80 (d, J = 12.1 Hz, 8H), 3.58 (s, 4H), 3.1 1 (s, 2H), 2.37 - 2.18 (m, 2H), 1.40 (s, 8H), 1.19 (s, 20H).

Example 112. Preparation of 4-((ll -((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-ll- oxoundecyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-l,3-dione (Degronimer 109) Scheme 327

(R)-2-((4-( 11-Aminoundecanoyl)-2-methylpiperazin- 1-yl)methyl)-7-(3 - (cyclopropylmethoxy)pyridin-4-yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one hydrochloride (208-2) (.04 g, 63.6 µηιοΐ), 2-(2,6-dioxopiperidin-3-yl)-4-fluoroisoindoline-l,3-dione (1-3) (22.8 mg, 82.6

µηιοΐ) andN-ethyl-N-isopropylpropan-2-amine (35.0 , 190 µηιοΐ) were dissolved in DMF (636 µL· ), the atmosphere purged with argon and heated to 90 °C for 16 hrs. The reaction was diluted with DMSO and purified by reverse phase HPLC to give 4-((l l-((R)-4-((7-(3- (cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)- 3-methylpiperazin- 1-yl)- 11-oxoundecyl)amino)-2-(2,6-dioxopiperidin-3 -yl)isoindoline- 1,3-dione isolated as the TFA salt (Degronimer 109) (29.0 mg, 53.8 %) as a yellow powder. LC/MS (ES+): m/z 425.3 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.10 (s, 1H), 8.60 (s, 1H), 8.47 (d, J = 5.4 Hz, 1H), 8.31 (s, 1H), 7.94 (d, J = 5.5 Hz, 2H), 7.32 (d, J = 6.8 Hz, 1H), 7.07 (dd, J = 8.5, 3.1 Hz, 1H), 7.00 (d, J = 7.0 Hz, 1H), 5.03 (dd, J = 12.7, 5.4 Hz, 1H), 4.74 (s, 1H), 4.69 - 4.54 (m, 2H), 4.07 (d, J = 7 .1Hz,

3H), 3.32 - 3.06 (m, 4H), 2.35 - 2.23 (m, 2H), 1.50 - 1.1 1 (m, 28H), 0.73 - 0.45 (m, 3H), 0.36 - 0.23 (m, 2H).

Example 113. Preparation of N -(ll -((R)-4-((7-(3-(Cyclopropylmethoxy)pyridin-4-yl)-5- methyl-4-oxo-4,5-dihydrofuro[3,2-c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-ll- oxoundecyl)-2-((2-(2,6-dioxopiperidin-3-yl)-l,3-dioxoisoindolin-4-yl)oxy)acetamide (Degronimer 110) Scheme 328

HATU (31.3 mg, 82.6 µ οΐ) was added to a solution of 2-((2-(2,6-dioxopiperidin-3-yl)- l,3-dioxoisoindolin-4-yl)oxy)acetic acid (208-2) (27.4 mg, 82.6 µιηοΐ) and N-ethyl-N- isopropylpropan-2-amine (35.0 , 190 µ οΐ) in DMF (636 µΙ_, ) . After stirring for 10 min (R)-2- ((4-(l l-aminoundecanoyl)-2-methylpiperazin-l-yl)methyl)-7-(3-(cyclopropylmethoxy)pyridin-4- yl)-5-methylfuro[3,2-c]pyridin-4(5H)-one hydrochloride (311-1) (.04 g, 63.6 µηιοΐ) was added and the reaction stirred for 2 hrs, diluted with DMSO and purified by reverse phase HPLC to give N-(l l-((R)-4-((7-(3-(cyclopropylmethoxy)pyridin-4-yl)-5-methyl-4-oxo-4,5-dihydrofuro[3,2- c]pyridin-2-yl)methyl)-3-methylpiperazin-l-yl)-l l-oxoundecyl)-2-((2-(2,6-dioxopiperidin-3-yl)- l,3-dioxoisoindolin-4-yl)oxy)acetamide isolated as the TFA salt (Degronimer 110) (33.0 mg, 57.2 %) as a white powder. LC/MS (ES+): m/z 906.8 [M + H]+ ¾ MR (400 MHz, DMSO-de) δ 11.12 (s, 1H), 8.53 (s, 1H), 8.37 (d, J = 5.1 Hz, 1H), 8.19 (s, 1H), 7.93 (t, J = 5.8 Hz, 1H), 7.84 - 7.74 (m, 1H), 7.74 - 7.66 (m, OH), 7.48 (d, J = 7.3 Hz, 1H), 7.37 (d, J = 8.6 Hz, 1H), 7.28 (d, J = 7.1 Hz, 1H), 5.1 1 (dd, J = 12.9, 5.4 Hz, 1H), 4.75 (s, 2H), 4.58 (d, J = 22.9 Hz, 1H), 4.03 (d, J = 6.9 Hz, 2H), 3.59 (s, 3H), 3.1 1 (q, J = 6.7 Hz, 2H), 2.96 - 2.79 (m, 1H), 2.67 - 2.52 (m, 1H), 2.28 (t, J = 7.6 Hz, 2H), 2.13 - 1.95 (m, 1H), 1.47 - 1.27 (m, 7H), 1.28 - 1.09 (m, 14H), 0.50 (d, J = 7.8 Hz, 2H), 0.31 - 0.16 (m, 2H).

