US20190255041A1 - Compositions and methods for treating ezh2-mediated cancer - Google Patents

Compositions and methods for treating ezh2-mediated cancer Download PDF

Info

Publication number
US20190255041A1
US20190255041A1 US16/345,591 US201716345591A US2019255041A1 US 20190255041 A1 US20190255041 A1 US 20190255041A1 US 201716345591 A US201716345591 A US 201716345591A US 2019255041 A1 US2019255041 A1 US 2019255041A1
Authority
US
United States
Prior art keywords
mmol
ezh2
cancer
methyl
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/345,591
Other languages
English (en)
Inventor
Jian Jin
Ramon Parsons
Ilias Stratikopoulos
Xiaobao Yang
Anqi Ma
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Icahn School of Medicine at Mount Sinai
Original Assignee
Icahn School of Medicine at Mount Sinai
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Icahn School of Medicine at Mount Sinai filed Critical Icahn School of Medicine at Mount Sinai
Priority to US16/345,591 priority Critical patent/US20190255041A1/en
Publication of US20190255041A1 publication Critical patent/US20190255041A1/en
Assigned to ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI reassignment ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PARSONS, RAMON, JIN, JIAN, MA, ANQI, STRATIKOPOULOS, Ilias, YANG, XIAOBAO
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • This disclosure relates to compositions and methods for administering one or more bivalent compounds which selectively degrade/disrupt enhancer of zeste homologue 2 (EZH2) to a subject for the treatment of EZH2-mediated cancer, and to methods for designing such degraders/disruptors.
  • EZH2 zeste homologue 2
  • EZH2 (enhancer of zeste homolog 2) is the main catalytic subunit of the polycomb repressive complex 2 (PRC2) that catalyzes methylation of histone H3 lysine 27 (H3K27) (Cao et al., 2002; Czermin et al., 2002; Kuzmichev et al., 2002; Muller et al., 2002).
  • PRC2 polycomb repressive complex 2
  • H3K27 histone H3 lysine 27
  • H3K27me3 The trimethylation of H3K27 (H3K27me3) is a transcriptionally repressive epigenetic mark that regulates gene expression, differentiation, and development.
  • Dysregulation of EZH2, other PRC2 components (e.g., EED and SUZ12), and/or H3K27 trimethylation have been associated with a number of cancers.
  • EZH2 is overexpressed in a broad spectrum of cancers, including prostate cancer, breast cancer, myeloma, and lymphoma.
  • High EZH2 expression correlates with poor prognosis (Bachmann et al., 2006; Bodor et al., 2011; Bracken et al., 2003; Kim and Roberts, 2016; Kleer et al., 2003; Morin et al., 2010; Sauvageau and Sauvageau, 2010; Varambally et al., 2002).
  • Hyper-trimethylation of H3K27 catalyzed by PRC2 drives tumorigenesis and progression of cancers including diffused large B cell lymphoma (DLBCL) and malignant rhabdoid tumor (MRT) (Majer et al., 2012; McCabe et al., 2012a; Sneeringer et al., 2010).
  • DLBCL diffused large B cell lymphoma
  • MRT malignant rhabdoid tumor
  • EZH2 inhibitors which effectively inhibit the methyltransferase activity of EZH2, display robust antiproliferative activity in DLBCL and MRT cellular and animal models (Kaniskan et al., 2017; Wang et al., 2015; Xu et al., 2015).
  • TNBC triple-negative breast cancer
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2 human epidermal growth factor receptor 2
  • EZH2 Overexpression of EZH2 has been identified as a major driver for breast cancer development and progression (Bachmann et al., 2006; Bracken et al., 2003; Chang et al., 2011; Holm et al., 2012; Fujii et al., 2011; Gonzalez et al., 2014; Kleer et al., 2003; Mahara et al., 2016).
  • EZH2 downregulates the tumor and metastasis suppressor RKIP (Raf-1 kinase inhibitor protein) (Ren et al., 2012), tumor suppressor KLF2 (Kruppel-like factor) (Taniguchi et al., 2012), forkhead box transcription factor FOXC1 (Du et al., 2012), and tumor suppressor RUNX3 (Runt-related transcription factor 3) (Fujii et al., 2008). Knockdown of EZH2 via RNA interference blocks proliferation of breast cancer cells (Fujii et al., 2008; Gonzalez et al., 2008).
  • RKIP Raf-1 kinase inhibitor protein
  • the present disclosure relates generally to bivalent compounds which selectively degrade/disrupt EZH2 (“EZH2 degraders/disruptors”), and to methods for the treatment of EZH2-mediated cancers, which include, but are not limited to, cancers that overexpress EZH2 relative to wild-type tissues of the same species and tissue types, with the EZH2 degraders/disruptors.
  • EZH2 degraders/disruptors include, but are not limited to, cancers that overexpress EZH2 relative to wild-type tissues of the same species and tissue types, with the EZH2 degraders/disruptors.
  • the bivalent compounds disclosed/claimed here can be significantly more effective therapeutic agents than current EZH2 inhibitors, which inhibit the enzymatic activity of EZH2 but do not affect EZH2 protein levels.
  • the present disclosure further provides methods for identifying EZH2 degraders/disruptors as described herein.
  • the document provides a bivalent compound including an EZH2 ligand conjugated to a degradation/disruption tag.
  • the EZH2 ligand can be an EZH2 inhibitor.
  • the EZH2 ligand can, for example, include UNC1999, EPZ005687, EPZ-6438, GSK126, EI1, CPI-1205, GSK343, CPI-360, EPZ011989, N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-1-isopropyl-6-(6-(4-isopropylpiperazin-1-yl)pyridin-3-yl)-1H-indazole-4-carboxamide (“compound 24”) (see, e.g., Yang et al., 2016), 3-chloro-4-(2-cyano-3-(pyridazin-4-yl)phenoxy)-N-(2,2,6,6-tetramethylpiperid
  • the degradation/disruption tag can bind to a ubiquitin ligase (e.g., an E3 ligase such as a cereblon E3 ligase or a VHL E3 ligase) and/or mimic EZH2 protein misfolding.
  • the degradation/disruption tag can include a bulky and/or hydrophobic group.
  • the degradation/disruption tag can, for example, include adamantane, 1-((4,4,5,5,5-pentafluoropentyl)sulfinyl)nonane, pomalidomide, thalidomide, lenalidomide, VHL-1, and analogs thereof.
  • an EZH2 ligand can be conjugated to a degradation/disruption tag through a linker.
  • the linker can, for example, include an acyclic or cyclic saturated or unsaturated carbon, ethylene glycol, amide, amino, ether, or carbonyl containing group.
  • the linker can, for example, include one or more of Formulas I-XIV:
  • any of the above-described bivalent compounds can include, for example, AM16-10A, AM16-11A, AM16-37A, AM16-38A, XY019-43, XY019-44, XY019-079, XY019-080, AM16-91A, AM16-92A, AM16-93A, AM16-97A, AM16-100A, AM16-101A, AM16-102A, AM16-105A, AM16-106A, XY012-120, AM29-21A, AM29-22A, AM29-32A, AM29-33A, AM16-103A, AM29-182A, AM29-55A, AM29-151A, AM29-152A, AM29-137A, AM29-153A, AM29-138A, AM29-154A, AM29-139A, AM29-155A, AM29-170A, AM29-156A, AM29-171A, AM29-157A, AM29-172A, AM29-17
  • a bivalent compound which can include, for example, AM16-10A, AM16-11A, AM16-37A, AM16-38A, XY019-43, XY019-44, XY019-079, XY019-080, AM16-91A, AM16-92A, AM16-93A, AM16-97A, AM16-100A, AM16-101A, AM16-102A, AM16-105A, AM16-106A, XY012-120, AM29-21A, AM29-22A, AM29-32A, AM29-33A, AM16-103A, AM29-182A, AM29-55A, AM29-151A, AM29-152A, AM29-137A, AM29-153A, AM29-138A, AM29-154A, AM29-139A, AM29-155A, AM29-170A, AM29-156A, AM29-171A, AM29-157A, AM29-172A, AM
  • an EZH2-mediated cancer which includes administering to a subject in a subject in need thereof with an EZH2-mediated cancer bivalent compound including an EZH2 ligand conjugated to a degradation/disruption tag.
  • the EZH2-mediated cancer can overexpress EZH2 relative to a wild-type tissue of the same species and tissue type.
  • the EZH2-mediated cancer can express hyper-trimethylated H3K27.
  • the bivalent compound can include, for example, AM16-10A, AM16-11A, AM16-37A, AM16-38A, XY019-43, XY019-44, XY019-079, XY019-080, AM16-91A, AM16-92A, AM16-93A, AM16-97A, AM16-100A, AM16-101A, AM16-102A, AM16-105A, AM16-106A, XY012-120, AM29-21A, AM29-22A, AM29-32A, AM29-33A, AM16-103A, AM29-182A, AM29-55A, AM29-151A, AM29-152A, AM29-137A, AM29-153A, AM29-138A, AM29-154A, AM29-139A, AM29-155A, AM29-170A, AM29-156A, AM29-171A, AM29-157A, AM29-172A, AM29-173A, AM16-
  • the bivalent compound can be administered to the subject orally, parenterally, intradermally, subcutaneously, topically, or rectally.
  • any of the above-described methods can further include treating the subject with one or more additional therapeutic regimens for treating cancer.
  • the additional therapeutic regimens for treating cancer can include, for example, surgery, chemotherapy, radiation therapy (e.g., ionizing radiation or ultraviolet light), hormone therapy, or immunotherapy (e.g., antibody therapy).
  • radiation therapy e.g., ionizing radiation or ultraviolet light
  • immunotherapy e.g., antibody therapy
  • one or more bivalent compounds can be administered to the subject in conjunction with an effective amount of at least one established chemotherapeutic agent (e.g., actinomycin D, cyclophosphamide, doxorubicin, etoposide, and/or paclitaxel).
  • the EZH2-mediated cancer can include breast cancer (e.g., triple-negative breast cancer), glioblastoma, prostate cancer, uterine cancer, ovarian cancer, pancreatic cancer, melanoma, renal cell carcinoma, bladder cancer, colorectal cancer, lymphoma, leukemia, malignant rhabdoid tumor, and oropharyngeal cancer.
  • breast cancer e.g., triple-negative breast cancer
  • glioblastoma e.g., prostate cancer, uterine cancer, ovarian cancer, pancreatic cancer, melanoma, renal cell carcinoma, bladder cancer, colorectal cancer, lymphoma, leukemia, malignant rhabdoid tumor, and oropharyngeal cancer.
  • the EZH2-mediated cancer can include a relapsed cancer.
  • the EZH2-mediated cancer can be (known, predicted, or determined to be) refractory to one or more previous treatments (e.g., surgery, chemotherapy, radiation therapy, hormone therapy, or immunotherapy).
  • the document additionally provides identifying a bivalent compound which mediates degradation/disruption of EZH2, the method including:
  • bivalent test compound including an EZH2 ligand conjugated to a degradation/disruption tag
  • the cell can be a cancer cell (e.g., an EZH2-mediated cancer cell).
  • the terms “about” and “approximately” are defined as being within plus or minus 10% of a given value or state, preferably within plus or minus 5% of said value or state.
  • FIG. 1 depicts exemplary structures of bivalent compounds as described in the instant disclosure. Thalidomide/pomalidomide-based EZH2 degraders/disruptors and exemplary linkers 1-4.
  • FIG. 2 depicts exemplary structures of VHL-1-based EZH2 degraders/disruptors and exemplary linkers 5-7.
  • FIG. 3 depicts exemplary structures of adamantane-based EZH2 degraders/disruptors.
  • FIG. 4 is a graph depicting the GI 50 for AM16-10A for MCF-7 cells.
  • FIG. 5 is a graph depicting the GI 50 for AM16-10A for MDA-MB-468 cells.
  • FIG. 6 is a graph depicting the GI 50 for AM16-10A for HCC1187 cells.
  • FIG. 7 is a graph depicting the GI 50 for AM16-10A for HCC1170 cells.
  • FIG. 8 is a graph depicting the GI 50 for AM16-11A for HCC1187 cells.
  • FIG. 9 is a graph depicting the GI 50 for AM16-37A for HCC1187 cells.
  • FIG. 10 is a graph depicting the GI 50 for AM16-38A for HCC1187 cells.
  • FIG. 11 is a graph depicting the GI 50 for XY019-43 for MCF-7 cells.
  • FIG. 12 is a graph depicting the GI 50 for XY019-43 for MCF-7 and MCF-10A (control) cells.
  • FIG. 13 is a graph depicting the GI 50 for XY019-43 for MDA-MB-468 cells.
  • FIG. 14 is a graph depicting the GI 50 for XY019-43 for HCC1187 cells.
  • FIG. 15 is a graph depicting the GI 50 for XY019-43 for BT549 cells.
  • FIG. 16 is a graph depicting the GI 50 for XY019-43 for HCC1954 cells.
  • FIG. 17 is a graph depicting the GI 50 for XY019-44 for HCC1187 cells.
  • FIG. 18 is a graph depicting the GI 50 for AM16-92A for MCF-7 cells.
  • FIG. 19 is a graph depicting the GI 50 for AM16-92A for HCC1187 cells.
  • FIG. 20 is a graph depicting the GI 50 for AM16-93A for HCC1187 cells.
  • FIG. 21 is a graph depicting the GI 50 for AM16-97A for HCC1187 cells.
  • FIG. 22 is a graph depicting the GI 50 for AM16-101A for MCF-7 cells.
  • FIG. 23 is a graph depicting the GI 50 for AM16-101A for MDA-MB-468 cells.
  • FIG. 24 is a graph depicting the GI 50 for AM16-105A for MCF-7 cells.
  • FIG. 25 is a graph depicting the GI 50 for AM16-105A for HCC1187 cells.
  • FIG. 26 is a graph depicting the GI 50 for AM16-106A for HCC1187 cells.
  • FIG. 27 is a graph depicting the GI 50 for AM29-21A for MCF-7 cells.
  • FIG. 28 is a graph depicting the GI 50 for AM29-21A for MDA-MB-468 cells.
  • FIG. 29 is a graph depicting the GI 50 for AM29-22A for MCF-7 cells.
  • FIG. 30 is a graph depicting the GI 50 for AM29-22A for MDA-MB-468 cells.
  • FIG. 31 is a graph depicting the GI 50 for AM29-33A for MCF-7 cells.
  • FIG. 32 is a graph depicting the GI 50 for AM29-33A for MDA-MB-468 cells.
  • FIG. 33 is a graph depicting the GI 50 for AM16-103A for MDA-MB-468 cells.
  • FIG. 34 is a graph depicting the GI 50 for AM16-103A for BT549 cells.
  • FIG. 35 is a graph depicting the GI 50 for AM16-103A for HCC1954 cells.
  • FIG. 36 is a graph depicting the GI 50 for AM29-182A for MDA-MB-468 cells.
  • FIG. 37 is a graph depicting the GI 50 for AM29-182A for BT549 cells.
  • FIG. 38 is a graph depicting the GI 50 for AM29-182A for HCC1954 cells.
  • FIG. 39 is a graph depicting the GI 50 for AM29-177A for MDA-MB-468 cells.
  • FIG. 40 is a graph depicting the GI 50 for XY028-086 for MDA-MB-468 cells.
  • FIG. 41 is a graph depicting the GI 50 for CZ40-75 for MDA-MB-468 cells.
  • FIG. 42 is a graph depicting the GI 50 for CZ40-149 for MDA-MB-468 cells.
  • FIG. 43 is a graph depicting the GI 50 for CZ40-131 for MDA-MB-468 cells.
  • FIG. 44 is a graph depicting the GI 50 for AM41-41A for MDA-MB-468 cells.
  • FIG. 45 is a graph depicting the GI 50 for XF042-95 for MDA-MB-468 cells.
  • FIG. 46 is a graph depicting the GI 50 for XF042-90 for MDA-MB-468 cells.
  • FIG. 47 is a graph depicting the GI 50 for XF042-93 for MDA-MB-468 cells.
  • FIG. 48 is a graph depicting the GI 50 for XF042-133 for MDA-MB-468 cells.
  • FIG. 49 is a graph depicting the GI 50 for XF042-92 for MDA-MB-468 cells.
  • FIG. 50 is a graph depicting Western blot results showing EZH2 (2-day treatment) and H3K27me3 (1-day treatment) levels in MCF-7 cells treated with 1 ⁇ M AM16-10A, UNC1999 (negative control), or DMSO.
  • FIG. 51 is a Western blot showing EZH2 and H3K27me3 levels in MDA-MB-468 cells treated for 2 days with 2.5 or 5 ⁇ M XY019-43, AM29-182A, or DMSO.
  • FIG. 52 is a Western blot showing EZH2 and H3K27me3 levels in MDA-MB-468 cells treated for 2 days with various concentrations of AM29-177A or DMSO.
  • FIG. 53 is a Western blot showing EZH2 and H3K27me3 levels in MDA-MB-468 cells treated for 1 day with various concentrations of XY019-43, UNC1999 (negative control), or DMSO.
  • FIG. 54 is a Western blot showing EZH2 and H3K27me3 levels in HCC1187 cells treated for various times (h) with 1 ⁇ M AM16-100A, UNC1999 (negative control), or DMSO.
  • FIG. 55 is a graph depicting the in vitro IC 50 of AM16-10A for PRC2-EZH2.
  • FIG. 56 is a graph depicting the in vitro IC 50 of XY019-43 for PRC2-EZH2.
  • FIG. 57 is a graph depicting the in vitro IC 50 of AM16-101A for PRC2-EZH2.
  • the present disclosure is based, in part, on the discovery that novel bivalent compounds which selectively degrade/disrupt EZH2 (“EZH2 degraders/disruptors”) are useful in the treatment of EZH2-mediated cancers, including but not limited to TNBC.
  • EZH2 degraders/disruptors novel bivalent compounds which selectively degrade/disrupt EZH2
  • this disclosure provides specific examples of novel EZH2 degraders/disruptors.
  • the effect of exemplary degraders/disruptors on the proliferation of different tumor cell lines was examined.
  • the effect of exemplary degraders/disruptors on EZH2 and H3K27me3 protein levels and the enzymatic activity of the PRC2-EZH2 complex were also evaluated.
  • This novel therapeutic approach can be beneficial, particularly since the standard of care for TNBC is primarily chemotherapy and radiation.
  • EZH2 degraders/disruptors disclosed herein have dual functions (enzyme inhibition plus protein degradation/disruption), they can be significantly more effective than current EZH2 inhibitors, which inhibit the enzymatic activity of EZH2 but do not affect EZH2 protein levels, for treating other EZH2-mediated cancers.
  • Some of these inhibitors have been in clinical trials for treating diffused large B cell lymphoma (DLBCL), follicular lymphoma (FL), and malignant rhabdoid tumor (MRT).
  • DLBCL diffused large B cell lymphoma
  • FL follicular lymphoma
  • MRT malignant rhabdoid tumor
  • these inhibitors have exhibited very limited success in treating breast cancers and prostate cancers mainly because these compounds only inhibit the methyltransferase activity of EZH2, but do not change EZH2 protein levels.
  • Representative examples of selective EZH2 inhibitors are provided below.
  • an EZH2 ligand or targeting moiety e.g., an EZH2 inhibitor such as UNC1999 or compound 24
  • a ubiquitin ligase e.g., an E3 ligase
  • a hydrophobic group e.g., adamantane
  • EZH2 degraders/disruptors recruit the ubiquitination machinery to EZH2, leading to selective degradation of EZH2 via the ubiquitin-proteasome pathway, and/or mimic EZH2 protein misfolding and subsequent degradation at the proteasome or loss of function. Therefore, these degraders/disruptors can be effective therapeutic agents for treating breast cancers (including TNBC), prostate cancers, and other cancers while current EZH2 inhibitors are ineffective.
  • these EZH2 degraders/disruptors can be more effective than EZH2 inhibitors for the treatment of those EZH2-mediated cancers where EZH2 inhibitors are effective, including, e.g., DLBCB, FL, and MRT.
  • the present disclosure provides bivalent compounds, referred to herein as “EZH2 degraders/disruptors”, comprising an enhancer of zeste homologue 2 (EZH2) ligand (or targeting moiety) conjugated to a degradation/disruption tag.
  • EZH2 degraders/disruptors comprising an enhancer of zeste homologue 2 (EZH2) ligand (or targeting moiety) conjugated to a degradation/disruption tag.
  • Linkage of the EZH2 ligand to the degradation/disruption tag can be direct, or indirect via a linker.
  • the term “enhancer of zeste homologue 2 ligand” or “EZH2 ligand” refers to compound that associates and/or binds to EZH2.
  • the EZH2 ligand can be, e.g., a small-molecule compound (i.e., a molecule of molecular weight less than about 1.5 kilodaltons (kDa)), a peptide, or an antibody or fragment thereof which is capable of binding to EZH2 and/or interfering with the methyltransferase enzymatic activity of EZH2.
  • the EZH2 ligand can be an EZH2 inhibitor, which is capable of interfering with the methyltransferase enzymatic activity of EZH2.
  • an “inhibitor” refers to an agent that restrains, retards, or otherwise causes inhibition of a physiological, chemical or enzymatic action or function. An inhibitor may cause at least 5% decrease in enzyme activity. An inhibitor may also refer to a drug, compound or agent that prevents or reduces the expression, transcription or translation of a gene or protein. An inhibitor may reduce or prevent the function of a protein, for instance by binding to and/or activating/inactivating another protein or receptor.
  • the EZH2 inhibitors of the present disclosure include, for example, UNC1999, EPZ005687, EPZ-6438, GSK126, EI1, CPI-1205, GSK343, CPI-360, EPZ011989, compound 24, compound 3, compound 31, ZLD1039, PF-06821497, JQEZ5, and analogs thereof.
  • degradation/disruption tag refers to a moiety, which associates with/binds to a ubiquitin ligase for recruitment of the corresponding ubiquitination machinery to the EHZ2/PRC2 complex, or mimics EZH2 protein misfolding and subsequent degradation at the proteasome or loss of function.
  • One or more degradation/disruption tags can be introduced to the solvent-exposed portion of an EZH2 ligand to create EZH2 degraders/disruptors. Exemplary structures of EZH2 degraders/disruptors containing such tags are illustrated in FIGS. 1-3 .
  • a docking model of UNC1999 and its close analogs in PRC2 crystal structures shows that two regions of UNC1999 and its analogs are solvent-exposed, thus presenting suitable handles to introduce a degradation/disruption tag without interfering with the inhibitors' ability to bind to EZH2. These regions are the piperazine portion (marked in red in FIGS. 1-3 ) and isopropyl group (marked in blue in FIGS. 1-3 ).
  • Structure-activity relationship (SAR) studies showed that modifying these two portions resulted in negligible effects on the molecule's potency towards EZH2 (Konze et al., 2013; Yang et al., 2016).
  • the degradation/disruption tags of the present disclosure include, for example, immunomodulatory drugs (e.g., thalidomide, pomalidomide, and lenalidomide), VHL-1, bulky hydrophobic groups (e.g., adamantane and 1-((4,4,5,5,5-pentafluoropentyl)sulfinyl)nonane), and their analogs.
  • immunomodulatory drugs e.g., thalidomide, pomalidomide, and lenalidomide
  • VHL-1 e.g., adamantane and 1-((4,4,5,5,5-pentafluoropentyl)sulfinyl)nonane
  • Immunomodulatory drugs such as thalidomide and pomalidomide (structures shown in FIG.
  • VHL-1 structure shown in FIG.
  • VHL or CRL2 VHL van Hippel-Lindau protein
  • VHL or CRL2 VHL van Hippel-Lindau protein
  • Bulky hydrophobic groups e.g., adamantane
  • mimic protein misfolding, leading to the degradation of the target protein by proteasome Buckley and Crews, 2014.
  • a “linker” is a bond, molecule or group of molecules that binds (i.e., bridges) two separate entities to one another.
  • a Linker can provide for optimal spacing of the two entities.
  • the term “linker” in some aspects refers to any agent or molecule that bridges the EZH2 ligand to the degradation/disruption tag.
  • sites on the EZH2 ligand and/or the degradation/disruption tag which are not necessary for the function of the bivalent compound of the present disclosures, are ideal sites for attaching a linker, provided that the linker, once attached to the conjugate of the present disclosures, does not interfere with the function of the bivalent compound, i.e., the ability to target EZH2 and recruit a ubiquitin ligase or mimic protein misfolding.
  • the length of the linker can be adjusted to minimize the molecular weight of the degrader/disruptor, avoid any steric interference of EZH2 with the E3 ligase, and/or enhance mimicry of EZH2 protein misfolding by the hydrophobic tag at the same time.
  • linkers include, but are not limited to, the linkers of Formulas I-XIV below:
  • the EZH2 degraders/disruptors have the form “X-linker-Y”, as shown below:
  • X comprises a degradation/disruption tag (e.g., adamantane) and Y comprises an EZH2 ligand (e.g., an EZH2 inhibitor).