Example 114. Preparation of 3-(5-(4-(7-(4-(6-(4-c Chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3-a][l,4]diazepine-4,l'- cyclopropan]-8-yl)-lH-pyrazol-l-yl)heptyl)piperazine-l-carbonyl)-2-oxopyridin-l(2H)- yl)piperidine-2,6-dione (Degronimer 111) Scheme 329. In a round bottom flask, l-(2,6-dioxopiperidin-3-yl)-6-oxo-l,6-dihydropyridine-3- carboxylic acid (329-1) (64.3 mg, 0.2571 mmol) and 6-(4-chlorophenyl)-l-methyl-8-(l-(7- (piperazin- 1-yl)heptyl)- lH-pyrazol-4-yl)spiro[benzo[f] [1,2,4]triazolo[4,3 -a] [1,4]diazepine-4, 1'- cyclopropane] (329-2) (100 mg, 0.1714 mmol) was dissolved in 5 mL dry DMF under inert atmosphere at 0°C. HATU (97.7 mg, 0.2571 mmol) was added followed by DIEA (89.4

µΐ 0.5142 mmol). The reaction mixture was stirred at room temp for overnight and LCMS indicate limiting starting material was completely consumed. The reaction mixture was diluted with 50 mL EtOAc, wash with water 50 mL three times and dry the organic phase with anhyd. MgS04. The solvent was removed under reduced pressure and oil residue was subjected to Prep HPLC to afford 3-(5-(4-(7-(4-(6-(4-chlorophenyl)-l-methylspiro[benzo[f][l,2,4]triazolo[4,3- a][1,4]diazepine-4, 1'-cyclopropan]-8-yl)- lH-pyrazol- 1-yl)heptyl)piperazine- 1-carbonyl)-2- oxopyridin-l(2H)-yl)piperidine-2,6-dione (Degronimer 111) (95.0 mg, 0.1 165 mmol, 68.3 %) as a white solid. LC/MS (ES+): m/z 815 [M + H]+ H MR (400 MHz, DMSO-de) δ 11.22 (s, 1H), 8.21 (s, 1H), 7.91 (dd, J = 13.3, 7.4 Hz, 3H), 7.73 (d, J = 8.5 Hz, 1H), 7.58 (d, J = 8.3 Hz, 2H), 7.47 (q, J = 10.4, 8.9 Hz, 4H), 6.44 (d, J = 9.5 Hz, 1H), 5.41 (br, 1H), 4.06 (t, J = 6.9 Hz, 2H), 3.60 - 3.42 (m, 5H), 2.49 (s, 2H), 2.29 (dt, J = 36.4, 6.0 Hz, 6H), 2.03 (dq, J = 11.3, 5.9, 5.3 Hz, 1H), 1.74 (p, J = 7.5, 7.1 Hz, 3H), 1.38 (p, J = 5.5, 4.7 Hz, 3H), 1.32 - 1.08 (m, 6H), 0.76 (dp, J = 17.3, 5.5 Hz, 2H).

Example 115. Cell Viability Assay of Representative Compounds of the Present Invention.

RPMI 1640 medium and fetal bovine serum (FBS) were purchased from Gibco (Grand Island, NY, USA). CellTiter-Glo® 2.0 Assay was purchased from Promega (Medison, WI, USA). MOLT4. 1 (WT) cell line was purchased from ATCC (Manassas, VA, USA) and MOLT4.2 (CRBN Knock Out) cell line was generated in house. Cell culture flasks and 384-well microplates were acquired from VWR (Radnor, PA , USA). MOLT4.1 and MOLT4.2 cell viability was determined based on quantification of ATP using CellTiter-Glo® 2.0 luminescent Assay kit, which signals the presence of metabolically- active cells. Briefly, MOLT4.1 and MOLT4.2 cells were seeded into 384-well plates at a cell density of 750 cells per well, the plates were kept at 37 °C with 5% C02 overnight. On the following day, test compounds were added to the cells from a top concentration of 1 µΜ with 10 points, half log titration in duplicates. The cells treated in the absence of the test compound were the negative control and the cells treated in the absence of CellTiter-Glo® 2.0 were the positive control. At the same day of compound treatment, CellTiter-Glo® 2.0 was added to a plate with cells treated in the absence of the test compound to establish Cytostatic control value (CT0). Cells treated with the test compound were incubated for 72 hr. CellTiter-Glo reagent was then added to the cells and Luminescence was acquired on EnVision™ Multilabel Reader (PerkinElmer, Santa Clara, CA, USA). Results of the assays for representative Targeting Ligands (Degron) compounds are provided in Table 1. Results of the assays for representative Degronimer compounds are provided in Table 2 . Table 1 - Cell Viability Assay of Representative Targeting Ligands (Degrons).