  • EZH2 ligand e.g., an EZH2 inhibitor.
  • Exemplary degradation/disruption tags (X) and exemplary EZH2 ligands (Y) are described above and are also illustrated below:
  • Novel EZH2 degraders/disruptors developed using the principles and methods described herein are shown in Table 1. Additional EZH2 degraders/disruptors can also be developed using the principles and methods disclosed herein. For example, other linkers, other degradation/disruptor tags, and/or other EZH2 ligands can be synthesized and tested. Some exemplary compounds are shown in the Figures following Table 1.
  • EZH2 degraders/disruptors can be assessed by EZH2 biochemical assays known in the art (Konze et al., 2013; Yang et al., 2016); see, e.g., Example 115. Their binding affinity to EZH2 can be assessed using standard biophysical assays known in the art (e.g., ITC, SPR). Cellular assays (e.g., as depicted in Examples 113 and 114) can be used to assess the compounds' ability to induce EZH2 degradation/disruption, reduce the H3K27me3 mark, and/or inhibit cancer cell proliferation.
  • Assays suitable for use in any or all of these steps are known in the art, and include, e.g., Western blotting and MTT.
  • Suitable cell lines for use in any or all of these steps are known in the art and include, e.g., HCC70, HCC1170, HCC1187, MDA-MB-468, MDA-MB-231, MCF-7, BT549, HCC1954, HeLa S3, HEK 293, U2OS, and HFF cells.
  • compositions and methods described herein include the manufacture and use of pharmaceutical compositions and medicaments that include compounds identified by a method described herein as active ingredients. Also included are the pharmaceutical compositions themselves.
  • compositions disclosed herein can include other compounds, drugs, and/or agents used for the treatment of cancer.
  • therapeutic compositions disclosed herein can be combined with one or more (e.g., one, two, three, four, five, or less than ten) compounds.
  • compositions disclosed herein can include EZH2 degraders/disruptors such as AM16-10A, XY019-43, AM29-182A, AM19-177A, AM16-103A, CZ40-75, CZ40-149, AM41-41A, XF042-95, XF042-93, XF042-133, XY028-086, CZ40-131, and XF042-92.
  • EZH2 degraders/disruptors such as AM16-10A, XY019-43, AM29-182A, AM19-177A, AM16-103A, CZ40-75, CZ40-149, AM41-41A, XF042-95, XF042-93, XF042-133, XY028-086, CZ40-131, and XF042-92.
  • An EZH2 degrader/disruptor can selectively affect EZH2-mediated cancer cells (e.g., TNBC cells) compared to WT, normal or non-tumor cells (i.e., a degrader/disruptor able to kill or inhibit the growth of EZH2-mediated cancer cells while also having a relatively low ability to lyse or inhibit the growth of WT, normal or non-tumor cells), e.g., possess a GI 50 for one or more EZH2-mediated cancer cells more than 1.5-fold lower, more than 2-fold lower, more than 2.5-fold lower, more than 3-fold lower, more than 4-fold lower, more than 5-fold lower, more than 6-fold lower, more than 7-fold lower, more than 8-fold lower, more than 9-fold lower, more than 10-fold lower, more than 15-fold lower, or more than 20-fold lower than its GI 50 for one or more WT, normal or non-tumor cells, e.g., WT, normal or non-tum
  • One or more of the EZH2 degraders/disruptors disclosed herein can be formulated for use as or in pharmaceutical compositions.
  • Such compositions can be formulated or adapted for administration to a subject via any route, e.g., any route approved by the Food and Drug Administration (FDA). Exemplary methods are described in the FDA Data Standards Manual (DSM) (available at http://www.fda.gov/Drugs/DevelopmentApprovalProcess/FormsSubmissionRequirements/ElectronicSubmissions/DataStandardsManualmonographs).
  • DSM Food and Drug Administration
  • the pharmaceutical compositions may be formulated for oral, parenteral, or transdermal delivery.
  • the compound of the invention may also be combined with other pharmaceutical agents.
  • compositions disclosed herein can be administered, e.g., orally, parenterally, by inhalation spray or nebulizer, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, by injection (e.g., intravenously, intra-arterially, subdermally, intraperitoneally, intramuscularly, and/or subcutaneously), in an ophthalmic preparation, or via transmucosal administration. Suitable dosages may range from about 0.001 to about 100 mg/kg of body weight, or according to the requirements of the particular drug.
  • the pharmaceutical compositions of this invention can contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation can be adjusted with pharmaceutically acceptable acids, bases, or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intra-arterial, intrasynovial, intrasternal, intrathecal, intralesional, and intracranial injection or infusion techniques.
  • the present invention may be administered according to any of the methods as described in the FDA DSM.
  • compositions typically include a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier or adjuvant refers to a carrier or adjuvant that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
  • pharmaceutically acceptable carrier includes saline, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the phrase “pharmaceutically acceptable” refers to molecular entities and compositions that are generally believed to be physiologically tolerable and do not typically produce an allergic or similar untoward reaction, such as gastric upset, dizziness and the like, when administered to a human.
  • pharmaceutically acceptable derivative means any pharmaceutically acceptable salt, solvate or prodrug, e.g., ester, of an atovaquone-related compound described herein, which upon administration to the recipient is capable of providing (directly or indirectly) a compound described herein, or an active metabolite or residue thereof. Such derivatives are recognizable to those skilled in the art, without undue experimentation.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intraarterial, intrasynovial, intrastemal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • compositions are typically formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • the EZH2 degraders/disruptors disclosed herein are defined to include pharmaceutically acceptable derivatives or prodrugs thereof.
  • a “pharmaceutically acceptable derivative or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound or agent disclosed herein which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention.
  • Particularly favored derivatives and prodrugs are those that increase the bioavailability of the compounds disclosed herein when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Preferred prodrugs include derivatives where a group that enhances aqueous solubility or active transport through the gut membrane is appended to the structure of formulae described herein.
  • the EZH2 degraders/disruptors disclosed herein include pure enantiomers, mixtures of enantiomers, pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates, mixtures of diastereoisomeric racemates and the meso-form and pharmaceutically acceptable salts, solvent complexes, morphological forms, or deuterated derivative thereof.
  • compositions can include an effective amount of one or more EZH2 degraders/disruptors.
  • effective amount and “effective to treat,” as used herein, refer to an amount or a concentration of one or more compounds or a pharmaceutical composition described herein utilized for a period of time (including acute or chronic administration and periodic or continuous administration) that is effective within the context of its administration for causing an intended effect or physiological outcome (e.g., treatment or prevention of cell growth, cell proliferation, or cancer).
  • the present disclosure provides methods for using a composition comprising an EZH2 degrader/disruptor, including pharmaceutical compositions (indicated below as ‘X’) disclosed herein in the following methods:
  • Substance X for use as a medicament in the treatment of one or more diseases or conditions disclosed herein e.g., neurodegenerative disease, referred to in the following examples as ‘Y’).
  • compositions disclosed herein can be formulated for sale in the US, import into the US, and/or export from the US.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • compositions of this invention will be administered from about 1 to about 6 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations can contain from about 20% to about 80% active compound.
  • an effective dose of an EZH2 degrader/disruptor can include, but is not limited to, e.g., about 0.00001, 0.0001, 0.001, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.15, 0.2, 0.25, 0.3, 0.35, 0.4, 0.45, 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 1, 1.25, 1.5, 1.75, 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2500, 5000, or 10000 mg/kg/day.
  • compositions of this invention can include one or more EZH2 degraders/disruptors and any pharmaceutically acceptable carrier and/or vehicle.
  • pharmaceuticals can further include one or more additional therapeutic agents in amounts effective for achieving a modulation of disease or disease symptoms.
  • additional therapeutic agents may include conventional chemotherapeutic agents known in the art.
  • EZH2 degraders/disruptors disclosed herein can operate in conjunction with conventional chemotherapeutic agents to produce mechanistically additive or synergistic therapeutic effects.
  • compositions of this invention comprise a combination of a compound of the formulae described herein and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 1 to 100%, and more preferably between about 5 to 95% of the dosage normally administered in a monotherapy regimen.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of this invention in a single composition.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d- ⁇ -tocopherol polyethylene glycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tween®s or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-
  • compositions can be in the form of a solution or powder for injection. Such compositions may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween® 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • surfactants such as Tween®s, Spann's, and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions can be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions can be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • Pharmaceutically acceptable salts of the EZH2 degraders/disruptors of this disclosure include, e.g., those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acid salts include acetate, adipate, benzoate, benzenesulfonate, butyrate, citrate, digluconate, dodecylsulfate, formate, fumarate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, palmoate, phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate, tartrate, tosylate, trifluoromethylsulfonate, and undecanoate.
  • Salts derived from appropriate bases include, e.g., alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), ammonium and N-(alkyl) 4+ salts.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., ammonium
  • N-(alkyl) 4+ salts e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium e.g., sodium