Table 2 - Cell Viability Assay of Representative Degronimers. 650

652 653 654 655 656 657 658 659

661 662 663

665 666

668 669 670

672 673 674 675 676

680

682 683 684 685 686

Ķ88

690 691 692

694 695 696

Ķ99

704 705 All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to one of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the invention as defined in the appended claims. We claim:

1. A com ound of Formula I, II, III, IV, V, VI, VII, VIII, IX, or X : or a pharmaceutically acceptable salt, N-oxide or isotopic derivative; wherein: A is independently selected from C(R ) and N wherein the number of nitrogen atoms is 0,

1, 2, 3, or 4 in the ring, which can be a pyridine, diazine, triazine, pyrimidine, pyridazine, pyrazine, triazine or tetrazine;

n is 0, 1, 2, 3, 4, 5, 6 or 7; is a single or double bond;

X is independently selected from C(R ), N, N(R9), N(O), O, and S; X' is selected from O, S, and -N(R9); 9 Y is selected from CH(R ), C(R )2, O, S, H, N(R ), P=0(aliphatic), P=0(Oaliphatic),

P=0(Oaryl), P=0(OH), P=0(NHaliphatic), P=0(NHaryl or heteroaryl), P=0 ( H2), S(0) 2 and S(O); Q is either C or N; T is hydrogen, deuterium, fluorine, chlorine, bromine, iodine, or or T and R together with the carbon to which they are attached form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle; or T and R form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle that is substituted with R , SChheterocycle, or SCtealkyl; W is selected from C(R ) and N; R2 and R3 are independently selected from hydrogen, deuterium, alkyl, hydroxyl, alkoxy, aliphaticoxy, amine, -NH(aliphatic), -N(aliphatic)2, fluorine, chlorine, bromine, iodine, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heteroaliphatic, aliphatic, and carbocyclic; R , R2', R4, R4', R5, R , R6, R6', R7, R , R 3, and R 13' are independently selected from hydrogen, deuterium, alkyl, aliphatic, hydroxyl, alkoxy, aliphaticoxy, amine, cyano, - NH(aliphatic), -N(aliphatic)2, fluorine, chlorine, bromine, iodine, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclo and carbocyclic;

or two R substituents can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

4 or two R substituents can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 5 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 6 6 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; 7 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

3 or R and R can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O;

or two R4 substituents can form a 1, 2, 3 or 4 carbon bridged ring; or R4 and R can form a 3, 4, 5, or 6 membered fused ring; or R and R6 can form a 3, 4, 5, or 6 membered fused ring;

or R4' and R can form a 1, 2, 3, or 4 carbon bridged ring;

or R4' and R6 can form a 1, 2, 3, or 4 carbon bridged ring; 8 9 9 R is N(R ), CH2, CHR , C(R )2, O, or S; R9 is selected at each instance from hydrogen, deuterium, alkyl, -C(0)H, -C(0)OH, - C(0)(aliphatic), -C(0)0(aliphatic), alkenyl, and alkynyl; aiyl, heteroaiyl, heterocyclic, aliphatic and heteroaliphatic; R 0 is Linker-Degron; R and R 11A are independently selected at each instance from hydrogen, alkyl, alkenyl, alkynyl, fluorine, chlorine, bromine, iodine, hydroxyl, alkoxy, cyano, azide, amino, -

NH(aliphatic), -N(aliphatic) 2, -NHS0 2(aliphatic), -N(aliphatic)S0 2(aliphatic), -NHS0 2(aryl or heteroaiyl), -N(aliphatic)S0 2aryl, -NHS0 2alkenyl, -N(alkyl)S0 2alkenyl, -NHS0 2alkynyl, -

N(alkyl)S0 2alkynyl, cyano, nitro, nitroso, -SH, -Salkyl, -S(0)alkyl, -S(0) 2alkyl, -C(0)H, - C(0)OH, -C(0)alkyl, -C(0)Oalkyl, aliphatic, heteroaliphatic, aiyl, heteroaiyl, carbocyclic, and haloalkyl; Linker is a chemical group that attaches the Degron to a Targeting Ligand; Targeting Ligand is selected from those in FIG 1A - FIG 8PPPPP; CIV: wherein:

W 2oi i s C R 206R 207 c=0 c = C=CH2 S02 S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; 202 0 209 W is CR R , C=0, C=S, C=CH 2, SO2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; 200 203 203 0 X is independently selected from NH, R , CH2, CHR , C(R )2, O, and S; n200 is 0, 1, 2, or 3; is a single or double bond;

wherein when represents a single bond, n is 0, 1, 2, or 3;

wherein when represents a double bond, n is 0, 1, or 2;

1 712 R202 is alkyl or hydrogen; or R201 and R202 are combined to form a 4, 5, 6, 7, 8, 9, or 10 membered heterocyclo or heteroaryl species, wherein the heterocyclo or heteroaryl species is substituted with R212 at any desired position, wherein the heterocyclo or heteroaryl species is optionally further substituted with one or more substituents selected from R

R is selected at each instance from: alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, alkene, alkyne, aliphatic, heteroaliphatic, aryl, heteroaryl or heteroalkyl; R204 is selected at each instance from: alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -NHalkyl, -N(alkyl)2, - H S0 2alkyl, -N(alkyl)S0 2alkyl, - H S0 2aryl, -

N(alkyl)S0 2aryl, - H S0 2alkenyl, -N(alkyl)S0 2alkenyl, - HS0 2alkynyl, -N(alkyl)S0 2alkynyl, and haloalkyl; and in addition to these can also be selected from aliphatic, heteroaliphatic, aryl, heteroaryl and heteroalkyl; or two R substituents together with the carbon atom(s) to which they are bound can form a 3, 4, 5 or 6 membered ring; R205 and R214 are selected at each instance from: hydrogen, alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -NHalkyl, -N(alkyl)2, -NHSChalkyl, -N(alkyl)S0 2alkyl,

-NH S aryl, -N(alkyl)S0 2aryl, - H S0 2alkenyl, -N(alkyl)S0 2alkenyl, - H S0 2alkynyl,

-N(alkyl)S0 2alkynyl, and haloalkyl; and in addition to these can also be selected from aliphatic, heteroaliphatic, aryl, heteroaryl or heterocyclic; or in the alternative, R205 is independently selected from C(0)R 204, cyano, aryl, aryloxy, heterocyclo, heteroaryl, arylalkyl, alkoxy, hydroxyl, O-aiylalkyl, or cycloalkyl; each of which R205 can be optionally substituted, for example, with one or more substituents selected from alkyl, alkene, alkyne, halogen, hydroxyl, alkoxy, azide, amino, -

204 NHalkyl, -N(alkyl)2, C(0)R , aryl, heterocyclo, heteroaryl, haloalkyl, and cycloalkyl; R206, R207, R208, R209, R210, and R211, are independently selected from hydrogen, alkyl, hydroxyl, alkoxy, amine, -NHalkyl, and -Nalkyl2; or R206 and R207 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R208 and R209 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R210 and R211 together with the carbon to which they are bound form a 3-, 4-, 5-, or 6- membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; or R206 and R208 form a 1 or 2 carbon bridged ring; or R206 and R210 form a 1 or 2 carbon bridged ring; or R208 and R210 form a 1 or 2 carbon bridged ring; or R214 and R206 form a 3, 4, 5, or 6 carbon fused ring; or R214 and R210 form a 3, 4, 5, or 6 carbon fused ring; or R214 and R208 form a 1 or 2 carbon bridged ring; or R214 and R204 form a 3, 4, 5, or 6 carbon fused ring wherein R205 is on the carbon alpha to R214 or a 1, 2, 3, or 4 carbon bridged ring wherein R205 is not on the carbon alpha to R214; R212 is the point at which the linker is attached; R222 is independently selected at each occurrence from hydrogen, alkyl, alkene, alkyne, haloalkyl, alkoxy, hydroxyl, aryl, heteroaryl, heterocycle, arylalkyl, heteroarylalkyl, heterocycloalkyl, aryloxy, heteroaryloxy, CN, -COOalkyl, COOH, NO2, F, CI, Br, I, CF3, NH2,

225 225 204 NHalkyl, N(alkyl) 2, NR R , NHR , aliphatic, heteroaliphatic, and COR ; R225 is selected at each instance from: alkyl, -C(0)H, -C(0)OH. - C(0)alkyl, -C(0)Oalkyl, alkenyl, or alkynyl or alternatively can be aliphatic, heteroaliphatic, aryl, heteroaryl or heteroc clic;

when R212 is bonded to a A that is carbon, then A is CR212; 401 401 402 W is CR R , C=0, C=S, C=CH2, S0 2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; 402 40 404 W is CR R , C=0, C=S, C=CH2, SO2, S(O), P(0)Oalkyl, P(0)NHalkyl,

P(0)N(alkyl) 2, P(0)alkyl, P(0)OH, P(0 ) H2; 400 412 412 412 X is independently selected from NH, R , CH2, CHR , C(R )2, O, or S; n200 is 0, 1, 2, or 3;

=-=-= is a sin le or double bond; 717 A is independently selected from C(R ), and N ;

Z20 is H, O, S, or R 412; and when R410 is bonded to a A that is carbon, then A is CR410, and when R410 is bonded to a Z20 or Z402 that is nitrogen, then Z20 or Z402 is R 410, etc; R is independently selected at each instance from hydrogen, alkyl, alkenyl, alkynyl, fluorine, chlorine, bromine, iodine, hydroxyl, alkoxy, cyano, azide, amino, -NH(aliphatic), -

N(aliphatic)2, -NHS02(aliphatic), -N(aliphatic)S02(aliphatic), - HS0 2(aryl or heteroaryl), -

N(aliphatic)S0 2aryl, -NHSChalkenyl, -N(alkyl)S0 2alkenyl, - H S0 2alkynyl,

N(alkyl)S0 2alkynyl, cyano, nitro, nitroso, -SH, -Salkyl, -S(0)alkyl, -S(0) 2alkyl, -C(0)H, - C(0)OH, -C(0)alkyl, -C(0)Oalkyl, aliphatic, heteroaliphatic, aryl, heteroaryl, carbocyclic, and haloalkyl; R401, R402, R403, R404, R407, R408, and R415 are independently selected from hydrogen, alkyl, aliphatic, heteroaliphatic, heterocyclic, carbocyclic, aryl, heteroaryl, hydroxyl, halo, azide, CN-, alkoxy, amine, -NHalkyl, and -Nalkyh, -NH(aliphatic), and -N(independently aliphatic) 2; or R401 and R402 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R403 and R404 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R407 and R408 together with the carbon to which they are attached form a 3-, 4-, 5-, or 6- membered spiro-carbocycle, or a 4-, 5-, or 6-membered spiro-heterocycle comprising 1 or 2 heteroatoms selected from N and O; or R401 and R403 form a 1 or 2 carbon bridged ring; or R401 and R407 form a 1 or 2 carbon bridged ring; or R403 and R407 form a 1 or 2 carbon bridged ring; or R415 and R401 form a 3, 4, 5, or 6 carbon fused ring; or R415 and R407 form a 3, 4, 5, or 6 carbon fused ring; or R415 and R403 form a 1 or 2 carbon bridged ring; or R 4 and R405 form a 3, 4, 5, or 6 carbon fused ring wherein R405 is on the carbon alpha to R415 or a 1, 2, 3, or 4 carbon bridged ring wherein R405 is not on the carbon alpha to R415 ; R is selected at each instance from: alkyl, alkene, alkyne, aliphatic, heteroaliphatic, heterocyclic, aryl, heteroaryl, halogen, hydroxyl, alkoxy, azide, amino, -NH(aliphatic), - N(independently aliphatic)2, -NHS02(aliphatic), -N(aliphatic)S02(aliphatic), -NHS02aryl, -

N(aliphatic)S0 2aryl, - H S0 2alkenyl, -N(aliphatic)S0 2alkenyl, - H S0 2alkynyl, N(aliphatic)S02alkynyl, and halo(aliphatic); or two R405 substituents together with the carbon atom(s) to which they are bound can form a 3, 4, 5 or 6 membered ring; R410 is the point at which the linker is attached; R411 is selected at each instance from: hydrogen, alkyl, alkenyl, alkynyl, halogen, hydroxyl, heterocyclic, heteroalkyl, carbocyclic, heteroaliphatic, aliphatic, alkoxy, aryl, heteroaryl, alkylamino, alkylhydroxyl, -NHalkyl, -Nalkyh, -NH(aliphatic), -N(independently aliphatic)2, amino, cyano, nitro, nitroso, sulfone, sulfoxide, thioalkyl, thiol and haloalkyl; R412 is selected at each instance from: hydrogen, alkyl, aliphatic, heteroaliphatic, heterocyclic, heteroaryl, aryl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, -C(0)(aliphatic, aryl, heteroaliphatic, aryl or heteroaryl), -C(0)0(aliphatic, aryl, heteroaliphatic, aryl or heteroaryl), alkene, and alkyne; R413 and R414 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, alkoxy, haloalkoxy, hydroxy, amino, -NHalkyl, and -N(alkyl)2; or R413 and R414 together with the carbon atom to which they are attached, form C(O), C(S), C=CH2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle com ri sing 1 or 2 heteroatoms selected from N and O . 2 . A compound of Formula XI, Formula XII, Formula XIII, Formula XIV, Formula XV, Formula XVI:

or a pharmaceutically acceptable salt, N-oxide or isotopic derivative; =-=-= is a single or double bond; Q is either C or N;

T is hydrogen, deuterium, fluorine, chlorine, bromine, iodine, or ; or T and R together with the carbon to which they are attached form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle; or T and R form a 3, 4, 5, or 6 membered spirocarbocycle or a 3, 4, 5, or 6 membered spiroheterocycle that is substituted with R , SChheterocycle, or SCtealkyl; W is selected from C(R ) and N; 2 9 W is selected from C(R )2 and N(R ); R is independently selected from hydrogen, deuterium, alkyl, hydroxyl, alkoxy, amine, -