  • the methods described herein include methods for the treatment of disorders associated with EZH2-mediated cancer, the methods include administering a therapeutically effective amount of an EZH2 degrader/disruptor as described herein, to a subject (e.g., a mammalian subject, e.g., a human subject) who is in need of, or who has been determined to be in need of, such treatment.
  • a subject e.g., a mammalian subject, e.g., a human subject
  • methods can include selection of a human subject who has or had a condition or disease.
  • suitable subjects include, for example, subjects who have or had a condition or disease but that resolved the disease or an aspect thereof, present reduced symptoms of disease (e.g., relative to other subjects (e.g., the majority of subjects) with the same condition or disease), and/or that survive for extended periods of time with the condition or disease (e.g., relative to other subjects (e.g., the majority of subjects) with the same condition or disease), e.g., in an asymptomatic state (e.g., relative to other subjects (e.g., the majority of subjects) with the same condition or disease).
  • treat refers to partially or completely alleviating, inhibiting, ameliorating, and/or relieving the disease or condition from which the subject is suffering. This means any manner in which one or more of the symptoms of a disease or disorder (e.g., cancer) are ameliorated or otherwise beneficially altered.
  • amelioration of the symptoms of a particular disorder refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with treatment by the compositions and methods of the present invention.
  • treatment can promote or result in, for example, a decrease in the number of tumor cells (e.g., in a subject) relative to the number of tumor cells prior to treatment; a decrease in the viability (e.g., the average/mean viability) of tumor cells (e.g., in a subject) relative to the viability of tumor cells prior to treatment; and/or reductions in one or more symptoms associated with one or more tumors in a subject relative to the subject's symptoms prior to treatment.
  • a decrease in the number of tumor cells e.g., in a subject
  • a decrease in the viability e.g., the average/mean viability
  • the term “treating cancer” means causing a partial or complete decrease in the rate of growth of a tumor, and/or in the size of the tumor and/or in the rate of local or distant tumor metastasis, and/or the overall tumor burden in a subject, and/or any decrease in tumor survival, in the presence of a degrader/disruptor (e.g., an EZH2 degrader/disruptor) described herein.
  • a degrader/disruptor e.g., an EZH2 degrader/disruptor
  • the term “preventing a disease” in a subject means for example, to stop the development of one or more symptoms of a disease in a subject before they occur or are detectable, e.g., by the patient or the patient's doctor.
  • the disease e.g., cancer
  • the disease does not develop at all, i.e., no symptoms of the disease are detectable.
  • it can also result in delaying or slowing of the development of one or more symptoms of the disease.
  • it can result in the decreasing of the severity of one or more subsequently developed symptoms.
  • Exemplary EZH2-mediated cancers that can be treated with EZH2 degraders/disruptors include, for example, INI1-negative tumors, lymphoma (including diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and non-Hodgkin's lymphoma (NHL)), malignant rhabdoid tumor, multiple myeloma, relapsed/refractory synovial sarcoma, breast cancers (including TNBC), prostate cancers, other solid tumors, acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, AIDS-related cancers, anal cancer, astrocytoma, childhood cerebellar cancer, basal cell carcinoma, skin cancer (non-melanoma), bile duct cancer, bladder cancer, bone cancer, osteosarcoma/malignant fibrous histiocytoma, brain stem glioma, brain tumor, cerebellar a
  • prevent shall refer to a decrease in the occurrence of a disease or decrease in the risk of acquiring a disease or its associated symptoms in a subject.
  • the prevention may be complete, e.g., the total absence of disease or pathological cells in a subject.
  • the prevention may also be partial, such that the occurrence of the disease or pathological cells in a subject is less than that which would have occurred without the present invention.
  • subject refers to any animal. In some instances, the subject is a mammal. In some instances, the term “subject,” as used herein, refers to a human (e.g., a man, a woman, or a child).
  • subject selection can include obtaining a sample from a subject (e.g., a candidate subject) and testing the sample for an indication that the subject is suitable for selection.
  • the subject can be confirmed or identified, e.g. by a health care professional, as having had or having a condition or disease.
  • exhibition of a positive immune response towards a condition or disease can be made from patient records, family history, and/or detecting an indication of a positive immune response.
  • multiple parties can be included in subject selection. For example, a first party can obtain a sample from a candidate subject and a second party can test the sample.
  • subjects can be selected and/or referred by a medical practitioner (e.g., a general practitioner).
  • subject selection can include obtaining a sample from a selected subject and storing the sample and/or using the in the methods disclosed herein. Samples can include, for example, cells or populations of cells.
  • methods include selecting a subject and administering to the subject an effective amount of one or more of the EZH2 degraders/disruptors described herein, e.g., in or as a pharmaceutical composition, and optionally repeating administration as required for the prophylaxis or treatment of cancer and can be administered, e.g., orally, intravenously or topically.
  • Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician.
  • treatments methods can include a single administration, multiple administrations, and repeating administration as required for the prophylaxis or treatment of the disease or condition from which the subject is suffering (e.g., an EZH2-mediated cancer, e.g., breast cancers including TNBC).
  • treatment methods can include assessing a level of disease in the subject prior to treatment, during treatment, and/or after treatment. In some instances, treatment can continue until a decrease in the level of disease in the subject is detected.
  • administer refers to implanting, absorbing, ingesting, injecting, or inhaling, the inventive drug, regardless of form.
  • one or more of the compounds disclosed herein can be administered to a subject topically (e.g., nasally) and/or orally.
  • the methods herein include administration of an effective amount of compound or compound composition to achieve the desired or stated effect.
  • Specific dosage and treatment regimens for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health status, sex, diet, time of administration, rate of excretion, drug combination, the severity and course of the disease, condition or symptoms, the patient's disposition to the disease, condition or symptoms, and the judgment of the treating physician.
  • the subject can be evaluated to detect, assess, or determine their level of disease. In some instances, treatment can continue until a change (e.g., reduction) in the level of disease in the subject is detected.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • An effective amount can be administered in one or more administrations, applications or dosages.
  • a therapeutically effective amount of a therapeutic compound depends on the therapeutic compounds selected.
  • the compositions can be administered one from one or more times per day to one or more times per week; including once every other day. The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present.
  • treatment of a subject with a therapeutically effective amount of the therapeutic compounds described herein can include a single treatment or a series of treatments.
  • effective amounts can be administered at least once.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary.
  • the dosage or frequency of administration, or both may be reduced, as a function of the symptoms, to a level at which the improved condition is retained.
  • Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • HPLC HPLC spectra for all compounds were acquired using an AgilentTM 1200 Series system with DAD detector. Chromatography was performed on a 2.1 ⁇ 150 mm ZorbaxTM 300SB-C18 5 ⁇ m column with water containing 0.1% formic acid as solvent A and acetonitrile containing 0.1% formic acid as solvent B at a flow rate of 0.4 ml/min. The gradient program was as follows: 1% B (0-1 min), 1-99% B (1-4 min), and 99% B (4-8 min). High-resolution mass spectra (HRMS) data were acquired in positive ion mode using an AgilentTM G1969A API-TOF with an electrospray ionization (ESI) source.
  • HRMS high-resolution mass spectra
  • intermediate 2 (302 mg, 73% over 2 steps).
  • Intermediate 2 100 mg, 0.15 mmol
  • HOAt (1-hydroxy-7-azabenzo-triazole)
  • 1-adamantaneacetic acid 35 mg, 0.18 mmol, Sigma®, #127272
  • DMSO 2.0 mL
  • NMM 66 ⁇ L, 0.60 mmol
  • EDCI 43 mg, 0.23 mmol
  • AM16-11A was synthesized according to the procedures for preparing AM16-10A from intermediate 2 (92 mg, 0.13 mmol), HOAt (27 mg, 0.20 mmol), 3-(1-adamantyl)propanoic acid (33 mg, 0.16 mmol, Matrix ScientificTM, #038155), NMM (57 ⁇ L, 0.52 mmol), EDCI (39 mg, 0.20 mmol), and DMSO (2.0 mL).
  • AM16-11A was obtained as white solid in TFA salt form (58 mg, 51%).
  • AM16-37A was synthesized according to the procedures for preparing AM16-10A from intermediate 1 (100 mg, 0.16 mmol), HOAt (33 mg, 0.24 mmol), 1-adamantaneacetic acid (38 mg, 0.19 mmol), NMM (71 ⁇ L, 0.64 mmol), EDCI (46 mg, 0.24 mmol), and DMSO (2.0 mL).
  • AM16-37A was obtained as yellow solid (73 mg, 65%).
  • AM16-38A was synthesized according to the procedures for preparing AM16-10A from intermediate 1 (100 mg, 0.16 mmol), HOAt (33 mg, 0.24 mmol), 1-adamantaneacetic acid (38 mg, 0.19 mmol), NMM (71 ⁇ L, 0.64 mmol), EDCI (46 mg, 0.24 mmol), and DMSO (2.0 mL).
  • AM16-38A was obtained as brown solid (69 mg, 60%).
  • AM16-92A was synthesized according to the procedures for preparing AM16-10A from intermediate 3 (100 mg, 0.16 mmol), HOAt (33 mg, 0.24 mmol), 1-adamantaneacetic acid (38 mg, 0.20 mmol), NMM (71 ⁇ L, 0.64 mmol), EDCI (46 mg, 0.24 mmol), and DMSO (2.0 mL).
  • AM16-92A was obtained as white solid in TFA salt form (77 mg, 61%).
  • AM16-93A was synthesized according to the procedures for preparing AM16-10A from intermediate 3 (100 mg, 0.16 mmol), HOAt (33 mg, 0.24 mmol), 3-(1-adamantyl)propanoic acid (42 mg, 0.20 mmol), NMM (71 ⁇ L, 0.64 mmol), EDCI (46 mg, 0.24 mmol), and DMSO (2.0 mL).
  • AM16-93A was obtained green solid in TFA salt form (85 mg, 66%).
  • AM16-97A was synthesized according to the procedures for preparing AM16-10A from intermediate 3 (67 mg, 0.11 mmol), HOAt (23 mg, 0.17 mmol), (2R)-4-((1r,3S)-adamantan-1-yl)-2-methylbutanoic acid (25 mg, 0.11 mmol), NMM (49 ⁇ L, 0.44 mmol), EDCI (33 mg, 0.17 mmol), and DMSO (2.0 mL). (2R)-4-((1r,3S)-Adamantan-1-yl)-2-methylbutanoic acid was synthesized according to the procedures reported previously(Neklesa et al., 2011).
  • AM16-97A was obtained as brown solid in TFA salt form (58 mg, 63%).
  • 1 H NMR 600 MHz, CD 3 OD
  • ⁇ 8.43 (brs, 1H), 8.37 (s, 1H), 8.23 (brs, 1H), 7.98 (s, 1H), 7.77 (s, 1H), 7.17 (brs, 1H), 6.13 (s, 1H), 5.13-5.03 (m, 1H), 4.57 (s, 2H), 3.89-3.79 (m, 4H), 3.76 (brs, 2H), 3.70 (brs, 2H), 2.84-2.74 (m, 1H), 2.43 (s, 3H), 2.25 (s, 3H), 1.92 (brs, 3H), 1.77-1.70 (m, 3H), 1.67-1.63 (m, 3H), 1.59-1.54 (m, 6H), 1.50 (brs, 6H), 1.46-1.29 (m, 2H), 1.13 (d, J 6.6 Hz, 3
  • AM16-100A was synthesized according to the procedures for preparing AM16-10A from intermediate 3 (75 mg, 0.12 mmol), HOAt (25 mg, 0.18 mmol), 1-adamantanecarboxylic acid (27 mg, 0.15 mmol, Sigma®, #106399), NMM (53 ⁇ L, 0.48 mmol), EDCI (35 mg, 0.18 mmol), and DMSO (1.5 mL).
  • AM16-100A was obtained as brown solid in TFA salt form (92 mg, 99%).
  • AM16-101A was synthesized according to the procedures for preparing AM16-10A from intermediate 7 (75 mg, 0.11 mmol), HOAt (23 mg, 0.17 mmol), 1-adamantanecarboxylic acid (25 mg, 0.14 mmol), NMM (51 ⁇ L, 0.46 mmol), EDCI (33 mg, 0.17 mmol), and DMSO (1.5 mL).
  • AM16-101A was obtained as off-white solid in TFA salt form (80 mg, 86%).
  • AM16-102A was synthesized according to the procedures for preparing AM16-10A from intermediate 8 (116 mg, 0.17 mmol), HOAt (35 mg, 0.26 mmol), 1-adamantaneacetic acid (41 mg, 0.21 mmol), NMM (75 ⁇ L, 0.68 mmol), EDCI (50 mg, 0.26 mmol), and DMSO (1.5 mL).
  • AM16-102A was obtained as white solid in TFA salt form (101 mg, 70%).
  • AM16-105A was synthesized according to the procedures for preparing AM16-10A from intermediate 7 (100 mg, 0.15 mmol), HOAt (31 mg, 0.23 mmol), (2R)-4-((1r,3S)-adamantan-1-yl)-2-methylbutanoic acid (36 mg, 0.15 mmol), NMM (66 ⁇ L, 0.60 mmol), EDCI (44 mg, 0.23 mmol), and DMSO (1.5 mL).
  • AM16-105A was obtained as white solid in TFA salt form (102 mg, 77%).
  • AM16-106A was synthesized according to the procedures for preparing AM16-10A from intermediate 8 (100 mg, 0.15 mmol), HOAt (31 mg, 0.23 mmol), (2R)-4-((1r,3S)-adamantan-1-yl)-2-methylbutanoic acid (36 mg, 0.15 mmol), NMM (66 ⁇ L, 0.60 mmol), EDCI (44 mg, 0.23 mmol), and DMSO (1.5 mL).
  • AM16-106A was obtained as solid in TFA salt form (101 mg, 76%).
  • AM29-21A was synthesized according to the procedures for preparing AM16-10A from intermediate 5 (80 mg, 0.09 mmol), HOAt (19 mg, 0.14 mmol), 3,5-dimethyladamantane-1-acetic acid (25 mg, 0.11 mmol, Sigma®, #679976), NMM (40 ⁇ L, 0.36 mmol), EDCI (27 mg, 0.14 mmol), and DMSO (1.0 mL).
  • AM29-21A was obtained as off-white solid in TFA salt form (58 mg, 74%).
  • AM29-22A was synthesized according to the procedures for preparing AM16-10A from intermediate 5 (80 mg, 0.09 mmol), HOAt (19 mg, 0.14 mmol), 3,5-dimethyladamantane-1-carboxylic acid (23 mg, 0.11 mmol, Sigma®, #679984), NMM (40 ⁇ L, 0.36 mmol), EDCI (27 mg, 0.14 mmol), and DMSO (1.0 mL).
  • AM29-22A was obtained as off-white solid in TFA salt form (67 mg, 87%).
  • AM29-32A was synthesized according to the procedures for preparing AM16-10A from intermediate 12 (55 mg, 0.08 mmol), HOAt (17 mg, 0.12 mmol), amantadine hydrochloride (19 mg, 0.10 mmol, Sigma®, #A1260), NMM (35 ⁇ L, 0.32 mmol), EDCI (23 mg, 0.12 mmol), and DMSO (1.0 mL).
  • AM29-32A was obtained as off-white solid in TFA salt form (18 mg, 27%).
  • AM29-33A was synthesized according to the procedures for preparing AM16-10A from intermediate 12 (55 mg, 0.08 mmol), HOAt (17 mg, 0.12 mmol), 1-adamantanemethylamine (16 mg, 0.10 mmol, Acros Organics, #177420010), NMM (35 ⁇ L, 0.32 mmol), EDCI (23 mg, 0.12 mmol), and DMSO (1.0 mL).
  • AM29-33A was obtained as off-white solid in TFA salt form (60 mg, 90%).
  • AM29-182A was synthesized according to the procedures for preparing XY019-43 from intermediate 7 (30 mg, 0.05 mmol), HOAt (9 mg, 0.07 mmol), 2-(adamantan-2-yl)acetic acid (11 mg, 0.06 mmol), NMM (20 ⁇ L, 0.18 mmol), EDCI (14 mg, 0.07 mmol), and DMSO (1.0 mL).
  • AM29-182 was obtained as white solid in TFA salt form (27 mg, 72%).
  • AM29-55A was synthesized according to the procedures for preparing AM16-10A from intermediate 2 (60 mg, 0.08 mmol), HOAt (17 mg, 0.12 mmol), intermediate 13 (31 mg, 0.08 mmol), NMM (44 ⁇ L, 0.40 mmol), EDCI (23 mg, 0.12 mmol) and DMF (1.0 mL). AM29-55A was obtained as yellow solid in TFA salt form (22 mg, 27%).
  • AM29-152A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 3-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)propanoic acid (6 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-152A was obtained as yellow solid in TFA salt form (9.6 mg, 65%).
  • AM29-137A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((2-aminoethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-137A was obtained as yellow solid in TFA salt form (11 mg, 75%).
  • AM29-153A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)butanoic acid (6 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-153A was obtained as yellow solid in TFA salt form (11 mg, 78%).
  • AM29-138A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((3-aminopropyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-138A was obtained as yellow solid in TFA salt form (14 mg, 96%).
  • AM29-154A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 5-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)pentanoic acid (7 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-154A was obtained as yellow solid in TFA salt form (12 mg, 82%).
  • AM29-139A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((4-aminobutyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-139A was obtained as yellow solid in TFA salt form (9.8 mg, 66%).
  • AM29-155A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 6-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)hexanoic acid (7 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-155A was obtained as yellow solid in TFA salt form (12 mg, 76%).
  • AM29-170A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (20 mg, 0.02 mmol), HATU (22 mg, 0.06 mmol), 4-((5-aminopentyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (17 mg, 0.04 mmol), DIPEA (20 ⁇ L, 0.12 mmol), and DMF (1.0 mL). AM29-170A was obtained as yellow solid in TFA salt form (20 mg, 64%).
  • AM29-156A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 7-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)heptanoic acid (7 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-156A was obtained as yellow solid in TFA salt form (13 mg, 83%).
  • AM29-171A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (20 mg, 0.02 mmol), HATU (22 mg, 0.06 mmol), 4-((6-aminohexyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (17 mg, 0.04 mmol), DIPEA (20 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-171A was obtained as yellow solid in TFA salt form (20 mg, 66%).
  • AM29-157A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 8-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)octanoic acid (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-157A was obtained as yellow solid in TFA salt form (11 mg, 67%).
  • AM29-172A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (20 mg, 0.02 mmol), HATU (22 mg, 0.06 mmol), 4-((7-aminoheptyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (18 mg, 0.04 mmol), DIPEA (20 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-172A was obtained as yellow solid in TFA salt form (23 mg, 73%).
  • AM29-173A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (20 mg, 0.02 mmol), HATU (22 mg, 0.06 mmol), 4-((8-aminooctyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (18 mg, 0.04 mmol), DIPEA (20 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-173A was obtained as yellow solid in TFA salt form (26 mg, 82%).
  • AM29-177A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)propanoic acid (14 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-177A was obtained as yellow solid in TFA salt form (30 mg, 95%).
  • AM29-141A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((2-(2-aminoethoxy) ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-141A was obtained as yellow liquid in TFA salt form (4.8 mg, 32%).
  • AM29-178A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 3-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)ethoxy)propanoic acid (16 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-178A was obtained as yellow solid in TFA salt form (27 mg, 85%).
  • AM29-142A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((2-(2-(2-aminoethoxy) ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (8 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-142A was obtained as brown liquid in TFA salt form (4 mg, 25%).
  • AM29-179A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 3-(2-(2-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)ethoxy)ethoxy)propanoic acid (18 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-179A was obtained as yellow solid in TFA salt form (26 mg, 78%).
  • AM29-143A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((2-(2-(2-(2-aminoethoxy) ethoxy)ethoxy)ethyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (9 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-143A was obtained as brown liquid in TFA salt form (11 mg, 67%).
  • AM29-180A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 1-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12-tetraoxapentadecan-15-oic acid (20 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-180A was obtained as yellow solid in TFA salt form (31 mg, 90%).
  • AM29-144A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((14-amino-3,6,9,12-tetraoxatetradecyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (10 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-144A was obtained as yellow solid in TFA salt form (9 mg, 53%).
  • AM29-145A was synthesized according to the procedures for preparing AM29-151A from intermediate 12 (10 mg, 0.02 mmol), HATU (11 mg, 0.03 mmol), 4-((17-amino-3,6,9,12,15-pentaoxaheptadecyl)amino)-2-(2,6-dioxopiperidin-3-yl)isoindoline-1,3-dione (11 mg, 0.02 mmol), DIPEA (11 ⁇ L, 0.06 mmol), and DMF (1.0 mL).
  • AM29-145A was obtained as yellow liquid in TFA salt form (12 mg, 70%).
  • AM29-181A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 1-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12,15-pentaoxaoctadecan-18-oic acid (21 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM29-181A was obtained as yellow solid in TFA salt form (9 mg, 25%).
  • AM41-16A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (18 mg, 0.03 mmol), HATU (21 mg, 0.05 mmol), 1-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12,15,18,21,24-octaoxaheptacosan-27-oic acid (19 mg, 0.03 mmol), DIPEA (19 ⁇ L, 0.11 mmol), and DMF (1.0 mL). AM41-16A was obtained as yellow liquid in TFA salt form (31 mg, 86%).
  • AM41-17A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (18 mg, 0.03 mmol), HATU (18 mg, 0.05 mmol), 1-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12,15,18,21,24,27,30-decaoxatritriacontan-33-oic acid (19 mg, 0.02 mmol), DIPEA (17 ⁇ L, 0.10 mmol), and DMF (1.0 mL). AM41-17A was obtained as yellow liquid in TFA salt form (27 mg, 79%).
  • AM41-18A was synthesized according to the procedures for preparing AM29-151A from intermediate 7 (18 mg, 0.03 mmol), HATU (14 mg, 0.04 mmol), 1-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)-3,6,9,12,15,18,21,24,27,30,33,36-dodecaoxanonatriacontan-39-oic acid (16 mg, 0.02 mmol), DIPEA (13 ⁇ L, 0.07 mmol), and DMF (1.0 mL). AM41-18A was obtained as yellow liquid in TFA salt form (18 mg, 66%).
  • XF034-165A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG1-CH 2 CH 2 COOH (10.6 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-165A was obtained as white solid in TFA salt form (20 mg, 98%).