NH(aliphatic), -N(aliphatic) 2, fluorine, chlorine, bromine, iodine, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, heterocyclo, aliphatic, heteroaliphatic,

or two R substituents can form C=0, C=S, C=CH 2, a 3-, 4-, 5-, or 6-membered spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O; and the other variables are as described in claim 1. 3 . A compound of Formula XVII, Formula XVIII, Formula XIX, Formula XX, Formula XXI, Formula XXII, Formula XXIII, Formula XXIV, Formula XXV, Formula XXVI, Formula XXVII, Formula XXVIII, or Formula XXIX: or a pharmaceutically acceptable salt, N -oxide or isotopic derivative; wherein:

X 11 is bond, CH2 or CH(aliphatic), and the other variables are as defined in claim 1.

4 . The compound of claim 1, 2 or 3 wherein the Linker has a chain of 2 to 20 carbon

atoms of which one or more carbons can be replaced by a heteroatom such as O, N ,

S, or P or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 ethylene glycol units.

5. The compound of claim 1, 2 or 3 wherein Linker is a moiety selected from Formula LI, Formula LII, Formula LIII, Formula LIV, Formula LV, Formula LVI, and Formula LVII:

x r 24 22 .HHeetteerrooaarryyll — X '2

x 3 2 (LII),

- χ 23 Heieroaryi R2 X2 (LIII),

R ¾ / \ ,' ' ' X R R X (LVI), and wherein:

1 2 25 25 X and X are independently selected from bond, H, , CH2, CHR , C(R )2, O, and S; R20, R2 , R22, R23, and R24 are independently selected from bond, alkyl, -C(O)- -C(0)0-, - OC(O)-, -C(0)alkyl, -C(0)Oalkyl, -C(S)-, -SO2-, -S(O)-, -C(S)-, -C(0)NH-, -NHC(O)-, - N(alkyl)C(0)-, -C(0)N(alkyl)-, -0-, -S-, -NH-, -N(alkyl)-, -CH(-0-R 26)-, -CH(-NHR 25)-, -CH(-

6 H 2)-, -CH(-NR 2)-, -C(-0-R )alkyl, -C(-NHR )alkyl-, -C(- H2)alkyl-, -C(- R 2)alkyl-, - C(R4R4)-, -alkyl(R )-alkyl(R 28)-, -C(R R28)-, -P(0)(OR 6)0-, -P(0)(OR 26)-, - HC(0 ) H-, - N(R )C(0)N(R 25)-, -N(H)C(0)N(R 25), polyethylene glycol, poly(lactic-co-glycolic acid), alkene, haloalkyl, alkoxy, alkyne, heteroarylalkyl, aryl, arylalkyl, heterocycle, aliphatic, heteroaliphatic, heteroaryl, polypropylene glycol, lactic acid, glycolic acid, carbocycle, or -0-(CH 2)i-i2-0-, - H-

(CH2)i-i2-NH-, -NH-(CH 2)i-i2-0-, or -0-(CH 2)i-i2-NH-, -S-(CH2)i-i2-0-, -0-(CH 2)i-i2-S-, -S-

(CH2)i-i2-S-, -S-(CH2)i-i2-NH- or -NH-(CH 2)i-i2-S-; R25 is selected at each instance from: alkyl, -C(0)H, -C(0)OH, -C(0)alkyl, -C(0)Oalkyl, alkenyl, or alkynyl or alternatively can be aliphatic, heteroaliphatic, aryl, heteroaryl or heterocyclic; R26 is hydrogen, alkyl, silane, arylalkyl, heteroarylalkyl, alkene, and alkyne; or in addition to these can also be selected from aryl, heteroaryl, heterocyclic, aliphatic and heteroaliphatic; and R27 and R28 are independently selected from hydrogen, alkyl, amine, or together with the carbon atom to which they are attached, form C(O), C(S), C=CH2, a C 3- C 6 spirocarbocycle, or a 4-, 5-, or 6-membered spiroheterocycle comprising 1 or 2 heteroatoms selected from N and O, or form a 1 or 2 carbon bridged ring.

6 . The compound of claim 1, 2 or 3 wherein Linker is a moiety selected from Formula LVIII, LIX, and LX:

(LVIII), R 2 j / \ Heterocycle R X X1 R ' (LIX), and

p22 p20

Heterocycle R23 R X wherein each variable is as defined in claim 5.

7 . The compound of claim 1, 2 or 3, wherein the Linker is selected from 727 8. The compound of claim 1, 2, or 3, wherein the Linker is wherein the variables are as defined in claim 5.