  • XF034-166A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG2-CH 2 COOH (10.9 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-166A was obtained as white solid in TFA salt form (16 mg, 77%).
  • XF034-167A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG2-CH 2 CH 2 COOH (11.4 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-167A was obtained as white solid in TFA salt form (16 mg, 74%).
  • XF034-168A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG3-CH 2 CH 2 COOH (12.2 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.05 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-168A was obtained as white solid in TFA salt form (13 mg, 58%).
  • XF034-169A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG4-CH 2 CH 2 COOH (13 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-169A was obtained as white solid in TFA salt form (20 mg, 90%).
  • XF034-170A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-PEG5-CH 2 COOH (13 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-170A was obtained as white solid in TFA salt form (17 mg, 76%).
  • XY019-084 (20 mg, 49%) was synthesized according to the procedures for preparing XY019-083 from intermediate 16.
  • 1 H NMR 600 MHz, CD 3 OD
  • XF034-172A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C2-COOH (9.8 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-172A was obtained as white solid in TFA salt form (10 mg, 51%).
  • XF034-173A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C3-COOH (10 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-173A was obtained as white solid in TFA salt form (14 mg, 70%).
  • XF034-174A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C4-COOH (11 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-174A was obtained as white solid in TFA salt form (14 mg, 72%).
  • XF034-175A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C5-COOH (11 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-175A was obtained as white solid in TFA salt form (17 mg, 83%).
  • XF034-176A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C6-COOH (11 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-176A was obtained as white solid in TFA salt form (12 mg, 61%).
  • XF034-177A was synthesized according to the procedures for preparing XF034-164A from intermediate 7 (10 mg, 0.02 mmol), HOAt (3.7 mg, 0.03 mmol), VHL-C9-COOH (12 mg, 0.02 mmol), NMM (5.3 ⁇ L, 0.06 mmol), EDCI (4.3 mg, 0.03 mmol), and DMSO (1.0 mL).
  • XF034-177A was obtained as white solid in TFA salt form (9 mg, 41%).
  • YS36-49 was synthesized according to the procedures for preparing YS36-48 from intermediate 23 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-49 was obtained as white solid in TFA salt form (11 mg, 65%).
  • YS36-50 was synthesized according to the procedures for preparing YS36-48 from intermediate 24 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-50 was obtained as white solid in TFA salt form (8 mg, 46%).
  • YS36-51 was synthesized according to the procedures for preparing YS36-48 from intermediate 25 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-51 was obtained as white solid in TFA salt form (9 mg, 60%).
  • YS36-52 was synthesized according to the procedures for preparing YS36-48 from intermediate 26 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-52 was obtained as white solid in TFA salt form (13 mg, 85%).
  • YS36-53 was synthesized according to the procedures for preparing YS36-48 from intermediate 27 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-53 was obtained as white solid in TFA salt form (15 mg, 96%).
  • YS36-54 was synthesized according to the procedures for preparing YS36-48 from intermediate 28 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-54 was obtained as white solid in TFA salt form (10 mg, 60%).
  • YS36-55 was synthesized according to the procedures for preparing YS36-48 from intermediate 29 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-55 was obtained as white solid in TFA salt form (9 mg, 59%).
  • YS36-56 was synthesized according to the procedures for preparing YS36-48 from intermediate 30 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-56 was obtained as white solid in TFA salt form (9 mg, 58%).
  • YS36-57 was synthesized according to the procedures for preparing YS36-48 from intermediate 31 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-57 was obtained as white solid in TFA salt form (9 mg, 58%).
  • YS36-58 was synthesized according to the procedures for preparing YS36-48 from intermediate 32 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-58 was obtained as white solid in TFA salt form (11 mg, 70%).
  • YS36-59 was synthesized according to the procedures for preparing YS36-48 from intermediate 33 (10 mg, 0.01 mmol), HOAt (4.3 mg, 0.03 mmol), intermediate 7 (10 mg, 0.01 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (6.1 mg, 0.03 mmol), and DMSO (1.0 mL). YS36-59 was obtained as white solid in TFA salt form (12 mg, 76%).
  • AM41-37A was synthesized according to the procedures for preparing AM41-36A from AM41-35A (20 mg, 0.03 mmol), HOAt (6 mg, 0.05 mmol), 2-(adamantan-2-yl)acetic acid (7 mg, 0.04 mmol), EDCI (9 mg, 0.05 mmol), NMM (14 ⁇ L, 0.12 mmol), and DMSO (1.0 mL).
  • AM41-37A was obtained as off-white solid in TFA salt form (21 mg, 83%).
  • AM41-39A was synthesized according to the procedures for preparing AM16-103A from AM41-35A (20 mg, 0.03 mmol), intermediate 6 (16 mg, 0.09 mmol), sodium triacetoxyborohydride (26 mg, 0.12 mmol), DCM (0.5 mL), and methanol (0.5 mL). AM41-39A was obtained as white solid (6 mg, 24%).
  • AM41-41A was synthesized according to the procedures for preparing AM16-103A from AM41-35A (20 mg, 0.03 mmol), intermediate 6 (5 mg, 0.03 mmol), sodium triacetoxyborohydride (26 mg, 0.12 mmol), DCM (0.5 mL), and methanol (0.5 mL). AM41-41A was obtained as white solid (10 mg, 46%).
  • AM41-38A was synthesized according to the procedures for preparing AM29-151A from AM41-35A (20 mg, 0.03 mmol), HATU (23 mg, 0.06 mmol), 3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)propanoic acid (14 mg, 0.04 mmol), DIPEA (21 ⁇ L, 0.12 mmol), and DMF (1.0 mL).
  • AM41-38A was obtained as yellow solid in TFA salt form (16 mg, 51%).
  • AM41-40A was synthesized according to the procedures for preparing XY028-086.
  • AM41-40A was obtained as off-white solid in TFA salt form (15 mg, 44%).
  • XF042-85 was synthesized according to the procedures for preparing XF042-84 from intermediate 48 (20 mg, 0.04 mmol), HOAt (8.6 mg, 0.06 mmol), 2-adamantaneacetic acid (8.1 mg, 0.04 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (12 mg, 0.06 mmol), and DMSO (1.0 mL).
  • XF042-85 was obtained as white solid in TFA salt form (24 mg, 85%).
  • XF042-132 was synthesized according to the procedures for preparing XF042-95 from intermediate 48 (15 mg, 0.03 mmol) and intermediate 6 (5.8 mg, 0.03 mmol). XF042-132 was obtained as white solid (8.2 mg, 98%).
  • XF042-86 was synthesized according to the procedures for preparing XF042-84 from intermediate 48 (20 mg, 0.04 mmol), HOAt (8.6 mg, 0.06 mmol), 3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)propanoic acid (16.2 mg, 0.04 mmol), NMM (14 ⁇ L, 0.13 mmol), EDCI (12 mg, 0.06 mmol), and DMSO (1.0 mL). XF042-86 was obtained as yellow solid in TFA salt form (29 mg, 82%).
  • XF042-94 was synthesized according to the procedures for preparing XF042-84 from intermediate 48 (10 mg, 0.02 mmol), HOAt (4.1 mg, 0.03 mmol), intermediate 34 (19 mg, 0.02 mmol), NMM (6.5 ⁇ L, 0.06 mmol), EDCI (5.8 mg, 0.03 mmol), and DMSO (1.0 mL). XF042-94 was obtained as white solid in TFA salt form (22 mg, 81%).
  • XF042-89 was synthesized according to the procedures for preparing XF042-84 from intermediate 49 (21 mg, 0.05 mmol), HOAt (8.7 mg, 0.07 mmol), 1-adamantaneacetic acid (8.7 mg, 0.05 mmol), NMM (15 ⁇ L, 0.14 mmol), EDCI (14 mg, 0.07 mmol), and DMSO (1.0 mL). XF042-89 was obtained as white solid in TFA salt form (20 mg, 71%).
  • XF042-90 was synthesized according to the procedures for preparing XF042-84 from intermediate 2 (21 mg, 0.05 mmol), HOAt (8.7 mg, 0.07 mmol), 2-adamantaneacetic acid (8.7 mg, 0.05 mmol), NMM (15 ⁇ L, 0.14 mmol), EDCI (13.5 mg, 0.07 mmol), and DMSO (1.0 mL).
  • XF042-90 was obtained as white solid in TFA salt form (24 mg, 83%).
  • XF042-93 was synthesized according to the procedures for preparing XF042-95 from intermediate 49 (29 mg, 0.06 mmol) and intermediate 6 (33 mg, 0.19 mmol). XF042-93 was obtained as white solid (10 mg, 26%).
  • XF042-133 was synthesized according to the procedures for preparing XF042-95 from intermediate 49 (15 mg, 0.03 mmol) and intermediate 6 (5.8 mg, 0.03 mmol). XF042-133 was obtained as white solid (16.3 mg, 87%).
  • XF042-91 was synthesized according to the procedures for preparing XF042-84 from intermediate 49 (21 mg, 0.05 mmol), HOAt (9.2 mg, 0.07 mmol), 3-(2-((2-(2,6-dioxopiperidin-3-yl)-1,3-dioxoisoindolin-4-yl)amino)ethoxy)propanoic acid (18 mg, 0.05 mmol), NMM (15 ⁇ L, 0.14 mmol), EDCI (14 mg, 0.06 mmol), and DMSO (1.0 mL). XF042-91 was obtained as yellow solid in TFA salt form (33 mg, 89%).
  • XF042-92 was synthesized according to the procedures for preparing XF042-84 from intermediate 49 (11 mg, 0.02 mmol), HOAt (4.5 mg, 0.03 mmol), intermediate 34 (21 mg, 0.02 mmol), NMM (7.2 ⁇ L, 0.07 mmol), EDCI (6.3 mg, 0.03 mmol), and DMSO (1.0 mL). XF042-92 was obtained as white solid in TFA salt form (19 mg, 60%).
  • GI 50 concentration for 50% of maximal inhibition of cell proliferation
  • GI 50 GI 50 ( ⁇ M) Cmpd ( ⁇ M) MDA- GI 50 ( ⁇ M) GI 50 ( ⁇ M) # Structure MCF-7 MB-468 HCC1187 HCC1170 AM16- 10A 1.2 1.4 0.57 1.2 AM16- 11A N/A N/A 2.6 N/A AM16- 37A N/A N/A 2.1 N/A AM16- 38A N/A N/A 2.0 N/A XY019- 43 0.35 0.54 0.65 N/A XY019- 44 N/A N/A 2.2 N/A AM16- 92A 1.0 N/A 1.1 N/A AM16- 93A N/A N/A 1.5 N/A AM16- 97A N/A N/A 1.2 N/A AM16- 101A 0.69 1.1 N/A N/A AM16- 105A 2.3 N/A 1.2 N/A AM16- 106A N/A N/A 4.3 N/A AM29- 21A 0.28 0.20 N
  • FIG. 50 Cellular EZH2 and H3K27me3 levels in MCF-7 cells treated with XY019-43 or UNC1999 (negative control) at 1 ⁇ M are shown in FIG. 50 .
  • Cellular EZH2 and H3K27me3 levels in MDA-MB-468 cells treated with XY019-43, AM19-182A, AM29-177, or UNC1999 (negative control) at various concentrations for various time points are shown in FIGS. 51-53 .
  • FIG. 54 cellular EZH2 and H3K27me3 levels in HCC1187 cells treated with 1 ⁇ M AM16-10A or UNC1999 (negative control) for various time points are shown in FIG. 54 .
  • Methyltransferase activity assays were performed by monitoring the incorporation of tritiumlabeled methyl group from S-adenosylmethionine ( 3 H-SAM) to biotinylated peptide substrates using Scintillation Proximity Assay (SPA) for EZH2/PRC2 5-component complex.
  • 3 H-SAM S-adenosylmethionine
  • SPA Scintillation Proximity Assay
  • EZH2/PRC2 5-component complex Compounds were dissolved in DMSO to a stock concentration of 10 mM. Compounds were tested in a 10-dose IC 50 mode with 3-fold serial dilution, in duplicate, at 10 ⁇ M. Reactions were carried out at 1 ⁇ M SAM and 5 ⁇ M Histone H3. Results for AM16-10A, XY019-43 and AM16-101A are shown in FIGS. 55-57 .
US16/345,591 2016-10-28 2017-10-27 Compositions and methods for treating ezh2-mediated cancer Abandoned US20190255041A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/345,591 US20190255041A1 (en) 2016-10-28 2017-10-27 Compositions and methods for treating ezh2-mediated cancer

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662414195P 2016-10-28 2016-10-28
US16/345,591 US20190255041A1 (en) 2016-10-28 2017-10-27 Compositions and methods for treating ezh2-mediated cancer
PCT/US2017/058718 WO2018081530A1 (fr) 2016-10-28 2017-10-27 Compositions et procédés pour le traitement du cancer à médiation par l'ezh2

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/058718 A-371-Of-International WO2018081530A1 (fr) 2016-10-28 2017-10-27 Compositions et procédés pour le traitement du cancer à médiation par l'ezh2

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/926,418 Continuation US11510920B2 (en) 2016-10-28 2020-07-10 Compositions and methods for treating EZH2-mediated cancer

Publications (1)

Publication Number Publication Date
US20190255041A1 true US20190255041A1 (en) 2019-08-22

Family

ID=62024052

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/345,591 Abandoned US20190255041A1 (en) 2016-10-28 2017-10-27 Compositions and methods for treating ezh2-mediated cancer
US16/926,418 Active US11510920B2 (en) 2016-10-28 2020-07-10 Compositions and methods for treating EZH2-mediated cancer
US17/978,696 Pending US20230114294A1 (en) 2016-10-28 2022-11-01 Compositions and methods for treating ezh2-mediated cancer

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/926,418 Active US11510920B2 (en) 2016-10-28 2020-07-10 Compositions and methods for treating EZH2-mediated cancer
US17/978,696 Pending US20230114294A1 (en) 2016-10-28 2022-11-01 Compositions and methods for treating ezh2-mediated cancer

Country Status (12)

Country Link
US (3) US20190255041A1 (fr)
EP (1) EP3532464A4 (fr)
JP (1) JP2019537585A (fr)
KR (1) KR20190084063A (fr)
CN (1) CN110167924A (fr)
AU (2) AU2017348322B8 (fr)
CA (1) CA3041840A1 (fr)
EA (1) EA201991071A1 (fr)
IL (1) IL266053A (fr)
MX (1) MX2019004950A (fr)
SG (2) SG11201903483VA (fr)
WO (1) WO2018081530A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10978068B2 (en) * 2016-10-27 2021-04-13 Samsung Electronics Co., Ltd. Method and apparatus for executing application on basis of voice commands
US11472799B2 (en) 2018-03-06 2022-10-18 Icahn School Of Medicine At Mount Sinai Serine threonine kinase (AKT) degradation / disruption compounds and methods of use
US11510920B2 (en) 2016-10-28 2022-11-29 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating EZH2-mediated cancer
US11541051B2 (en) 2016-12-08 2023-01-03 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating CDK4/6-mediated cancer
US20230346953A1 (en) * 2021-04-30 2023-11-02 Northwestern University Substituted 3-amino-5-phenylbenzamide compounds as covalent inhibitors of enhancer zeste homolog 2 (ezh2) and proteolysis-targeting chimeric derivatives thereof (protacs) that induce degradation of ezh2

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11191741B2 (en) 2016-12-24 2021-12-07 Arvinas Operations, Inc. Compounds and methods for the targeted degradation of enhancer of zeste homolog 2 polypeptide
LT3784663T (lt) 2018-04-23 2023-10-25 Celgene Corporation Pakeistieji 4-aminoizoindolin-1,3-diono junginiai ir jų naudojimas limfomos gydymui
CN112457365B (zh) * 2019-09-09 2023-06-09 中国医学科学院药物研究所 一类靶向蛋白质水解通路的功能分子及其制备和应用
CN110862434B (zh) * 2019-10-28 2021-07-06 中山大学 一种可促进TGF-β1蛋白靶向降解的化合物及其制备方法与应用
KR102505664B1 (ko) * 2019-10-31 2023-03-03 재단법인 대구경북첨단의료산업진흥재단 Ezh2 저해제 및 e3 리가제 바인더를 포함하는 화합물 및 이를 유효성분으로 함유하는 ezh2 관련 질환의 예방 또는 치료용 약학적 조성물
CN111303133A (zh) * 2020-03-25 2020-06-19 清华大学 降解ezh2蛋白的小分子化合物
CN112920176B (zh) * 2020-05-25 2022-11-04 四川大学华西医院 可诱导prc2蛋白复合物核心亚基降解的双功能化合物和药物组合物及应用
JP7387907B2 (ja) * 2020-08-13 2023-11-28 ハンミ ファーマシューティカル カンパニー リミテッド 新規なジオキソロイソキノリノン誘導体化合物及びその用途{novel dioxoloisoquinolinone derivatives and use thereof}
CN113214203A (zh) * 2020-12-16 2021-08-06 中山大学肿瘤防治中心(中山大学附属肿瘤医院、中山大学肿瘤研究所) 基于ezh2蛋白降解的小分子化合物及其应用
WO2022159650A1 (fr) * 2021-01-22 2022-07-28 Icahn School Of Medicine At Mount Sinai Composés hétérobifonctionnels utilisés en tant qu'agents de dégradation de l'eef1a2
WO2023205701A1 (fr) 2022-04-20 2023-10-26 Kumquat Biosciences Inc. Hétérocycles macrocycliques et leurs utilisations