9 . The compound of claim 1, 2, or 3, wherein the Linker is selected from:

6 6 - (CH2 )ni -(lower alkyl)-, - (CH2 )ni -(lower alkoxyl)-,

6 )ni 6 )ni - (CH2 -(lower alkoxyl)-OCH2-, - (CH2 -(lower alkoxyl)-(lower alkyl)-OCH2-,

6 )ni 6 )ni (CH2 -(cycloalkyl)-(lower alkyl)-OCH2-, - (CH2 -(heterocycloalkyl)-, 6 6 (CH2CH20 )ni -(lower alkyl)-0-CH 2-, - (CH2CH20 )ni -(heterocycloalkyl)-0-CH 2-, 6 6 - (CH2CH20 )ni -Aryl-0-CH 2-, - (CH2CH20 )ni -(heteroaryl)-0-CH 2-, 6 - (CH2CH20 )ni -(cycloalkyl)-0-(heteroaiyl)-0-CH 2-, 6 - (CH2CH20 )ni -(cycloalkyl)-0-Aryl-0-CH 2-, 6 - (CH2CH20 )ni -(lower alkyl)-NH-Aryl-0- CH2-, 6 - (CH2CH20 )ni -(lower alkyl)-0-Aryl-CH 2, 6 - (CH2CH20 )ni -cycloalkyl-0-Aryl-, 6 - (CH2CH20 )ni -cycloalkyl-0-heteroaryl-,

6 )ni - (CH2CH2 -(cycloalkyl)-0-(heterocycle)-CH 2,

6 )ni 6 - (CH2CH2 -(heterocycle)-(heterocycle)-CH 2, and - -(heterocycle)-CH2; wherein nl is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and R6 is H, methyl, or ethyl. 10. The compound of claim 1, 2, or 3, wherein the Linker is selected from:

{CH2) O(GH ) 2OCH

732 1 734 735 736 737 738 739 740 741 742

745

7 is -0-, - H, - Me, -Nalkyl, N(aliphatic), -N(heteroaliphatic);

ml, n2, ol, pi, q2, and rl are independently 1, 2, 3, 4, or 5.

11. A method for treating a patient with a medical disorder that can be treated by degrading a Target Protein that binds to a Targeting Ligand, comprising administering an effective amount of a compound of claims 1-10, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. 12. A method for treating a patient with a medical disorder that can be treated by binding to the protein cereblon in vivo, comprising administering an effective amount of a compound of claims 1-10, or a pharmaceutically acceptable salt thereof, optionally in a pharmaceutically acceptable carrier. 13. The method of claim 11 or 12, wherein the disorder is selected from abnormal cellular proliferation, a tumor, cancer, an immune disorder, autoimmune disorder, arthritis, lupus, diabetes, cardiovascular disease, an infectious disease, or an inflammatory condition. 14. The method of claim 11 or 12, wherein the infectious disease is selected from HIV, HBV, HCV, HSV, HPV, RSV, CMV, Ebola, Flavivirus, Pestivirus, Rotavirus, Influenza, Coronavirus, EBV, viral pneumonia, drug-resistant viruses, Bird flu, RNA virus, DNA virus, adenovirus, poxvirus, Picornavirus, Togavirus, Orthomyxovirus, Retrovirus or Hepadnovirus, a Gram- negative, Gram-positive, Atypical, Staphylococcus, Streptococcus, E . Coli, Salmonella, Helicobacter pylori, meningitis, gonorrhea, Chlamydiaceae, Mycoplasmataceae, fungus, protozoa, helminth, worms, prion or parasite. 15. The method of claim 11 or 12, wherein the disorder is a cancer selected from the group consisting of squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinoma, renal cell carcinoma, cancer of the bladder, bowel, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreatic, prostate, stomach, leukemia, lymphoma, Burkitt's lymphoma, Non-Hodgkin's lymphoma; melanoma; myeloproliferative disease; sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcoma, peripheral neuroepithelioma, synovial sarcoma, glioma, astrocytoma, oligodendroglioma, ependymoma, glioblastoma, neuroblastoma, ganglioneuroma, ganglioglioma, medulloblastoma, pineal cell tumor, meningioma, meningeal sarcoma, neurofibroma, and Schwannoma; breast cancer, uterine cancer, testicular cancer, thyroid cancer, astrocytoma, esophageal cancer, carcinosarcoma, Hodgkin's disease, Wilms' tumor and teratocarcinoma. 16. A pharmaceutical composition comprising an effective amount of a compound of claims 1-10 in a pharmaceutically acceptable carrier.

INTERNATIONAL SEARCH REPORT International application No. PCT/US 17/32053

A . CLASSIFICATION O F SUBJECT MATTER IPC(8) - A61 K47/48; C07D 471/04; C07D 471/14 (201 7.01 ) CPC - C07D471/04; C07D471/14; A61 K47/48023; A61 K31/551

According to International Patent Classification (IPC) or to both national classification and IPC B. FIELDS SEARCHED Minimum documentation searched (classification system followed by classification symbols) See Search History Document Documentation searched other than minimum documentation to the extent that such documents are included in the fields searched See Search History Document Electronic data base consulted during the international search (name of data base and, where practicable, search terms used) See Search History Document

C. DOCUMENTS CONSIDERED TO BE RELEVANT Category* Citation of document, with indication, where appropriate, of the relevant passages Relevant to claim No.

US 2016/0058872 A 1 (CREW et al.) 03 March 2016 (03.03.2016) para [01 14], [0125]-[0129], 1-7, 9-10 [0205]-[0207], [0210], [0219] 8

US 2013/0190340 A 1 (HEDSTROM at al.) 25 July 2013 (25.07.2013) para [0009], [0253], 8 [0353], Fig 14

"DAWSON et al. 'Inhibition of BET recruitment to chromatin as a n effective treatment for MLL- 1-10 fusion leukaemia', Nature, 201 1, Vol.478(7370), pp 529-533. Entire Document, esp. Fig 1

US 2016/0016966 A 1 (GLAXOSMITHKLINE INTELLECTUAL PROPERTY (NO.2) LIMITED) 2 1 1-10 January 2016 (21 .01 .201 6) para [0596], Example 20; para [0662], [0667]

hALBRECHT et al. 'Identification of a Benzoisoxazoloazepine Inhibitor (CPI-0610) of the Bromodomain and Extra-Terminal (BET) Family as a Candidate for Human Clinical Trials', J . 1-10 Med. Chem. 01.2016, Vol.59, pp.1330-1339. [Published January 2016] abstract; pg 1332, Table 2, compounds 3-21

J--ZENGERLE et al. 'Selective Small Molecule Induced Degradation of the BET Bromodomain 1-10 Protein BRD4' ACS Chem. Biol. 2015, Vol.10, pages 1770-1777. pg 1771, Fig 1

I Further documents are listed in the continuation of Box C. | | See patent family annex.

• Special categories of cited documents: "T" later document published after the international filing date or priority "A" document defining the general state of the art which is not considered date and not in conflict with the application but cited to understand to be of particular relevance the principle or theory underlying the invention "E" earlier application or patent but published on or after the international "X" document of particular relevance; the claimed invention cannot be filing date considered novel or cannot be considered to involve an inventive "L" document which may throw doubts on priority claim(s) or which is step when the document is taken alone cited to establish the publication date of another citation or other "Y" document of particular relevance; the claimed invention cannot be special reason (as specified) considered to involve an inventive step when the document is "O" document referring to an oral disclosure, use, exhibition or other combined with one or more other such documents, such combination means being obvious to a person skilled in the art "P" document published prior to the international filing date but later than "&" document member of the same patent family the priority date claimed Date of the actual completion of the international search Date of mailing of the international search report 05 August 2017 0 6 S EP 2017

Name and mailing address of the ISA/US Authorized officer: Mail Stop PCT, Attn: ISA/US, Commissioner for Patents Lee W. Young P.O. Box 1450, Alexandria, Virginia 22313-1450 Facsimile No. 571-273-8300 Form PCT/lSA/210 (second sheet) (January 2015) INTERNATIONAL SEARCH REPORT International application No. PCT/US 17/32053

Box No. II Observations where certain claims were found unsearchable (Continuation of item 2 of first sheet)

This international search report has not been established in respect of certain claims under Article 17(2)(a) for the following reasons:

Claims Nos.: because they relate to subject matter not required to be searched by this Authority, namely:

□ Claims Nos.: because they relate to parts of the international application that do not comply with the prescribed requirements to such an extent that no meaningful international search can be carried out, specifically:

3. Claims Nos.: 11-16 because they are dependent claims and are not drafted in accordance with the second and third sentences of Rule 6 .4(a).

Box No. Ill Observations where unity of invention is lacking (Continuation of item 3 of first sheet)

This International Searching Authority found multiple inventions in this international application, as follows:

1. I I As all required additional search fees were timely paid by the applicant, this international search report covers all searchable claims.

2. I As all searchable claims could be searched without effort justifying additional fees, this Authority did not invite payment of additional fees.

3. □ As only some of the required additional search fees were timely paid by the applicant, this international search report covers only those claims for which fees were paid, specifically claims Nos.:

No required additional search fees were timely paid by the applicant. Consequently, this international search report is restricted to the invention first mentioned in the claims; it is covered by claims Nos.:

The additional search fees were accompanied by the applicant's protest and, where applicable, the payment of a protest fee. The additional search fees were accompanied by the applicant's protest but the applicable protest fee was not paid within the time limit specified in the invitation. No protest accompanied the payment of additional search fees.

Form PCT/ISA/210 (continuation of first sheet (2)) (January 2015)