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5691147A (en) 1994-06-02 1997-11-25 Mitotix, Inc. CDK4 binding assay
US6486214B1 (en) 1997-09-10 2002-11-26 Rutgers, The State University Of New Jersey Polyanhydride linkers for production of drug polymers and drug polymer compositions produced thereby
WO2001060814A2 (fr) 2000-02-15 2001-08-23 Sugen, Inc. Inhibiteurs de la proteine kinase 2-indolinone a substitution pyrrole
JP5567339B2 (ja) 2006-09-15 2014-08-06 セルジーン コーポレイション N−メチルアミノメチルイソインドール化合物、それを含む組成物、及びその使用方法
WO2008109104A1 (fr) 2007-03-07 2008-09-12 The Regents Of The University Of California Inhibiteurs bivalents de protéine kinase akt
US8377937B2 (en) 2007-07-05 2013-02-19 Array Biopharma Inc. Pyrimidyl cyclopentanes as AKT protein kinase inhibitors
US20110172107A1 (en) 2008-04-30 2011-07-14 Fox Chase Cancer Center Assay for identifying agents that modulate epigenetic silencing, and agents identified thereby
CN102984941B (zh) 2009-09-04 2016-08-17 密执安大学评议会 用于治疗白血病的组合物和方法
RS58523B1 (sr) 2010-02-11 2019-04-30 Celgene Corp Derivati arilmetoksi izoindolina i kombinacije koje ih obuhvataju i postupci njihove upotrebe
WO2012078492A1 (fr) 2010-12-06 2012-06-14 Celgene Corporation Traitement d'association avec du lénalidomide et un inhibiteur de cdk pour traiter le myélome multiple
KR102204989B1 (ko) 2012-01-12 2021-01-20 예일 유니버시티 E3 유비퀴틴 리가아제에 의한 표적 단백질 및 다른 폴리펩티드의 증진된 분해를 위한 화합물 및 방법
WO2013170147A1 (fr) 2012-05-11 2013-11-14 Yale University Composés utiles pour stimuler la dégradation des protéines et procédés utilisant ceux-ci
WO2014063061A1 (fr) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Petites molécules marquées de façon hydrophobe en tant qu'inducteurs de la dégradation de protéine
HUE040323T2 (hu) 2012-12-21 2019-02-28 Epizyme Inc PRMT5-inhibitorok és alkalmazásaik
RU2670762C2 (ru) 2013-12-31 2018-10-25 Хуаньчжу Фарма Ко., Лтд. Производные 6-(пиримидиноаминопиридин)бензоимидазола, полезные для лечения рака
WO2015104677A1 (fr) * 2014-01-10 2015-07-16 Piramal Enterprises Limited Composés hétérocycliques en tant qu'inhibiteurs de ezh2
MX2016013563A (es) * 2014-04-14 2017-05-09 Arvinas Inc Moduladores de la proteolisis basados en imida y metodos de uso asociados.
CA2952051C (fr) 2014-06-13 2023-09-12 Trustees Of Tufts College Agents therapeutiques actives par fap, et utilisations associees
CA2964629A1 (fr) * 2014-11-06 2016-05-12 Dana-Farber Cancer Institute, Inc. Inhibiteurs d'ezh2 et leurs utilisations
US10479997B2 (en) 2014-12-01 2019-11-19 Novartis Ag Compositions and methods for diagnosis and treatment of prostate cancer
CN107257800B (zh) * 2014-12-23 2020-06-30 达纳-法伯癌症研究所股份有限公司 通过双功能分子诱导靶蛋白降解的方法
WO2016106518A1 (fr) * 2014-12-29 2016-07-07 深圳市日上光电股份有限公司 Boîtier de lampe d'éclairage intégré de ligne conductrice
TWI698436B (zh) 2014-12-30 2020-07-11 美商佛瑪治療公司 作為泛素特異性蛋白酶7抑制劑之吡咯并及吡唑并嘧啶
EP3244927A1 (fr) 2015-01-16 2017-11-22 Vedantra Pharmaceuticals Inc. Compositions de vésicules lipidiques multilamellaires comprenant un variant de kinase du lymphome anaplasique (alk) conjugué et leurs utilisations
AU2016232705C1 (en) * 2015-03-18 2021-06-17 Arvinas, Inc. Compounds and methods for the enhanced degradation of targeted proteins
US10988737B2 (en) 2015-04-28 2021-04-27 Universite De Strasbourg Clinical gene signature-based human cell culture model and uses thereof
RU2719477C2 (ru) 2015-06-22 2020-04-17 Оно Фармасьютикал Ко., Лтд. Соединение, ингибирующее brk
CN105085620B (zh) 2015-06-25 2018-05-08 中山大学附属第一医院 一种靶向泛素化降解Smad3的化合物
WO2017007612A1 (fr) 2015-07-07 2017-01-12 Dana-Farber Cancer Institute, Inc. Procédés pour induire la dégradation ciblée de protéines par des molécules bifonctionnelles
WO2017011371A1 (fr) * 2015-07-10 2017-01-19 Arvinas, Inc Modulateurs de protéolyse à base de mdm2 et méthodes d'utilisation associées
WO2017011590A1 (fr) * 2015-07-13 2017-01-19 Arvinas, Inc. Modulateurs de protéolyse à base d'alanine et procédés d'utilisation associés
US9809603B1 (en) 2015-08-18 2017-11-07 Deuterx, Llc Deuterium-enriched isoindolinonyl-piperidinonyl conjugates and oxoquinazolin-3(4H)-yl-piperidinonyl conjugates and methods of treating medical disorders using same
CA2996685A1 (fr) 2015-09-03 2017-03-09 Aileron Therapeutics, Inc. Macrocycles peptidomimetiques et leurs utilisations
RU2018120330A (ru) * 2015-11-02 2019-12-04 Йейл Юниверсити Химерные соединения, осуществляющие нацеливание для протеолиза, и способы их получения и применения
US11174250B2 (en) 2016-03-01 2021-11-16 Propellon Therapeutics Inc. Substituted carboxamides as inhibitors of WDR5 protein-protein binding
EA038109B1 (ru) 2016-03-01 2021-07-07 Пропеллон Терапьютикс Инк. Ингибиторы связывания белка wdr5 с белками
CN109071552B (zh) 2016-04-22 2022-06-03 达纳-法伯癌症研究所公司 细胞周期蛋白依赖性激酶4/6(cdk4/6)通过cdk4/6抑制剂与e3连接酶配体的缀合的降解及使用方法
WO2017197051A1 (fr) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Dégronimères de c3-glutarimide liés à une amine pour la dégradation de protéines cibles
WO2017197055A1 (fr) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Dégronimères hétérocycliques pour la dégradation de protéines cibles
US20180072741A1 (en) 2016-09-09 2018-03-15 Incyte Corporation Pyrazolopyrimidine compounds and uses thereof
US10160763B2 (en) 2016-09-13 2018-12-25 Vanderbilt University WDR5 inhibitors and modulators
US20190255041A1 (en) 2016-10-28 2019-08-22 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating ezh2-mediated cancer
AU2017370694A1 (en) 2016-12-08 2019-07-25 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating CDK4/6-mediated cancer
US11052091B2 (en) 2016-12-21 2021-07-06 Ono Pharmaceutical Co., Ltd. BRK inhibitory compound
WO2019113071A1 (fr) 2017-12-05 2019-06-13 Icahn School Of Medicine At Mount Sinai Compositions et méthodes de traitement de cancer à médiation par alk
WO2019165189A1 (fr) 2018-02-22 2019-08-29 Icahn School Of Medicine At Mount Sinai Composés de dégradation/perturbation de la protéine arginine méthyltransférase 5 (prmt5) et méthodes d'utilisation
CA3092677A1 (fr) 2018-03-06 2019-09-12 Icahn School Of Medicine At Mount Sinai Composes de degradation/interruption de serine threonine kinase (akt) et procedes d'utilisation
WO2019222380A1 (fr) 2018-05-15 2019-11-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Formulations et méthodes de prévention et de traitement des métastases tumorales et de la tumorigenèse
EP3810145A4 (fr) 2018-06-21 2022-06-01 Icahn School of Medicine at Mount Sinai Composés de dégradation/désintégration de la protéine 5 à domaine de répétition wd40 (wdr5) et méthodes d'utilisation
CN114269749A (zh) 2019-06-10 2022-04-01 苏特罗生物制药公司 5H-吡咯并[3,2-d]嘧啶-2,4-二氨基化合物及其抗体偶联物
WO2021021904A1 (fr) 2019-07-30 2021-02-04 The Scripps Research Institute Inhibiteurs pharmacologiques du domaine enl yeats
US20210395244A1 (en) 2020-06-01 2021-12-23 Icahn School Of Medicine At Mount Sinai Mitogen-activated protein kinase kinase (mek) degradation compounds and methods of use
CN112778303A (zh) 2020-12-31 2021-05-11 武汉九州钰民医药科技有限公司 Cdk4/6激酶抑制剂shr6390的制备方法

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10978068B2 (en) * 2016-10-27 2021-04-13 Samsung Electronics Co., Ltd. Method and apparatus for executing application on basis of voice commands
US11510920B2 (en) 2016-10-28 2022-11-29 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating EZH2-mediated cancer
US11541051B2 (en) 2016-12-08 2023-01-03 Icahn School Of Medicine At Mount Sinai Compositions and methods for treating CDK4/6-mediated cancer
US11472799B2 (en) 2018-03-06 2022-10-18 Icahn School Of Medicine At Mount Sinai Serine threonine kinase (AKT) degradation / disruption compounds and methods of use
US20230346953A1 (en) * 2021-04-30 2023-11-02 Northwestern University Substituted 3-amino-5-phenylbenzamide compounds as covalent inhibitors of enhancer zeste homolog 2 (ezh2) and proteolysis-targeting chimeric derivatives thereof (protacs) that induce degradation of ezh2

Also Published As

Publication number Publication date
AU2017348322B8 (en) 2021-12-23
KR20190084063A (ko) 2019-07-15
EP3532464A1 (fr) 2019-09-04
US11510920B2 (en) 2022-11-29
JP2019537585A (ja) 2019-12-26
CA3041840A1 (fr) 2018-05-03
SG10202104386UA (en) 2021-05-28
AU2017348322A8 (en) 2021-12-23
AU2022201488A1 (en) 2022-03-24
SG11201903483VA (en) 2019-05-30
AU2017348322A1 (en) 2019-06-13
US20230114294A1 (en) 2023-04-13
EA201991071A1 (ru) 2019-11-29
AU2017348322B2 (en) 2021-12-09
CN110167924A (zh) 2019-08-23
IL266053A (en) 2019-06-30
US20200338070A1 (en) 2020-10-29
EP3532464A4 (fr) 2020-07-08
WO2018081530A1 (fr) 2018-05-03
MX2019004950A (es) 2019-09-26

Similar Documents

Publication Publication Date Title
US11510920B2 (en) Compositions and methods for treating EZH2-mediated cancer
US11541051B2 (en) Compositions and methods for treating CDK4/6-mediated cancer
CN109153675B (zh) 细胞周期蛋白依赖性激酶9通过cdk9抑制剂与e3连接酶配体的缀合的降解及使用方法
JP6882299B2 (ja) 多環式tlr7/8アンタゴニスト及び免疫障害の治療におけるそれらの使用
CA2852860C (fr) Peptides cytotoxiques et conjugues anticorps-medicaments de ceux-ci
JP6976618B2 (ja) 活性酸素種スカベンジャーの調製および使用
US20210047367A1 (en) Preparation and uses of reactive oxygen species scavenger derivatives
WO2021098859A1 (fr) Inhibiteur à cycle aza à sept chaînons, et son procédé de préparation et utilisation associée
US20230132823A1 (en) Bi-functional compounds and methods for targeted ubiquitination of androgen receptor
CA3096424A1 (fr) Derives de pladienolide en tant qu'agents ciblant un complexe d'epissage pour le traitement du cancer
US20230141981A1 (en) Novel compounds and composition for targeted therapy of kidney-associated cancers
JP2018509424A (ja) 疾患を処置するためのコンジュゲート
JP2022549353A (ja) プロテインキナーゼc(pkc)に作用するジテルペノイド化合物
EP3791897A1 (fr) Conjugués de type inhibiteur de hsp90-médicament
AU2015256266B2 (en) BH4 antagonists and methods related thereto
US11839659B2 (en) Proteolysis-targeting chimeric molecules (PROTACs) that induce degradation of indoleamine 2,3-dioxygenase (IDO) protein
RU2815064C2 (ru) Производные пладиенолида в качестве средств, оказывающих целенаправленное воздействие на сплайсосому, для лечения рака
WO2023155808A1 (fr) Conjugué d'anticorps-éribuline ou un dérivé de celui-ci, intermédiaire de celui-ci, son procédé de préparation, composition pharmaceutique de celui-ci et son utilisation
WO2022184103A1 (fr) Composé tricyclique et composition pharmaceutique et son utilisation
TW202342041A (zh) 一種藥物組合物及所含活性成分化合物的製備方法
AU2021284369A1 (en) Quinazoline-derived HCK inhibitors for use in the treatment of MYD88 mutated diseases
WO2023220722A2 (fr) Agents de dégradation de pak1 et leurs procédés d'utilisation
TW202313092A (zh) 細胞可穿透環狀肽及其用途

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: ICAHN SCHOOL OF MEDICINE AT MOUNT SINAI, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JIN, JIAN;PARSONS, RAMON;STRATIKOPOULOS, ILIAS;AND OTHERS;SIGNING DATES FROM 20191025 TO 20191114;REEL/FRAME:051005/0905

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION