METHODS OF TREATING DISORDERS

Abstract
The present invention relates to methods and compositions for the treatment of BAF-related disorders such as cancers and viral infections.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Jun. 20, 2019, is named 51121-023W02_Sequence_Listing_6.20.2019_ST25 and is 180,048 bytes in size.


BACKGROUND

Disorders can be affected by the BAF complex. BICRA is a component of the BAF complex. The present invention relates to useful methods and compositions for the treatment of BAF-related disorders, such as cancer and infection.


SUMMARY

BRD4 Interacting Chromatin Remodeling Complex Associated protein (BICRA) is a protein encoded by the BICRA gene on chromosome 19. BICRA is a component of the BAF (BRG1- or BRM-associated factors) complex, a SWI/SNF ATPase chromatin remodeling complex. BICRA is present in several SWI/SNF ATPase chromatin remodeling complexes and is upregulated in multiple cancer cell lines. Accordingly, agents which reduce the levels and/or activity of BICRA may provide new methods for the treatment of disease and disorders, such as cancer. Depleting BICRA in cells may result in the depletion of the SS18-SSX fusion protein in those cells. The SS18-SSX fusion protein has been detected in more than 95% of synovial sarcoma tumors and is often the only cytogenetic abnormality in synovial sarcoma. Thus, agents that degrade BICRA, e.g., antibodies, enzymes, polynucleotides, and compounds, may be useful in the treatment of cancers related to BICRA or SS18-SSX expression such as soft tissue sarcomas, e.g., synovial sarcoma.


The present disclosure features useful methods to treat cancer, e.g., in a subject in need thereof. In some embodiments, the methods described herein are useful in the treatment of disorders associated with BICRA expression, e.g., soft tissue sarcomas, e.g., adult soft tissue sarcomas. In some embodiments, the methods described herein are useful in the treatment of disorders associated with SS18-SSX fusion protein.


In one aspect, the invention features a method of treating soft tissue sarcoma (e.g., adult soft tissue sarcoma) in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the sarcoma.


In another aspect, the invention features a method of treating soft tissue sarcoma (e.g., adult soft tissue sarcoma) in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of a BAF complex (e.g., GBAF) in the sarcoma.


In another aspect, the invention features a method of reducing tumor growth of a (soft tissue sarcoma (e.g., an adult soft tissue sarcoma) in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the tumor.


In another aspect, the invention features a method of inducing apoptosis in a soft tissue sarcoma (e.g., an adult soft tissue sarcoma) cell, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell.


In another aspect, the invention features a method of reducing the level of BICRA in a soft tissue sarcoma (e.g., an adult soft tissue sarcoma) cell, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell.


In some embodiments of any of the above aspects, the soft tissue sarcoma (e.g., adult soft tissue sarcoma) cell is in a subject. In some embodiments, the subject or cell has been identified as expressing SS18-SSX fusion protein or BICRA fusion protein.


In another aspect, the invention features a method of modulating the level of an SS18-SSX fusion protein, SS18 wild-type protein, or SSX wild-type protein in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in a cell or subject. In some embodiments, the cell is in a subject.


In another aspect, the invention features a method of treating a disorder related to an SS18-SSX fusion protein, SS18 wild-type protein, or SSX wild-type protein in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in an SS18-SSX fusion protein-expressing cell in the subject.


In some embodiments of any of the above aspects, the effective amount of the agent reduces the level and/or activity of BICRA by at least 5% (e.g., 6%, 7%, 8%, 8%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) as compared to a reference. In some embodiments, the effective amount of the agent that reduces the level and/or activity of BICRA by at least 50% (e.g., 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) as compared to a reference. In some embodiments, the effective amount of the agent that reduces the level and/or activity of BICRA by at least 90% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%).


In some embodiments, the effective amount of the agent reduces the level and/or activity of BICRA by at least 5% (e.g., 6%, 7%, 8%, 8%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) as compared to a reference for at least 12 hours (e.g., 14 hours, 16 hours, 18 hours, 20 hours, 22 hours, 24 hours, 30 hours, 36 hours, 48 hours, 72 hours, or more). In some embodiments, the effective amount of the agent that reduces the level and/or activity of BICRA by at least 5% (e.g., 6%, 7%, 8%, 8%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95%) as compared to a reference for at least 4 days (e.g., 5 days, 6 days, 7 days, 14 days, 28 days, or more).


In some embodiments, the subject has cancer. In some embodiments, the cancer expresses SS18-SSX fusion protein and/or the cell or subject has been identified as expressing SS18-SSX fusion protein. In some embodiments, the disorder is synovial sarcoma or Ewing's sarcoma. In some embodiments, the disorder is synovial sarcoma.


In one aspect, the invention features a method of modulating the activity of a BAF complex in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.


In another aspect, the invention features a method of increasing the level of BAF47 in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.


In one aspect, the invention features a method of decreasing Wnt/β-catenin signaling in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.


In one aspect, the invention features a method treating a disorder related to BAF47 in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the subject.


In some embodiments, the disorder related to BAF47 is a cancer or viral infection. In some embodiments, the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer.


In some embodiments, the viral infection is an infection with a virus of the Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or Togaviridae family.


In an aspect, the invention features a method for treating cancer in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a cancer cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.


In an aspect, the invention features a method of reducing tumor growth of a cancer in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a tumor cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.


In another aspect, the invention features a method of inducing apoptosis in a cancer cell, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.


In another aspect, the invention features a method of reducing the level of BICRA in a cancer cell, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.


In some embodiments of any of the foregoing aspects, the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer. In some embodiments, the cancer is non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.


In one aspect, the invention features a method of modulating the activity of a BICRA fusion protein in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.


In another aspect, the invention features a method of modulating the level of a BICRA fusion protein in a cell or subject, the method including contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject. In some embodiments, the cell is in a subject.


In another aspect, the invention features a method of treating a disorder related to a BICRA fusion protein in a subject in need thereof, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a BICRA fusion protein-expressing cell.


In some embodiments of any of the above aspects, the subject has cancer. In some embodiments, the cancer expresses a BICRA fusion protein and/or the cell or subject has been identified as expressing a BICRA fusion protein. In some embodiments, the method further includes administering to the subject or contacting the cell with an anticancer therapy. In some embodiments, the anticancer therapy is a chemotherapeutic or cytotoxic agent or radiotherapy. In some embodiments, the chemotherapeutic or cytotoxic agent is doxorubicin or ifosfamide. In some embodiments, the anticancer therapy and the agent that reduces the level and/or activity of BICRA in a cell are administered within 28 days of each other and each in an amount that together are effective to treat the subject. In some embodiments, the subject or cancer has been identified as having an elevated level of an SS18-SSX fusion protein or a BICRA fusion protein as compared to a reference. In some embodiments, the subject or cancer has been identified as having a decreased level of SS18 wild-type protein or SSX wild-type protein as compared to a reference.


In one aspect, the invention features a method of treating a viral infection, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a cell of the subject.


In some embodiments, the disorder is a viral infection is an infection with a virus of the Retroviridae family such as the lentiviruses (e.g., Human immunodeficiency virus (HIV) and deltaretroviruses (e.g., human T cell leukemia virus I (HTLV-I), human T cell leukemia virus II (HTLV-II)), Hepadnaviridae family (e.g., hepatitis B virus (HBV)), Flaviviridae family (e.g., hepatitis C virus (HCV)), Adenoviridae family (e.g., Human Adenovirus), Herpesviridae family (e.g., Human cytomegalovirus (HCMV), Epstein-Barr virus, herpes simplex virus 1 (HSV-1), herpes simplex virus 2 (HSV-2), human herpesvirus 6 (HHV-6), Herpesvitus K*, CMV, varicella-zoster virus), Papillomaviridae family (e.g., Human Papillomavirus (HPV, HPV E1)), Parvoviridae family (e.g., Parvovirus B19), Polyomaviridae family (e.g., JC virus and BK virus), Paramyxoviridae family (e.g., Measles virus), Togaviridae family (e.g., Rubella virus). In some embodiments, the disorder is Coffin Siris, Neurofibromatosis (e.g., NF-1, NF-2, or Schwannomatosis), or Multiple Meningioma. In some embodiments, the viral infection is an infection with a virus of the Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or Togaviridae family.


In some embodiments of any of the above aspects, the agent that reduces the level and/or activity of BICRA in a cell is a small molecule compound, an antibody, an enzyme, and/or a polynucleotide. In some embodiments, the agent that reduces the level and/or activity of BICRA in a cell is an enzyme. In some embodiments, the enzyme is a clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein, a zinc finger nuclease (ZFN), a transcription activator-like effector nuclease (TALEN), or a meganuclease. In some embodiments, the CRISPR-associated protein is CRISPR-associated protein 9 (Cas9).


In some embodiments of any of the above aspects, the agent that reduces the level and/or activity of BICRA in a cell is a polynucleotide. In some embodiments, the polynucleotide is an antisense nucleic acid, a short interfering RNA (siRNA), a short hairpin RNA (shRNA), a CRISPR/Cas 9 nucleotide (e.g., a guide RNA (gRNA)), or a ribozyme. In some embodiments, the polynucleotide has a sequence having at least 70% sequence identity (e.g., 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9% identity, or more) to the nucleic acid sequence of any one of SEQ ID NOs: 3-124. In some embodiments, the polynucleotide comprises a sequence having at least 70% sequence identity (e.g., 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9% identity, or more) to the nucleic acid sequence of any one of SEQ ID NOs: 3-68.


In some embodiments of any of the above aspects, the agent that reduces the level and/or activity of BICRA in a cell is a small molecule compound, or a pharmaceutically acceptable salt thereof.


In some embodiments, the small molecule compound, or a pharmaceutically acceptable salt thereof is a degrader. In some embodiments, the degrader has the structure of Formula I:





A-L-B   Formula I


wherein A is a BICRA binding moiety; L is a linker; and B is a degradation moiety, or a pharmaceutically acceptable salt thereof. In some embodiments, the degradation moiety is a ubiquitin ligase moiety. In some embodiments, the ubiquitin ligase binding moiety includes Cereblon ligands, IAP (Inhibitors of Apoptosis) ligands, mouse double minute 2 homolog (MDM2), hydrophobic tag, or von Hippel-Lindau ligands, or derivatives or analogs thereof.


In some embodiments, the hydrophobic tag includes a diphenylmethane, adamantine, or tri-Boc arginine, i.e., the hydrophobic tag includes the structure:




embedded image


In some embodiments, the ubiquitin ligase binding moiety includes the structure of Formula A:




embedded image


wherein X1 is CH2, O, S, or NR1, wherein R1 is H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; X2 is C═O, CH2, or




embedded image


R3 and R4 are, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; m is 0, 1, 2, 3, or 4; and each R2 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure:




embedded image


or is a derivative or an analog thereof, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure of Formula B:




embedded image


wherein each R4, R4′, and R7 is, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R5 is optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; R6 is H, optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; n is 0, 1, 2, 3, or 4; each R8 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; and each R9 and R10 is, independently, H, halogen, optionally substituted C1-C6 alkyl, or optionally substituted C6-C10 aryl, wherein R4′ or R5 comprises a bond to the linker, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure:




embedded image


or is a derivative or analog thereof, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure of Formula C:




embedded image


wherein each R11, R13, and R15 is, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R12 is optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; R14 is optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; p is 0, 1, 2, 3, or 4; each R16 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; q is 0, 1, 2, 3, or 4; and each R17 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure:




embedded image


or is a derivative or an analog thereof, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure of Formula D:




embedded image


wherein each R18 and R19 is, independently, H, optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; r1 is 0, 1, 2, 3, or 4; each R20 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; r2 is 0, 1, 2, 3, or 4; and each R21 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino, or a pharmaceutically acceptable salt thereof.


In some embodiments, the ubiquitin ligase binding moiety includes the structure:




embedded image


or is a derivative or an analog thereof, or a pharmaceutically acceptable salt thereof.


In some embodiments, the linker has the structure of Formula II:





A1-(B1)f—(C1)g—(B2)h-(D)-(B3)i—(C2)j—(B4)k-A2   Formula II


wherein A1 is a bond between the linker and A; A2 is a bond between B and the linker; B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl, optionally substituted C1-C3 heteroalkyl, O, S, S(O)2, and NRN; RN is hydrogen, optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C2-6 heterocyclyl, optionally substituted C6-12 aryl, or optionally substituted C1-7 heteroalkyl; C1 and C2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, l, j, and k are each, independently, 0 or 1; and D is optionally substituted C1-10 alkyl, optionally substituted C2-10 alkenyl, optionally substituted C2-10 alkynyl, optionally substituted C2-6 heterocyclyl, optionally substituted C6-12 aryl, optionally substituted C2-C10 polyethylene glycol, or optionally substituted C1-10 heteroalkyl, or a chemical bond linking A1-(B1)f—(C1)g—(B2)h— to —(B3)i—(C2)j—(B4)k-A2.


In some embodiments, D is optionally substituted C2-C10 polyethylene glycol. In some embodiments, C1 and C2 are each, independently, a carbonyl or sulfonyl. In some embodiments, B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl, optionally substituted C1-C3 heteroalkyl, O, S, S(O)2, and NRN; RN is hydrogen or optionally substituted C1-4 alkyl. In some embodiments, B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl or optionally substituted C1-C3 heteroalkyl. In some embodiments, j is 0. In some embodiments, k is 0. In some embodiments, j and k are each, independently, 0. In some embodiments, f, g, h, and i are each, independently, 1.


In some embodiments, the linker of Formula II has the structure of Formula IIa:




embedded image


wherein A1 is a bond between the linker and A, and A2 is a bond between B and the linker.


In some embodiments, D is optionally substituted C1-10 alkyl. In some embodiments, C1 and C2 are each, independently, a carbonyl. In some embodiments, B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl, optionally substituted C1-C3 heteroalkyl, O, S, S(O)2, and NRN, wherein RN is hydrogen or optionally substituted C1-4 alkyl. In some embodiments, B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl, O, S, S(O)2, and NRN, wherein RN is hydrogen or optionally substituted C1-4 alkyl. In some embodiments, B1 and B4 each, independently, is optionally substituted C1-C2 alkyl. In some embodiments, B1 and B4 each, independently, is C1 alkyl. In some embodiments, B2 and B4 each, independently, is NRN, wherein RN is hydrogen or optionally substituted C1-4 alkyl. In some embodiments, B2 and B4 each, independently, is NH. In some embodiments, f, g, h, l, j, and k are each, independently, 1.


In some embodiments, the linker of Formula II has the structure of Formula Mb:




embedded image


wherein A1 is a bond between the linker and A, and A2 is a bond between B and the linker.


In an aspect, the invention features a method of treating cancer in a subject, the method including: (a) determining the level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein in the subject; and (b) administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a cell or subject if the subject has an elevated level of SS18-SSX fusion protein or BICRA fusion protein or a decreased level of SS18 wild-type protein or SSX wild-type protein as compared to a reference. In a related aspect, the invention features a method of treating cancer in a subject determined to have an elevated level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein, the method including administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.


In some embodiments, the level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein in the subject is measured in one or more cancer cells. In some embodiments, the level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein in the subject is measured systemically.


In one aspect, the invention features a composition including an adult soft tissue sarcoma cell and an agent that reduces the level and/or activity of BICRA in a cell.


Chemical Terms

For any of the following chemical definitions, a number following an atomic symbol indicates that total number of atoms of that element that are present in a particular chemical moiety. As will be understood, other atoms, such as hydrogen atoms, or substituent groups, as described herein, may be present, as necessary, to satisfy the valences of the atoms. For example, an unsubstituted C2 alkyl group has the formula —CH2CH3. When used with the groups defined herein, a reference to the number of carbon atoms includes the divalent carbon in acetal and ketal groups but does not include the carbonyl carbon in acyl, ester, carbonate, or carbamate groups. A reference to the number of oxygen, nitrogen, or sulfur atoms in a heteroaryl group only includes those atoms that form a part of a heterocyclic ring.


The term “acyl,” as used herein, represents a hydrogen or an alkyl group that is attached to a parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group), acetyl, trifluoroacetyl, propionyl, and butanoyl. Exemplary unsubstituted acyl groups include from 1 to 6, from 1 to 11, or from 1 to 21 carbons.


The term “alkyl,” as used herein, refers to a branched or straight-chain monovalent saturated aliphatic hydrocarbon radical of 1 to 20 carbon atoms (e.g., 1 to 16 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms).


An alkylene is a divalent alkyl group. The term “alkenyl,” as used herein, alone or in combination with other groups, refers to a straight chain or branched hydrocarbon residue having a carbon-carbon double bond and having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms).


The term “alkynyl,” as used herein, alone or in combination with other groups, refers to a straight chain or branched hydrocarbon residue having a carbon-carbon triple bond and having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms).


The term “amino,” as used herein, represents —N(RN1)2, wherein each RN1 is, independently, H, OH, NO2, N(RN2)2, SO2ORN2, SO2RN2, SORN2, an N-protecting group, alkyl, alkoxy, aryl, arylalkyl, cycloalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), wherein each of these recited RN1 groups can be optionally substituted; or two RN1 combine to form an alkylene or heteroalkylene, and wherein each RN2 is, independently, H, alkyl, or aryl. The amino groups of the compounds described herein can be an unsubstituted amino (i.e., —NH2) or a substituted amino (i.e., —N(RN1)2).


The term “aryl,” as used herein, refers to an aromatic mono- or polycarbocyclic radical of 6 to 12 carbon atoms having at least one aromatic ring. Examples of such groups include, but are not limited to, phenyl, naphthyl, 1,2,3,4-tetrahydronaphthyl, 1,2-dihydronaphthyl, indanyl, and 1H-indenyl.


The term “arylalkyl,” as used herein, represents an alkyl group substituted with an aryl group. Exemplary unsubstituted arylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-C6 alkyl C6-C10 aryl, C1-C10 alkyl C6-C10 aryl, or C1-C20 alkyl C6-C10 aryl), such as, benzyl and phenethyl. In some embodiments, the alkyl and the aryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups.


The term “azido,” as used herein, represents a —N3 group.


The term “bridged polycycloalkyl,” as used herein, refers to a bridged polycyclic group of 5 to 20 carbons, containing from 1 to 3 bridges.


The term “cyano,” as used herein, represents a —CN group.


The term “carbocyclyl,” as used herein, refers to a non-aromatic C3-C12 monocyclic, bicyclic, or tricyclic structure in which the rings are formed by carbon atoms. Carbocyclyl structures include cycloalkyl groups and unsaturated carbocyclyl radicals.


The term “cycloalkyl,” as used herein, refers to a saturated, non-aromatic, monovalent mono- or polycarbocyclic radical of 3 to 10, preferably 3 to 6 carbon atoms. This term is further exemplified by radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and adamantyl.


The term “halogen,” as used herein, means a fluorine (fluoro), chlorine (chloro), bromine (bromo), or iodine (iodo) radical.


The term “heteroalkyl,” as used herein, refers to an alkyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkyl groups. Examples of heteroalkyl groups are an “alkoxy” which, as used herein, refers alkyl-O— (e.g., methoxy and ethoxy). A heteroalkylene is a divalent heteroalkyl group. The term “heteroalkenyl,” as used herein, refers to an alkenyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkenyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkenyl groups. Examples of heteroalkenyl groups are an “alkenoxy” which, as used herein, refers alkenyl-O—. A heteroalkenylene is a divalent heteroalkenyl group. The term “heteroalkynyl,” as used herein, refers to an alkynyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkynyl group can be further substituted with 1, 2, 3, or 4 substituent groups as described herein for alkynyl groups. Examples of heteroalkynyl groups are an “alkynoxy” which, as used herein, refers alkynyl-O—. A heteroalkynylene is a divalent heteroalkynyl group.


The term “heteroaryl,” as used herein, refers to an aromatic mono- or polycyclic radical of 5 to 12 atoms having at least one aromatic ring containing 1, 2, or 3 ring atoms selected from nitrogen, oxygen, and sulfur, with the remaining ring atoms being carbon. One or two ring carbon atoms of the heteroaryl group may be replaced with a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazoyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, oxaxolyl, and thiazolyl.


The term “heteroarylalkyl,” as used herein, represents an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroarylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-C6 alkyl C2-C9 heteroaryl, C1-C10 alkyl C2-C9 heteroaryl, or C1-C20 alkyl C2-C9 heteroaryl). In some embodiments, the alkyl and the heteroaryl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups.


The term “heterocyclyl,” as used herein, refers a mono- or polycyclic radical having 3 to 12 atoms having at least one ring containing 1, 2, 3, or 4 ring atoms selected from N, O, or S, wherein no ring is aromatic. Examples of heterocyclyl groups include, but are not limited to, morpholinyl, thiomorpholinyl, furyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl, and 1,3-dioxanyl.


The term “heterocyclylalkyl,” as used herein, represents an alkyl group substituted with a heterocyclyl group. Exemplary unsubstituted heterocyclylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-C6 alkyl C2-C9 heterocyclyl, C1-C10 alkyl C2-C9 heterocyclyl, or C1-C20 alkyl C2-C9 heterocyclyl). In some embodiments, the alkyl and the heterocyclyl each can be further substituted with 1, 2, 3, or 4 substituent groups as defined herein for the respective groups.


The term “hydroxyalkyl,” as used herein, represents alkyl group substituted with an —OH group.


The term “hydroxyl,” as used herein, represents an —OH group.


The term “N-protecting group,” as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, “Protective Groups in Organic Synthesis,” 3rd Edition (John Wiley & Sons, New York, 1999). N-protecting groups include, but are not limited to, acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, α-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L, or D, L-amino acids such as alanine, leucine, and phenylalanine; sulfonyl-containing groups such as benzenesulfonyl, and p-toluenesulfonyl; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2,4-20 dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, α,α-dimethyl-3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl, diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl, arylalkyl groups such as benzyl, triphenylmethyl, and benzyloxymethyl, and silyl groups, such as trimethylsilyl. Preferred N-protecting groups are alloc, formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).


The term “nitro,” as used herein, represents an —NO2 group.


The term “thiol,” as used herein, represents an —SH group.


The alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, there will generally be 1 to 4 substituents present, unless otherwise specified. Substituents include, for example: alkyl (e.g., unsubstituted and substituted, where the substituents include any group described herein, e.g., aryl, halo, hydroxy), aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (e.g., substituted and unsubstituted cycloalkyl), halogen (e.g., fluoro), hydroxyl, heteroalkyl (e.g., substituted and unsubstituted methoxy, ethoxy, or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g., NH2 or mono- or dialkyl amino), azido, cyano, nitro, or thiol. Aryl, carbocyclyl (e.g., cycloalkyl), heteroaryl, and heterocyclyl groups may also be substituted with alkyl (unsubstituted and substituted such as arylalkyl (e.g., substituted and unsubstituted benzyl)).


Compounds described herein can have one or more asymmetric carbon atoms and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates, or mixtures of diastereoisomeric racemates. The optically active forms can be obtained for example by resolution of the racemates, by asymmetric synthesis or asymmetric chromatography (chromatography with a chiral adsorbent or eluant). That is, certain of the disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. “Enantiomer” means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that are not related as mirror images, most commonly because they contain two or more asymmetrically substituted carbon atoms and represent the configuration of substituents around one or more chiral carbon atoms. Enantiomers of a compound can be prepared, for example, by separating an enantiomer from a racemate using one or more well-known techniques and methods, such as, for example, chiral chromatography and separation methods based thereon. The appropriate technique and/or method for separating an enantiomer of a compound described herein from a racemic mixture can be readily determined by those of skill in the art. “Racemate” or “racemic mixture” means a compound containing two enantiomers, wherein such mixtures exhibit no optical activity; i.e., they do not rotate the plane of polarized light. “Geometric isomer” means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon-carbon double bond may be in an E (substituents are on opposite sides of the carbon-carbon double bond) or Z (substituents are oriented on the same side) configuration. “R,” “S,” “S*,” “R*,” “E,” “Z,” “cis,” and “trans,” indicate configurations relative to the core molecule. Certain of the disclosed compounds may exist in atropisomeric forms. Atropisomers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. The compounds described herein may be prepared as individual isomers by either isomer-specific synthesis or resolved from an isomeric mixture. Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by weight relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by weight optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by weight pure. Percent optical purity is the ratio of the weight of the enantiomer or over the weight of the enantiomer plus the weight of its optical isomer. Diastereomeric purity by weight is the ratio of the weight of one diastereomer or over the weight of all the diastereomers.


When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by mole fraction pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by mole fraction pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% by mole fraction pure. Percent purity by mole fraction is the ratio of the moles of the enantiomer or over the moles of the enantiomer plus the moles of its optical isomer. Similarly, percent purity by moles fraction is the ratio of the moles of the diastereomer or over the moles of the diastereomer plus the moles of its isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has at least one chiral center, it is to be understood that the name or structure encompasses either enantiomer of the compound free from the corresponding optical isomer, a racemic mixture of the compound, or mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomer(s), or mixtures of diastereomers in which one or more diastereomer is enriched relative to the other diastereomers. The invention embraces all of these forms.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.


Definitions

In this application, unless otherwise clear from context, (i) the term “a” may be understood to mean “at least one”; (ii) the term “or” may be understood to mean “and/or”; and (iii) the terms “including” and “including” may be understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps.


As used herein, the terms “about” and “approximately” refer to a value that is within 10% above or below the value being described. For example, the term “about 5 nM” indicates a range of from 4.5 to 5.5 nM.


As used herein, the term “administration” refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intratumoral, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreal.


As used herein, the term “soft tissue sarcoma” refers to a sarcoma that develops in the soft tissues of the body (e.g., an adult soft tissue sarcoma). Adult soft tissue sarcoma refers to a sarcoma that develops typically in adolescent and adult subjects (e.g., subjects who are at least 10 years old, 11 years old, 12 years old, 13 years old, 14 years old, 15 years old, 16 years old, 17 years old, 18 years old, or 19 years old). Non-limiting examples of soft tissue sarcoma include, but are not limited to, synovial sarcoma, fibrosarcoma, malignant fibrous histiocytoma, dermatofibrosarcoma, liposarcoma, leiomyosarcoma, hemangiosarcoma, Kaposi's sarcoma, lymphangiosarcoma, malignant peripheral nerve sheath tumor/neurofibrosarcoma, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, extraskeletal myxoid chondrosarcoma, and extraskeletal mesenchymal.


As used herein, the term “BAF complex” refers to the BRG1- or FIRBM-associated factors complex in a human cell.


As used herein, the terms “GBAF complex” and “GBAF” refer to a SWI/SNF ATPase chromatin remodeling complex in a human cell. GBAF complex subunits may include, but are not limited to, ACTB, ACTL6A, ACTL6B, BICRA, BICRAL, BRD9, SMARCA2, SMARCA4, SMARCC1, SMARCD1, SMARCD2, SMARCD3, and SS18.


The term “cancer” refers to a condition caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas.


As used herein, a “combination therapy” or “administered in combination” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In some embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.


As used herein, the term “BICRA” refers to BRD4 interacting chromatin remodeling complex associated protein (also called glioma tumor suppressor candidate region gene 1 protein or GLTSCR1), a component of the BAF (BRG1- or BRM-associated factors) complex, a SWI/SNF ATPase chromatin remodeling complex. BICRA is encoded by the BICRA gene. The nucleic acid sequence of an exemplary human BICRA is shown under NCBI Reference Sequence: NM_015711.3 or in SEQ ID NO: 1. The amino acid sequence of an exemplary protein encoded by human BICRA is shown under UniProt Accession No. Q9NZM4 or in SEQ ID NO: 2. The term “BICRA” also refers to natural variants of the wild-type BICRA protein, such as proteins having at least 85% identity (e.g., 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9% identity, or more) to the amino acid sequence of wild-type BICRA, an example of which is set forth in SEQ ID NO: 2.


As used herein, the term “degrader” refers to a small molecule compound including a degradation moiety, wherein the compound interacts with a protein (e.g., BICRA) in a way which results in degradation of the protein, e.g., binding of the compound results in at least 5% reduction of the level of the protein, e.g., in a cell or subject.


As used herein, the term “degradation moiety” refers to a moiety whose binding results in degradation of a protein, e.g., BICRA. In one example, the moiety binds to a protease or a ubiquitin ligase that metabolizes the protein, e.g., BICRA.


By “determining the level of a protein” is meant the detection of a protein, or an mRNA encoding the protein, by methods known in the art either directly or indirectly. “Directly determining” means performing a process (e.g., performing an assay or test on a sample or “analyzing a sample” as that term is defined herein) to obtain the physical entity or value. “Indirectly determining” refers to receiving the physical entity or value from another party or source (e.g., a third-party laboratory that directly acquired the physical entity or value). Methods to measure protein level generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, liquid chromatography (LC)-mass spectrometry, microcytometry, microscopy, fluorescence activated cell sorting (FACS), and flow cytometry, as well as assays based on a property of a protein including, but not limited to, enzymatic activity or interaction with other protein partners. Methods to measure mRNA levels are known in the art.


By “modulating the activity of a BAF complex,” is meant altering the level of an activity related to a BAF complex (e.g., GBAF), or a related downstream effect. The activity level of a BAF complex may be measured using any method known in the art, e.g., the methods described in Kadoch et al, Cell 153:71-85 (2013), the methods of which are herein incorporated by reference.


By “reducing the activity of BICRA,” is meant decreasing the level of an activity related to BICRA, or a related downstream effect. A non-limiting example of inhibition of an activity of BICRA is decreasing the level of a BAF complex (e.g., GBAF) in a cell. The activity level of BICRA may be measured using any method known in the art. In some embodiments, an agent which reduces the activity of BICRA is a small molecule BICRA inhibitor. In some embodiments, an agent which reduces the activity of BICRA is a small molecule BICRA degrader.


By “reducing the level of BICRA,” is meant decreasing the level of BICRA in a cell or subject. The level of BICRA may be measured using any method known in the art.


By “level” is meant a level of a protein, or mRNA encoding the protein, as compared to a reference. The reference can be any useful reference, as defined herein. By a “decreased level” or an “increased level” of a protein is meant a decrease or increase in protein level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 150%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01-fold, about 0.02-fold, about 0.1-fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about 1.2-fold, about 1.4-fold, about 1.5-fold, about 1.8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more). A level of a protein may be expressed in mass/vol (e.g., g/dL, mg/mL, μg/mL, ng/mL) or percentage relative to total protein or mRNA in a sample.


As used herein, the term “inhibitor” refers to any agent which reduces the level and/or activity of a protein (e.g., BICRA). Non-limiting examples of inhibitors include small molecule inhibitors, degraders, antibodies, enzymes, or polynucleotides (e.g., siRNA).


As used herein, the terms “effective amount,” “therapeutically effective amount,” and “a “sufficient amount” of an agent that reduces the level and/or activity of BICRA (e.g., in a cell or a subject) described herein refer to a quantity sufficient to, when administered to the subject, including a human, effect beneficial or desired results, including clinical results, and, as such, an “effective amount” or synonym thereto depends on the context in which it is being applied. For example, in the context of treating cancer, it is an amount of the agent that reduces the level and/or activity of BICRA sufficient to achieve a treatment response as compared to the response obtained without administration of the agent that reduces the level and/or activity of BICRA. The amount of a given agent that reduces the level and/or activity of BICRA described herein that will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, and/or weight) or host being treated, and the like, but can nevertheless be routinely determined by one of skill in the art. Also, as used herein, a “therapeutically effective amount” of an agent that reduces the level and/or activity of BICRA of the present disclosure is an amount which results in a beneficial or desired result in a subject as compared to a control. As defined herein, a therapeutically effective amount of an agent that reduces the level and/or activity of BICRA of the present disclosure may be readily determined by one of ordinary skill by routine methods known in the art. Dosage regimen may be adjusted to provide the optimum therapeutic response.


The term “inhibitory RNA agent” refers to an RNA, or analog thereof, having sufficient sequence complementarity to a target RNA to direct RNA interference. Examples also include a DNA that can be used to make the RNA. RNA interference (RNAi) refers to a sequence-specific or selective process by which a target molecule (e.g., a target gene, protein, or RNA) is down-regulated. Generally, an interfering RNA (“iRNA”) is a double-stranded short-interfering RNA (siRNA), short hairpin RNA (shRNA), or single-stranded micro-RNA (miRNA) that results in catalytic degradation of specific mRNAs, and also can be used to lower or inhibit gene expression.


The terms “short interfering RNA” and “siRNA” (also known as “small interfering RNAs”) refer to an RNA agent, preferably a double-stranded agent, of about 10-50 nucleotides in length, the strands optionally having overhanging ends comprising, for example 1, 2 or 3 overhanging nucleotides (or nucleotide analogs), which is capable of directing or mediating RNA interference. Naturally-occurring siRNAs are generated from longer dsRNA molecules (e.g., >25 nucleotides in length) by a cell's RNAi machinery (e.g., Dicer or a homolog thereof).


The term “shRNA”, as used herein, refers to an RNA agent having a stem-loop structure, comprising a first and second region of complementary sequence, the degree of complementarity and orientation of the regions being sufficient such that base pairing occurs between the regions, the first and second regions being joined by a loop region, the loop resulting from a lack of base pairing between nucleotides (or nucleotide analogs) within the loop region.


The terms “miRNA” and “microRNA” refer to an RNA agent, preferably a single-stranded agent, of about 10-50 nucleotides in length, preferably between about 15-25 nucleotides in length, which is capable of directing or mediating RNA interference. Naturally-occurring miRNAs are generated from stem-loop precursor RNAs (i.e., pre-miRNAs) by Dicer. The term “Dicer” as used herein, includes Dicer as well as any Dicer ortholog or homolog capable of processing dsRNA structures into siRNAs, miRNAs, siRNA-like or miRNA-like molecules. The term microRNA (“miRNA”) is used interchangeably with the term “small temporal RNA” (“stRNA”) based on the fact that naturally-occurring miRNAs have been found to be expressed in a temporal fashion (e.g., during development).


The term “antisense,” as used herein, refers to a nucleic acid comprising a polynucleotide that is sufficiently complementary to all or a portion of a gene, primary transcript, or processed mRNA, so as to interfere with expression of the endogenous gene (e.g., BICRA). “Complementary” polynucleotides are those that are capable of base pairing according to the standard Watson-Crick complementarity rules. Specifically, purines will base pair with pyrimidines to form a combination of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. It is understood that two polynucleotides may hybridize to each other even if they are not completely complementary to each other, provided that each has at least one region that is substantially complementary to the other.


The term “antisense nucleic acid” includes single-stranded RNA as well as double-stranded DNA expression cassettes that can be transcribed to produce an antisense RNA. “Active” antisense nucleic acids are antisense RNA molecules that are capable of selectively hybridizing with a primary transcript or mRNA encoding a polypeptide having at least 80% sequence identity (e.g., 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9% identity, or more) with the targeted polypeptide sequence (e.g., a BICRA polypeptide sequence). The antisense nucleic acid can be complementary to an entire coding strand, or to only a portion thereof. In some embodiments, an antisense nucleic acid molecule is antisense to a “coding region” of the coding strand of a nucleotide sequence. The term “coding region” refers to the region of the nucleotide sequence comprising codons that are translated into amino acid residues. In some embodiments, the antisense nucleic acid molecule is antisense to a “noncoding region” of the coding strand of a nucleotide sequence. The term “noncoding region” refers to 5′ and 3′ sequences that flank the coding region that are not translated into amino acids (i.e., also referred to as 5′ and 3′ untranslated regions). The antisense nucleic acid molecule can be complementary to the entire coding region of mRNA, or can be antisense to only a portion of the coding or noncoding region of an mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nucleotides in length.


“Percent (%) sequence identity” with respect to a reference polynucleotide or polypeptide sequence is defined as the percentage of nucleic acids or amino acids in a candidate sequence that are identical to the nucleic acids or amino acids in the reference polynucleotide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid or amino acid sequence identity can be achieved in various ways that are within the capabilities of one of skill in the art, for example, using publicly available computer software such as BLAST, BLAST-2, or Megalign software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.


For example, percent sequence identity values may be generated using the sequence comparison computer program BLAST. As an illustration, the percent sequence identity of a given nucleic acid or amino acid sequence, A, to, with, or against a given nucleic acid or amino acid sequence, B, (which can alternatively be phrased as a given nucleic acid or amino acid sequence, A that has a certain percent sequence identity to, with, or against a given nucleic acid or amino acid sequence, B) is calculated as follows:





100 multiplied by (the fraction X/Y)


where X is the number of nucleotides or amino acids scored as identical matches by a sequence alignment program (e.g., BLAST) in that program's alignment of A and B, and where Y is the total number of nucleic acids in B. It will be appreciated that where the length of nucleic acid or amino acid sequence A is not equal to the length of nucleic acid or amino acid sequence B, the percent sequence identity of A to B will not equal the percent sequence identity of B to A.


The term “pharmaceutical composition,” as used herein, represents a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient, and manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. Pharmaceutical compositions can be formulated, for example, for oral administration in unit dosage form (e.g., a tablet, capsule, caplet, gelcap, or syrup); for topical administration (e.g., as a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.


A “pharmaceutically acceptable excipient,” as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.


As used herein, the term “pharmaceutically acceptable salt” means any pharmaceutically acceptable salt of the compound of any of the compounds described herein. For example, pharmaceutically acceptable salts of any of the compounds described herein include those that are within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Berge et al., J. Pharmaceutical Sciences 66:1-19, 1977 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P. H. Stahl and C. G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the compounds described herein or separately by reacting a free base group with a suitable organic acid.


The compounds described herein may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the compounds described herein, be prepared from inorganic or organic bases. Frequently, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparation of the appropriate salts are well-known in the art. Salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, and valerate salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.


By a “reference” is meant any useful reference used to compare protein or mRNA levels. The reference can be any sample, standard, standard curve, or level that is used for comparison purposes. The reference can be a normal reference sample or a reference standard or level. A “reference sample” can be, for example, a control, e.g., a predetermined negative control value such as a “normal control” or a prior sample taken from the same subject; a sample from a normal healthy subject, such as a normal cell or normal tissue; a sample (e.g., a cell or tissue) from a subject not having a disease; a sample from a subject that is diagnosed with a disease, but not yet treated with a compound described herein; a sample from a subject that has been treated by a compound described herein; or a sample of a purified protein (e.g., any described herein) at a known normal concentration. By “reference standard or level” is meant a value or number derived from a reference sample. A “normal control value” is a pre-determined value indicative of non-disease state, e.g., a value expected in a healthy control subject. Typically, a normal control value is expressed as a range (“between X and Y”), a high threshold (“no higher than X”), or a low threshold (“no lower than X”). A subject having a measured value within the normal control value for a particular biomarker is typically referred to as “within normal limits” for that biomarker. A normal reference standard or level can be a value or number derived from a normal subject not having a disease or disorder (e.g., cancer); a subject that has been treated with a compound described herein. In preferred embodiments, the reference sample, standard, or level is matched to the sample subject sample by at least one of the following criteria: age, weight, sex, disease stage, and overall health. A standard curve of levels of a purified protein, e.g., any described herein, within the normal reference range can also be used as a reference.


As used herein, the term “subject” refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.


As used herein, the terms “treat,” “treated,” or “treating” mean both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.


As used herein, the terms “variant” and “derivative” are used interchangeably and refer to naturally-occurring, synthetic, and semi-synthetic analogues of a compound, peptide, protein, or other substance described herein. A variant or derivative of a compound, peptide, protein, or other substance described herein may retain or improve upon the biological activity of the original material.


The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a graph illustrating the effect of sgRNA targeting of the BICRA BAF complex subunit on synovial sarcoma cell growth. FIG. 1 corresponds to data obtained with SYO1 cell line. The Y-axis indicated the dropout ratio. The X-axis indicates the nucleotide position of the BICRA gene. The grey box indicates the range of the negative control sgRNAs in the screen. The SYO1 cell line carries SS18-SSX2 fusion protein. The linear protein sequence is shown with BICRA PFAM domains annotated from the PFAM database.



FIG. 2 is a graph illustrating the effect of sgRNA targeting of the BICRA BAF complex subunit on synovial sarcoma cell growth. FIG. 2 corresponds to data obtained with HS-SY-II cell line. The Y-axis indicated the dropout ratio. The X-axis indicates the nucleotide position of the BICRA gene. The grey box indicates the range of the negative control sgRNAs in the screen. The HS-SY-II cell line carries a SS18-SSX1 fusion protein. The linear protein sequence is shown with BICRA PFAM domains annotated from the PFAM database.





DETAILED DESCRIPTION

The present inventors have found that depletion of BICRA in cancer cells inhibits cell growth and may result in the depletion of the SS18-SSX fusion protein and further inhibits the proliferation of the cancer cells.


Accordingly, the invention features methods and compositions useful for the inhibition of the activity of the SS18-SSX fusion proteins, e.g., for the treatment of cancer such as soft tissue sarcomas, e.g., adult soft tissue sarcomas. The invention further features methods and compositions useful for inhibition of the activity of the BICRA protein, e.g., for the treatment of cancer such as soft tissue sarcomas, e.g., in a subject in need thereof. Exemplary methods are described herein.


BICRA-Reducing Agents

Agents described herein that reduce the level and/or activity of BICRA in a cell may be an antibody, a protein (such as an enzyme), a polynucleotide, or a small molecule compound. The agents reduce the level of an activity related to BICRA, or a related downstream effect, or reduce the level of BICRA in a cell or subject.


In some embodiments, the agent that reduces the level and/or activity of BICRA in a cell is an enzyme, a polynucleotide, or a small molecule compound such as a degrader or small molecule BICRA inhibitor.


Antibodies


The agent that reduces the level and/or activity of BICRA can be an antibody or antigen binding fragment thereof. For example, an agent that reduces the level and/or activity of BICRA described herein is an antibody that reduces or blocks the activity and/or function of BICRA through binding to BICRA. The making and use of therapeutic antibodies against a target antigen (e.g., BICRA) is known in the art. See, for example, the references cited herein above, as well as Zhiqiang An (Editor), Therapeutic Monoclonal Antibodies: From Bench to Clinic. 1st Edition. Wiley 2009, and also Greenfield (Ed.), Antibodies: A Laboratory Manual. (Second edition) Cold Spring Harbor Laboratory Press 2013, for methods of making recombinant antibodies, including antibody engineering, use of degenerate oligonucleotides, 5′-RACE, phage display, and mutagenesis; antibody testing and characterization; antibody pharmacokinetics and pharmacodynamics; antibody purification and storage; and screening and labeling techniques.


Polynucleotides


In some embodiments, the agent that reduces the level and/or activity of BICRA is a polynucleotide. In some embodiments, the polynucleotide is an inhibitory RNA molecule, e.g., that acts by way of the RNA interference (RNAi) pathway. An inhibitory RNA molecule can decrease the expression level (e.g., protein level or mRNA level) of BICRA. For example, an inhibitory RNA molecule includes a short interfering RNA (siRNA), short hairpin RNA (shRNA), and/or a microRNA (miRNA) that targets full-length BICRA. A siRNA is a double-stranded RNA molecule that typically has a length of about 19-25 base pairs. A shRNA is a RNA molecule including a hairpin turn that decreases expression of target genes via RNAi. A microRNA is a non-coding RNA molecule that typically has a length of about 22 nucleotides. miRNAs bind to target sites on mRNA molecules and silence the mRNA, e.g., by causing cleavage of the mRNA, destabilization of the mRNA, or inhibition of translation of the mRNA. Degradation is caused by an enzymatic, RNA-induced silencing complex (RISC).


In some embodiments, the agent that reduces the level and/or activity of BICRA is an antisense nucleic acid. Antisense nucleic acids include antisense RNA (asRNA) and antisense DNA (asDNA) molecules, typically about 10 to 30 nucleotides in length, which recognize polynucleotide target sequences or sequence portions through hydrogen bonding interactions with the nucleotide bases of the target sequence (e.g., BICRA). The target sequences may be single- or double-stranded RNA, or single- or double-stranded DNA.


In embodiments, the polynucleotide decreases the level and/or activity of a negative regulator of function or a positive regulator of function. In other embodiments, the polynucleotide decreases the level and/or activity of an inhibitor of a positive regulator of function.


A polynucleotide of the invention can be modified, e.g., to contain modified nucleotides, e.g., 2′-fluoro, 2′-o-methyl, 2′-deoxy, unlocked nucleic acid, 2′-hydroxy, phosphorothioate, 2′-thiouridine, 4′-thiouridine, 2′-deoxyuridine. Without being bound by theory, it is believed that certain modification can increase nuclease resistance and/or serum stability, or decrease immunogenicity. The polynucleotides mentioned above, may also be provided in a specialized form such as liposomes, microspheres, or may be applied to gene therapy, or may be provided in combination with attached moieties. Such attached moieties include polycations such as polylysine that act as charge neutralizers of the phosphate backbone, or hydrophobic moieties such as lipids (e.g., phospholipids, cholesterols, etc.) that enhance the interaction with cell membranes or increase uptake of the nucleic acid. These moieties may be attached to the nucleic acid at the 3′ or 5′ ends and may also be attached through a base, sugar, or intramolecular nucleoside linkage. Other moieties may be capping groups specifically placed at the 3′ or 5′ ends of the nucleic acid to prevent degradation by nucleases such as exonuclease, RNase, etc. Such capping groups include hydroxyl protecting groups known in the art, including glycols such as polyethylene glycol and tetraethylene glycol. The inhibitory action of the polynucleotide can be examined using a cell-line or animal based gene expression system of the present invention in vivo and in vitro. In some embodiments, the polynucleotide decreases the level and/or activity or function of BICRA. In embodiments, the polynucleotide inhibits expression of BICRA. In other embodiments, the polynucleotide increases degradation of BICRA and/or decreases the stability (i.e., half-life) of BICRA. The polynucleotide can be chemically synthesized or transcribed in vitro.


Inhibitory polynucleotides can be designed by methods well known in the art. siRNA, miRNA, shRNA, and asRNA molecules with homology sufficient to provide sequence specificity required to uniquely degrade any RNA can be designed using programs known in the art, including, but not limited to, those maintained on websites for Thermo Fisher Scientific, the German Cancer Research Center, and The Ohio State University Wexner Medical Center. Systematic testing of several designed species for optimization of the inhibitory polynucleotide sequence can be routinely performed by those skilled in the art. Considerations when designing interfering polynucleotides include, but are not limited to, biophysical, thermodynamic, and structural considerations, base preferences at specific positions in the sense strand, and homology. The making and use of inhibitory therapeutic agents based on non-coding RNA such as ribozymes, RNAse P, siRNAs, and miRNAs are also known in the art, for example, as described in Sioud, RNA Therapeutics: Function, Design, and Delivery (Methods in Molecular Biology). Humana Press 2010. Exemplary inhibitory polynucleotides, for use in the methods of the invention, are provided in Table 1, below. In some embodiments, the inhibitory polynucleotides have a nucleic acid sequence with at least 50% (e.g., at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%) sequence identity to the nucleic acid sequence of an inhibitory polynucleotide in Table 1. In some embodiments, the inhibitory polynucleotides have a nucleic acid sequence with at least 70% sequence identity (e.g., 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9% identity, or more) to the nucleic acid sequence of an inhibitory polynucleotide in Table 1.


Construction of vectors for expression of polynucleotides for use in the invention may be accomplished using conventional techniques which do not require detailed explanation to one of ordinary skill in the art. For generation of efficient expression vectors, it is necessary to have regulatory sequences that control the expression of the polynucleotide. These regulatory sequences include promoter and enhancer sequences and are influenced by specific cellular factors that interact with these sequences, and are well known in the art.


Gene Editing


In some embodiments, the agent that reduces the level and/or activity of BICRA is a component of a gene editing system. For example, the agent that reduces the level and/or activity of BICRA introduces an alteration (e.g., insertion, deletion (e.g., knockout), translocation, inversion, single point mutation, or other mutation) in BICRA. In some embodiments, the agent that reduces the level and/or activity of BICRA is a nuclease. Exemplary gene editing systems include the zinc finger nucleases (ZFNs), Transcription Activator-Like Effector-based Nucleases (TALENs), and the clustered regulatory interspaced short palindromic repeat (CRISPR) system. ZFNs, TALENs, and CRISPR-based methods are described, e.g., in Gaj et al., Trends Biotechnol. 31 (7):397-405 (2013).


CRISPR refers to a set of (or system including a set of) clustered regularly interspaced short palindromic repeats. A CRISPR system refers to a system derived from CRISPR and Cas (a CRISPR-associated protein) or other nuclease that can be used to silence or mutate a gene described herein. The CRISPR system is a naturally occurring system found in bacterial and archeal genomes. The CRISPR locus is made up of alternating repeat and spacer sequences. In naturally-occurring CRISPR systems, the spacers are typically sequences that are foreign to the bacterium (e.g., plasmid or phage sequences). The CRISPR system has been modified for use in gene editing (e.g., changing, silencing, and/or enhancing certain genes) in eukaryotes. See, e.g., Wiedenheft et al., Nature 482(7385):331-338 (2012). For example, such modification of the system includes introducing into a eukaryotic cell a plasmid containing a specifically-designed CRISPR and one or more appropriate Cas proteins. The CRISPR locus is transcribed into RNA and processed by Cas proteins into small RNAs that include a repeat sequence flanked by a spacer. The RNAs serve as guides to direct Cas proteins to silence specific DNA/RNA sequences, depending on the spacer sequence. See, e.g., Horvath et al., Science 327(5962):167-170 (2010); Makarova et al., Biology Direct 1:7 (2006); Pennisi, Science 341 (6148):833-836 (2013). In some examples, the CRISPR system includes the Cas9 protein, a nuclease that cuts on both strands of the DNA. See, e.g., Id.


In some embodiments, in a CRISPR system for use described herein, e.g., in accordance with one or more methods described herein, the spacers of the CRISPR are derived from a target gene sequence, e.g., from a BICRA sequence.


In some embodiments, the agent that reduces the level and/or activity of BICRA includes a guide RNA (gRNA) for use in a CRISPR system for gene editing. Exemplary gRNAs, for use in the methods of the invention, are provided in Table 1, below. In embodiments, the agent that reduces the level and/or activity of BICRA includes a ZFN, or an mRNA encoding a ZFN, that targets (e.g., cleaves) a nucleic acid sequence (e.g., DNA sequence) of BICRA. In embodiments, the agent that reduces the level and/or activity of BICRA includes a TALEN, or an mRNA encoding a TALEN, that targets (e.g., cleaves) a nucleic acid sequence (e.g., DNA sequence) of BICRA.


For example, the gRNA can be used in a CRISPR system to engineer an alteration in a gene (e.g., BICRA). In other examples, the ZFN and/or TALEN can be used to engineer an alteration in a gene (e.g., BICRA). Exemplary alterations include insertions, deletions (e.g., knockouts), translocations, inversions, single point mutations, or other mutations. The alteration can be introduced in the gene in a cell, e.g., in vitro, ex vivo, or in vivo. In some embodiments, the alteration decreases the level and/or activity of (e.g., knocks down or knocks out) BICRA, e.g., the alteration is a negative regulator of function. In yet another example, the alteration corrects a defect (e.g., a mutation causing a defect), in BICRA. In certain embodiments, the CRISPR system is used to edit (e.g., to add or delete a base pair) a target gene, e.g., BICRA. In other embodiments, the CRISPR system is used to introduce a premature stop codon, e.g., thereby decreasing the expression of a target gene. In yet other embodiments, the CRISPR system is used to turn off a target gene in a reversible manner, e.g., similarly to RNA interference. In embodiments, the CRISPR system is used to direct Cas to a promoter of a target gene, e.g., BICRA, thereby blocking an RNA polymerase sterically.


In some embodiments, a CRISPR system can be generated to edit BICRA using technology described in, e.g., U.S. Publication No. 20140068797; Cong et al., Science 339(6121):819-823 (2013); Tsai, Nature Biotechnd, 32(6):569-576 (2014); and U.S. Pat. Nos. 8,871,445; 8,865,406; 8,795,965; 8,771,945; and 8,697,359.


In some embodiments, the CRISPR interference (CRISPRi) technique can be used for transcriptional repression of specific genes, e.g., the gene encoding BICRA. In CRISPRi, an engineered Cas9 protein (e.g., nuclease-null dCas9, or dCas9 fusion protein, e.g., dCas9-KRAB or dCas9-SID4X fusion) can pair with a sequence specific guide RNA (sgRNA). The Cas9-gRNA complex can block RNA polymerase, thereby interfering with transcription elongation. The complex can also block transcription initiation by interfering with transcription factor binding. The CRISPRi method is specific with minimal off-target effects and is multiplexable, e.g., can simultaneously repress more than one gene (e.g., using multiple gRNAs). Also, the CRISPRi method permits reversible gene repression.


In some embodiments, CRISPR-mediated gene activation (CRISPRa) can be used for transcriptional activation, e.g., of one or more genes described herein, e.g., a gene that inhibits BICRA. In the CRISPRa technique, dCas9 fusion proteins recruit transcriptional activators. For example, dCas9 can be used to recruit polypeptides (e.g., activation domains) such as VP64 or the p65 activation domain (p65D) and used with sgRNA (e.g., a single sgRNA or multiple sgRNAs), to activate a gene or genes, e.g., endogenous gene(s). Multiple activators can be recruited by using multiple sgRNAs—this can increase activation efficiency. A variety of activation domains and single or multiple activation domains can be used. In addition to engineering dCas9 to recruit activators, sgRNAs can also be engineered to recruit activators. For example, RNA aptamers can be incorporated into a sgRNA to recruit proteins (e.g., activation domains) such as VP64. In some examples, the synergistic activation mediator (SAM) system can be used for transcriptional activation. In SAM, MS2 aptamers are added to the sgRNA. MS2 recruits the MS2 coat protein (MCP) fused to p65AD and heat shock factor 1 (HSF1). The CRISPRi and CRISPRa techniques are described in greater detail, e.g., in Dominguez et al., Nat. Rev. Mol. Cell Bid. 17(1):5-15 (2016), incorporated herein by reference.









TABLE 1







Exemplary Inhibitory Polynucleotides









SEQ
Type of



ID
Inhibitory



NO.
Polynucleotide
Nucleic Acid Sequence












3
CRISPR gRNA
GGAGGGCGCCCTGGTAGACA





4
CRISPR gRNA
ATATCGGCTCCTGCTCCTGG





5
CRISPR gRNA
TCCTGCTCCTGGAGGAGTCC





6
CRISPR gRNA
GGCGCCCTGGTAGACATGGT





7
CRISPR gRNA
TGCAGGGCGTCCTCAAAGGA





8
CRISPR gRNA
GCAGCTGCTGAAACGCACCC





9
CRISPR gRNA
GATCATTACCATCTCCGCTG





10
CRISPR gRNA
CCTGCCCTACCATGTCTACC





11
CRISPR gRNA
GAAGTCTAGGTCCACACTGG





12
CRISPR gRNA
CCTGGTAGACATGGTAGGGC





13
CRISPR gRNA
TCATTACCATCTCCGCTGAG





14
CRISPR gRNA
GGTAGGGCAGGAGGCGATGC





15
CRISPR gRNA
GCAGGGCGTCCTCAAAGGAG





16
CRISPR gRNA
TCAGGGACCAGGTGGAGGGT





17
CRISPR gRNA
TCTGCAGGGAGTCCTGAGTG





18
CRISPR gRNA
CTGCCCTACCATGTCTACCA





19
CRISPR gRNA
GTAGGGCAGGAGGCGATGCA





20
CRISPR gRNA
AGGCCATGCTCAATAAATAT





21
CRISPR gRNA
ACAGCTGGCCAAGGAGAAGC





22
CRISPR gRNA
ATGCAGGGCGTCCTCAAAGG





23
CRISPR gRNA
GCCTCCTCGGACCTTCCAGA





24
CRISPR gRNA
AGAAGTCATTGAGGGCCTGT





25
CRISPR gRNA
TCTCCTCCTGAATGAACATT





26
CRISPR gRNA
TTCTGGAGGATGATTCCGGA





27
CRISPR gRNA
GCAGGAAGGGCTGCACACTC





28
CRISPR gRNA
GGGGCCTGGTGAGGTAGTGA





29
CRISPR gRNA
CCTTGCTGGGCTGAAGCGTG





30
CRISPR gRNA
ATCATTACCATCTCCGCTGA





31
CRISPR gRNA
TTCCTTGCTGGGCTGAAGCG





32
CRISPR gRNA
GACGGCCTTCCCCTCCTTTG





33
CRISPR gRNA
GCTGGTGGCCTCGTCCACTT





34
CRISPR gRNA
AAGTGGACGAGGCCACCAGC





35
CRISPR gRNA
CAGCTGTTTATCCAAGGCAA





36
CRISPR gRNA
GGTAGACATGGTAGGGCAGG





37
CRISPR gRNA
GGAGCATTTGCACAAACACC





38
CRISPR gRNA
TTCAGGAGGTGGACGCTCAT





39
CRISPR gRNA
TCTAGGTCCACACTGGGGGC





40
CRISPR gRNA
GCCCCAGGACGATCTTCTCC





41
CRISPR gRNA
GACACACTCTGTGGCCGGGA





42
CRISPR gRNA
CTTGGCCAGGAGCTGGGAGG





43
CRISPR gRNA
GAGCTGTCCACCTGTGTGGG





44
CRISPR gRNA
CGGAAGAGGCTGCGATGGGG





45
CRISPR gRNA
GCGATGCAGGGCGTCCTCAA





46
CRISPR gRNA
GTTCAGGAGGTGGACGCTCA





47
CRISPR gRNA
TCCCCGCCGCCATGAACGTC





48
CRISPR gRNA
CTTGTTCTGGAGGATGATTC





49
CRISPR gRNA
GGCCTGGTGAGGTAGTGACG





50
CRISPR gRNA
GGGCCTCCCCGGGATTATCC





51
CRISPR gRNA
GTCCCCGTCACTACCTCACC





52
CRISPR gRNA
CTTGTAGTCGGGGTGCAGGA





53
CRISPR gRNA
CTCTGGGTTCAGGTGGTTGC





54
CRISPR gRNA
GCTGCCTGGGAAGAGGGCTT





55
CRISPR gRNA
CCTCCCCGGGATTATCCAGG





56
CRISPR gRNA
TCGAGAAGAGCCTTCGGCTG





57
CRISPR gRNA
GTTGTGACTGGAGGGTGGGT





58
CRISPR gRNA
GATGAGCTGTCCACCTGTGT





59
CRISPR gRNA
GCTGGAGGATGTCACAGGGC





60
CRISPR gRNA
TGCCGGATCACAAGCTTGGT





61
CRISPR gRNA
TCCCCCTCCAACCCTCCACC





62
CRISPR gRNA
AGTGGACGAGGAGTTTGAGA





63
CRISPR gRNA
GTAGTGACGGGGACGAAGAG





64
CRISPR gRNA
GGGTGCAAGGGTGGCTCTGA





65
CRISPR gRNA
GACCCCCTGGAGGACAGTGG





66
CRISPR gRNA
GAGCATTTGCACAAACACCA





67
CRISPR gRNA
GGGCCTGGTGAGGTAGTGAC





68
CRISPR gRNA
GGGCCTTGTTGACCACGTCC





69
CRISPR gRNA
GCCCAAAGTGCCTTCTATGA





70
CRISPR gRNA
CAACATCACGGAGCAGACGC





71
CRISPR gRNA
TGCCGGAAGCTTCTTGCACA





72
CRISPR gRNA
CCGTGATGTTGGCCTCTTGG





73
CRISPR gRNA
GCTCCGTGATGTTGGCCTCT





74
CRISPR gRNA
CAGCGTCTGCTCCGTGATGT





75
CRISPR gRNA
TGACCTCCTGGATAATCCCG





76
CRISPR gRNA
GGCCTTGTTGACCACGTCCT





77
CRISPR gRNA
CTGTGGCCACCACGCTCAAT





78
CRISPR gRNA
TGACGGGCTGGCCCACCACC





79
miRNA
CCACACAGCGGAGGGAGGCGGC





80
miRNA
GGGGAGAGCGAGAGCCCGGCUG





81
miRNA
CCUCCUUUCCGAGGGGCGUCGU





82
miRNA
UUCUUCAGCGGACUCAGUUUGC





83
miRNA
UGCACCGUCUCGACAGGCGCGG





84
miRNA
UUAUCCAACCGAAGGGUGGUCU





85
miRNA
UCUCUCACAGUCUUGUGCACAC





86
miRNA
UGUCGAUGCGCCUCUGCAGGUG





87
miRNA
UGCACACAGUGACACACACAGG





88
miRNA
UGUCAAGCAAGUCGGAUCCAUG





89
miRNA
UGCUCCAGCUUACAGGCUUCCU





90
miRNA
UUGUGCACCGGCUCGCUGAGCC





91
miRNA
UGUGCACAGUGACACACACACA





92
siRNA (guide
GGAGAATTCTGTACATTTA



strand)






93
siRNA (guide
GTATAACGATTTTTTTAAA



strand)






94
siRNA (guide
CTTTGAAATCTGAGCAAAA



strand)






95
siRNA (guide
CTGTAAGATAAATTTTTTT



strand)






96
siRNA (guide
CATTTAGAACTCTTGTAAA



strand)






97
siRNA (guide
GTGATGACCTCCTGGATAA



strand)






98
siRNA (guide
GCATCTTTGTCATCCAAAA



strand)






99
siRNA (guide
CCCAGGCCATGCTCAATAA



strand)






100
siRNA (guide
GGCCATGCTCAATAAATAT



strand)






101
siRNA (guide
CCTCAGCGGAGATGGTAAT



strand)






102
siRNA (guide
CCACCCTTGCCTTGGATAA



strand)






103
siRNA (guide
CACCCCTCGACTTTGAAAT



strand)






104
siRNA (guide
CCGCGCCAGATTTTGAAAT



strand)






105
siRNA (guide
CCTGTTTCCTGGAGCATTT



strand)






106
siRNA (guide
GGCCCAAAGTGCCTTCTAT



strand)






107
siRNA (guide
CCAGGACGTTGACCAGATA



strand)






108
siRNA (guide
CTGTATTTATTGTGTATAA



strand)






109
siRNA (guide
CCGGAATCATCCTCCAGAA



strand)






110
siRNA (guide
CCAGGCCATGCTCAATAAA



strand)






111
siRNA (guide
CAGGCCATGCTCAATAAAT



strand)






112
shRNA (loop
GAGGATGGGAGATGCTTACTATCAAGAGTAGTAAGCATCTCCCATCCTC



bolded)






113
shRNA (loop
GGATGGGAGATGCTTACTAGATCAAGAGTCTAGTAAGCATCTCCCATCC



bolded)






114
shRNA (loop
GATGCTTACTAGACGTGATTTTCAAGAGAAATCACGTCTAGTAAGCATC



bolded)






115
shRNA (loop
GGATCCGAGAAGCTTGACAGTTCAAGAGACTGTCAAGCTTCTCGGATCC



bolded)






116
shRNA (loop
GAAGCTTGACAGTGATGACCTTCAAGAGAGGTCATCACTGTCAAGCTTC



bolded)






117
shRNA (loop
GGCCCAAAGTGCCTTCTATGATCAAGAGTCATAGAAGGCACTTTGGGCC



bolded)






118
shRNA (loop
GCCCAAAGTGCCTTCTATGAATCAAGAGTTCATAGAAGGCACTTTGGGC



bolded)






119
shRNA (loop
GCAAACTGAGTGGCCTGAAGATCAAGAGTCTTCAGGCCACTCAGTTTGC



bolded)






120
shRNA (loop
GGTAATGATCGACCGAATGTTTCAAGAGAACATTCGGTCGATCATTACC



bolded)






121
shRNA (loop
GCCAGGACGTTGACCAGATAATCAAGAGTTATCTGGTCAACGTCCTGGC



bolded)






122
shRNA (loop
GTGCACAAGTGAGTGAGAGATTCAAGAGATCTCTCACTCACTTGTGCAC



bolded)






123
shRNA (loop
GCACAAGTGAGTGAGAGATTTTCAAGAGAAATCTCTCACTCACTTGTGC



bolded)






124
shRNA (loop
GCTAGTCTTCCCTCTGTTCTTTCAAGAGAAGAACAGAGGGAAGACTAGC



bolded)









Small Molecule Compounds


In some embodiments of the invention, the agent that reduces the level and/or activity of BICRA in a cell is a small molecule compound. In some embodiments, the small molecule compound is a structure of Formula I:





A-L-B   Formula I


wherein A is a BICRA binding moiety; L is a linker; and B is a degradation moiety.


In some embodiments, the degradation moiety has the structure of:




embedded image


wherein X1 is CH2, O, S, or NR1, wherein R1 is H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; X2 is C═O, CH2, or




embedded image


R3 and R4 are, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; m is 0, 1, 2, 3, or 4; and each R2 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino,


or a pharmaceutically acceptable salt thereof;




embedded image


wherein each R4, R4′, and R7 is, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R5 is optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; R6 is H, optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; n is 0, 1, 2, 3, or 4; each R8 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; and each R9 and R10 is, independently, H, halogen, optionally substituted C1-C6 alkyl, or optionally substituted C6-C10 aryl, wherein R4′ or R5 comprises a bond to the linker, or a pharmaceutically acceptable salt thereof;




embedded image


wherein each R11, R13, and R15 is, independently, H, optionally substituted C1-C6 alkyl, or optionally substituted C1-C6 heteroalkyl; R12 is optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; R14 is optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; p is 0, 1, 2, 3, or 4; each R16 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; q is 0, 1, 2, 3, or 4; and each R17 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino, or a pharmaceutically acceptable salt thereof; or




embedded image


wherein each R18 and R19 is, independently, H, optionally substituted C1-C6 alkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C6-C10 aryl, optionally substituted C1-C6 alkyl C3-C10 carbocyclyl, or optionally substituted C1-C6 alkyl C6-C10 aryl; r1 is 0, 1, 2, 3, or 4; each R20 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino; r2 is 0, 1, 2, 3, or 4; and each R21 is, independently, halogen, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 heteroalkyl, optionally substituted C3-C10 carbocyclyl, optionally substituted C2-C9 heterocyclyl, optionally substituted C6-C10 aryl, optionally substituted C2-C9 heteroaryl, optionally substituted C2-C6 alkenyl, optionally substituted C2-C6 heteroalkenyl, hydroxy, thiol, or optionally substituted amino, or a pharmaceutically acceptable salt thereof.


In some embodiments, the linker has the structure of Formula II:





A1-(B1)f—(C1)g—(B2)h-(D)-(B3)i—(C2)j—(B4)k-A2   Formula II


wherein A1 is a bond between the linker and A; A2 is a bond between B and the linker; B1, B2, B3, and B4 each, independently, is selected from optionally substituted C1-C2 alkyl, optionally substituted C1-C3 heteroalkyl, O, S, S(O)2, and NRN; RN is hydrogen, optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C2-6 heterocyclyl, optionally substituted C6-12 aryl, or optionally substituted C1-7 heteroalkyl; C1 and C2 are each, independently, selected from carbonyl, thiocarbonyl, sulphonyl, or phosphoryl; f, g, h, l, j, and k are each, independently, 0 or 1; and D is optionally substituted C1-10 alkyl, optionally substituted C2-10 alkenyl, optionally substituted C2-10 alkynyl, optionally substituted C2-6 heterocyclyl, optionally substituted C6-12 aryl, optionally substituted C2-C10 polyethylene glycol, or optionally substituted C1-10 heteroalkyl, or a chemical bond linking A1-(B1)f—(C1)g—(B2)h— to —(B3)i—(C2)j—(B4)k-A2.


Linkers include, but are not limited to, the structure of:




embedded image


Pharmaceutical Uses

The compounds described herein are useful in the methods of the invention and, while not bound by theory, are believed to exert their desirable effects through their ability to modulate the level, status, and/or activity of a BAF complex, e.g., by inhibiting the activity or level of the BRG and BRM proteins in a cell within the BAF complex in a mammal.


An aspect of the present invention relates to methods of treating disorders related to BRG and BRM proteins such as cancer in a subject in need thereof. In some embodiments, the compound is administered in an amount and for a time effective to result in one of (or more, e.g., two or more, three or more, four or more of): (a) reduced tumor size, (b) reduced rate of tumor growth, (c) increased tumor cell death (d) reduced tumor progression, (e) reduced number of metastases, (f) reduced rate of metastasis, (g) decreased tumor recurrence (h) increased survival of subject, and (i) increased progression free survival of a subject.


Treating cancer can result in a reduction in size or volume of a tumor. For example, after treatment, tumor size is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or greater) relative to its size prior to treatment. Size of a tumor may be measured by any reproducible means of measurement. For example, the size of a tumor may be measured as a diameter of the tumor.


Treating cancer may further result in a decrease in number of tumors. For example, after treatment, tumor number is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or greater) relative to number prior to treatment. Number of tumors may be measured by any reproducible means of measurement, e.g., the number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification (e.g., 2×, 3×, 4×, 5×, 10×, or 50×).


Treating cancer can result in a decrease in number of metastatic nodules in other tissues or organs distant from the primary tumor site. For example, after treatment, the number of metastatic nodules is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to number prior to treatment. The number of metastatic nodules may be measured by any reproducible means of measurement. For example, the number of metastatic nodules may be measured by counting metastatic nodules visible to the naked eye or at a specified magnification (e.g., 2×, 10×, or 50×).


Treating cancer can result in an increase in average survival time of a population of subjects treated according to the present invention in comparison to a population of untreated subjects. For example, the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days). An increase in average survival time of a population may be measured by any reproducible means. An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with the compound described herein. An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of a compound described herein.


Treating cancer can also result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population. For example, the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%). A decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of a compound described herein. A decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a pharmaceutically acceptable salt of a compound described herein.


Combination Therapies

A method of the invention can be used alone or in combination with an additional therapeutic agent, e.g., other agents that treat cancer or symptoms associated therewith, or in combination with other types of therapies to treat cancer. In combination treatments, the dosages of one or more of the therapeutic compounds may be reduced from standard dosages when administered alone. For example, doses may be determined empirically from drug combinations and permutations or may be deduced by isobolographic analysis (e.g., Black et al., Neurology 65:S3-S6 (2005)). In this case, dosages of the compounds when combined should provide a therapeutic effect.


In some embodiments, the second therapeutic agent is a chemotherapeutic agent (e.g., a cytotoxic agent or other chemical compound useful in the treatment of cancer). These include alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitors, interferons, platinum coordination complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog. Also included is 5-fluorouracil (5-FU), leucovorin (LV), irenotecan, oxaliplatin, capecitabine, paclitaxel, and doxetaxel. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (see, e.g., Agnew, Chem. Intl. Ed Engl. 33:183-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin, including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® (paclitaxel; Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANE®, cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Ill.), and TAXOTERE® doxetaxel (Rhone-Poulenc Rorer, Antony, France); chloranbucil; GEMZAR® gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g., CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Two or more chemotherapeutic agents can be used in a cocktail to be administered in combination with the first therapeutic agent described herein. Suitable dosing regimens of combination chemotherapies are known in the art and described in, for example, Saltz et al., Proc. Am. Soc. Clin. Oncol. 18:233a (1999), and Douillard et al., Lancet 355(9209):1041-1047 (2000).


In some embodiments, the second therapeutic agent is a therapeutic agent which is a biologic such a cytokine (e.g., interferon or an interleukin (e.g., IL-2)) used in cancer treatment. In some embodiments the biologic is an anti-angiogenic agent, such as an anti-VEGF agent, e.g., bevacizumab (AVASTIN®). In some embodiments the biologic is an immunoglobulin-based biologic, e.g., a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein or a functional fragment thereof) that agonizes a target to stimulate an anti-cancer response, or antagonizes an antigen important for cancer. Such agents include RITUXAN® (rituximab); ZENAPAX® (daclizumab); SIMULECT® (basiliximab); SYNAGIS® (palivizumab); REMICADE® (infliximab); HERCEPTIN® (trastuzumab); MYLOTARG® (gemtuzumab ozogamicin); CAMPATH® (alemtuzumab); ZEVALIN® (ibritumomab tiuxetan); HUMIRA® (adalimumab); XOLAIR® (omalizumab); BEXXAR® (tositumomab-I-131); RAPTIVA® (efalizumab); ERBITUX® (cetuximab); AVASTIN® (bevacizumab); TYSABRI® (natalizumab); ACTEMRA® (tocilizumab); VECTIBIX® (panitumumab); LUCENTIS® (ranibizumab); SOURIS® (eculizumab); CIMZIA® (certolizumab pegol); SIMPONI® (golimumab); ILARIS® (canakinumab); STELARA® (ustekinumab); ARZERRA® (ofatumumab); PROLIA® (denosumab); NUMAX® (motavizumab); ABTHRAX® (raxibacumab); BENLYSTA® (belimumab); YERVOY® (ipilimumab); ADCETRIS® (brentuximab vedotin); PERJETA® (pertuzumab); KADCYLA® (ado-trastuzumab emtansine); and GAZYVA® (obinutuzumab). Also included are antibody-drug conjugates.


The second agent may be a therapeutic agent which is a non-drug treatment. For example, the second therapeutic agent is radiation therapy, cryotherapy, hyperthermia, and/or surgical excision of tumor tissue.


The second agent may be a checkpoint inhibitor. In one embodiment, the inhibitor of checkpoint is an inhibitory antibody (e.g., a monospecific antibody such as a monoclonal antibody). The antibody may be, e.g., humanized or fully human. In some embodiments, the inhibitor of checkpoint is a fusion protein, e.g., an Fc-receptor fusion protein. In some embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with a checkpoint protein. In some embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein. In some embodiments, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA4 antibody or fusion a protein such as ipilimumab/YERVOY® or tremelimumab). In some embodiments, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PD-1 (e.g., nivolumab/OPDIVO®; pembrolizumab/KEYTRUDA®; pidilizumab/CT-011). In some embodiments, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PDL1 (e.g., MPDL3280A/RG7446; MEDI4736; MSB0010718C; BMS 936559). In some embodiments, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or Fc fusion or small molecule inhibitor) of PDL2 (e.g., a PDL2/Ig fusion protein such as AMP 224). In some embodiments, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of B7-H3 (e.g., MGA271), B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or a combination thereof.


In some embodiments, the anti-cancer therapy is a T cell adoptive transfer (ACT) therapy. In some embodiments, the T cell is an activated T cell. The T cell may be modified to express a chimeric antigen receptor (CAR). CAR modified T (CAR-T) cells can be generated by any method known in the art. For example, the CAR-T cells can be generated by introducing a suitable expression vector encoding the CAR to a T cell. Prior to expansion and genetic modification of the T cells, a source of T cells is obtained from a subject. T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. In certain embodiments of the present invention, any number of T cell lines available in the art, may be used. In some embodiments, the T cell is an autologous T cell. Whether prior to or after genetic modification of the T cells to express a desirable protein (e.g., a CAR), the T cells can be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.


In any of the combination embodiments described herein, the first and second therapeutic agents are administered simultaneously or sequentially, in either order. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours up to 24 hours or up to 1-7, 1-14, 1-21 or 1-30 days before or after the second therapeutic agent.


Delivery of Anti-BICRA Agents


A variety of methods are available for the delivery of anti-BICRA agents to a subject including viral and non-viral methods.


Viral Delivery Methods


In some embodiments, the agent that reduces the level and/or activity of BICRA is delivered by a viral vector (e.g., a viral vector expressing an anti-BICRA agent). Viral genomes provide a rich source of vectors that can be used for the efficient delivery of exogenous genes into a mammalian cell. Viral genomes are particularly useful vectors for gene delivery because the polynucleotides contained within such genomes are typically incorporated into the nuclear genome of a mammalian cell by generalized or specialized transduction. These processes occur as part of the natural viral replication cycle, and do not require added proteins or reagents in order to induce gene integration. Examples of viral vectors include a retrovirus (e.g., Retroviridae family viral vector), adenovirus (e.g., Ad5, Ad26, Ad34, Ad35, and Ad48), parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses, such as picornavirus and alphavirus, and double-stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus, replication deficient herpes virus), and poxvirus (e.g., vaccinia, modified vaccinia Ankara (MVA), fowlpox and canarypox). Other viruses include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, human papillomavirus, human foamy virus, and hepatitis virus, for example. Examples of retroviruses include: avian leukosis-sarcoma, avian C-type viruses, mammalian C-type, B-type viruses, D-type viruses, oncoretroviruses, HTLV-BLV group, lentivirus, alpharetrovirus, gammaretrovirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, Virology (Third Edition) Lippincott-Raven, Philadelphia, 1996). Other examples include murine leukemia viruses, murine sarcoma viruses, mouse mammary tumor virus, bovine leukemia virus, feline leukemia virus, feline sarcoma virus, avian leukemia virus, human T cell leukemia virus, baboon endogenous virus, Gibbon ape leukemia virus, Mason Pfizer monkey virus, simian immunodeficiency virus, simian sarcoma virus, Rous sarcoma virus and lentiviruses. Other examples of vectors are described, for example, in U.S. Pat. No. 5,801,030, the teachings of which are incorporated herein by reference.


Exemplary viral vectors include lentiviral vectors, AAVs, and retroviral vectors. Lentiviral vectors and AAVs can integrate into the genome without cell divisions, and both types have been tested in pre-clinical animal studies. Methods for preparation of AAVs are described in the art e.g., in U.S. Pat. Nos. 5,677,158, 6,309,634, and 6,683,058, each of which is incorporated herein by reference. Methods for preparation and in vivo administration of lentiviruses are described in US 20020037281 (incorporated herein by reference). Preferably, a lentiviral vector is a replication-defective lentivirus particle. Such a lentivirus particle can be produced from a lentiviral vector comprising a 5′ lentiviral LTR, a tRNA binding site, a packaging signal, a promoter operably linked to a polynucleotide signal encoding the fusion protein, an origin of second strand DNA synthesis and a 3′ lentiviral LTR.


Retroviruses are most commonly used in human clinical trials, as they carry 7-8 kb, and have the ability to infect cells and have their genetic material stably integrated into the host cell with high efficiency (see, e.g., WO 95/30761; WO 95/24929, each of which is incorporated herein by reference). Preferably, a retroviral vector is replication defective. This prevents further generation of infectious retroviral particles in the target tissue. Thus, the replication defective virus becomes a “captive” transgene stable incorporated into the target cell genome. This is typically accomplished by deleting the gag, env, and pol genes (along with most of the rest of the viral genome). Heterologous nucleic acids are inserted in place of the deleted viral genes. The heterologous genes may be under the control of the endogenous heterologous promoter, another heterologous promoter active in the target cell, or the retroviral 5′ LTR (the viral LTR is active in diverse tissues).


These delivery vectors described herein can be made target-specific by attaching, for example, a sugar, a glycolipid, or a protein (e.g., an antibody to a target cell receptor).


Reversible delivery expression systems may also be used. The Cre-loxP or FLP/FRT system and other similar systems can be used for reversible delivery-expression of one or more of the above-described nucleic acids. See WO2005/112620, WO2005/039643, US20050130919, US20030022375, US20020022018, US20030027335, and US20040216178. In particular, the reversible delivery-expression system described in US20100284990 can be used to provide a selective or emergency shut-off.


Non-Viral Delivery Methods


Several non-viral methods exist for delivery of anti-BICRA agents including polymeric, biodegradable microparticle, or microcapsule delivery devices known in the art. For example, a colloidal dispersion system may be used for targeted delivery an anti-BICRA agent described herein. Colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Liposomes are artificial membrane vesicles that are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles (LUV), which range in size from 0.2-4.0 μm can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules.


The composition of the liposome is usually a combination of phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations.


Lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidyl-ethanolamine, sphingolipids, cerebrosides, and gangliosides. Exemplary phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine, and distearoyl-phosphatidylcholine. The targeting of liposomes is also possible based on, for example, organ-specificity, cell-specificity, and organelle-specificity and is known in the art. In the case of a liposomal targeted delivery system, lipid groups can be incorporated into the lipid bilayer of the liposome in order to maintain the targeting ligand in stable association with the liposomal bilayer. Various linking groups can be used for joining the lipid chains to the targeting ligand. Additional methods are known in the art and are described, for example in U.S. Patent Application Publication No. 20060058255.


Pharmaceutical Compositions

The pharmaceutical compositions described herein are preferably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo.


The compounds described herein may be used in the form of the free base, in the form of salts, solvates, and as prodrugs. All forms are within the methods described herein. In accordance with the methods of the invention, the described compounds or salts, solvates, or prodrugs thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compounds described herein may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, intratumoral, or transdermal administration and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.


A compound described herein may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, a compound described herein may be incorporated with an excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, and wafers. A compound described herein may also be administered parenterally. Solutions of a compound described herein can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO, and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2012, 22nd ed.) and in The United States Pharmacopeia: The National Formulary (USP 41 NF 36), published in 2018. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that may be easily administered via syringe. Compositions for nasal administration may conveniently be formulated as aerosols, drops, gels, and powders. Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device. Alternatively, the sealed container may be a unitary dispensing device, such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use. Where the dosage form includes an aerosol dispenser, it will contain a propellant, which can be a compressed gas, such as compressed air or an organic propellant, such as fluorochlorohydrocarbon. The aerosol dosage forms can also take the form of a pump-atomizer. Compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, where the active ingredient is formulated with a carrier, such as sugar, acacia, tragacanth, gelatin, and glycerine. Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base, such as cocoa butter. A compound described herein may be administered intratumorally, for example, as an intratumoral injection. Intratumoral injection is injection directly into the tumor vasculature and is specifically contemplated for discrete, solid, accessible tumors. Local, regional, or systemic administration also may be appropriate. A compound described herein may advantageously be contacted by administering an injection or multiple injections to the tumor, spaced for example, at approximately, 1 cm intervals. In the case of surgical intervention, the present invention may be used preoperatively, such as to render an inoperable tumor subject to resection. Continuous administration also may be applied where appropriate, for example, by implanting a catheter into a tumor or into tumor vasculature.


The compounds described herein may be administered to an animal, e.g., a human, alone or in combination with pharmaceutically acceptable carriers, as noted herein, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration, and standard pharmaceutical practice.


Dosages

The dosage of the compounds described herein, and/or compositions including a compound described herein, can vary depending on many factors, such as the pharmacodynamic properties of the compound; the mode of administration; the age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of the treatment, and the type of concurrent treatment, if any; and the clearance rate of the compound in the animal to be treated. One of skill in the art can determine the appropriate dosage based on the above factors. The compounds described herein may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. In general, satisfactory results may be obtained when the compounds described herein are administered to a human at a daily dosage of, for example, between 0.05 mg and 3000 mg (measured as the solid form). Dose ranges include, for example, between 10-1000 mg (e.g., 50-800 mg). In some embodiments, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1000 mg of the compound is administered.


Alternatively, the dosage amount can be calculated using the body weight of the patient. For example, the dose of a compound, or pharmaceutical composition thereof, administered to a patient may range from 0.1-50 mg/kg (e.g., 0.25-25 mg/kg). In exemplary, non-limiting embodiments, the dose may range from 0.5-5.0 mg/kg (e.g., 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 mg/kg) or from 5.0-20 mg/kg (e.g., 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 mg/kg).


Kits

The invention also features kits including (a) a pharmaceutical composition including an agent that reduces the level and/or activity of BICRA in a cell or subject described herein, and (b) a package insert with instructions to perform any of the methods described herein. In some embodiments, the kit includes (a) a pharmaceutical composition including an agent that reduces the level and/or activity of BICRA in a cell or subject described herein, (b) an additional therapeutic agent (e.g., an anti-cancer agent), and (c) a package insert with instructions to perform any of the methods described herein.


EXAMPLES
Example 1—High Density Tiling sgRNA Screen Against Human BAF Complex Subunits in Synovial Sarcoma Cell Line SYO1

The following example shows that BICRA sgRNA inhibits cell growth in synovial sarcoma cells.


Procedure: To perform high density sgRNA tiling screen, an sgRNA library against BAF complex subunits was custom synthesized at Cellecta (Mountain View, Calif.). All BICRA-targeting sgRNAs used in this screen are listed in Table 2. Negative and positive control sgRNA were included in the library. Negative controls consisted of 200 sgRNAs that do not target human genome. The positive controls are sgRNAs targeting essential genes (CDC16, GTF2B, HSPA5, HSPA9, PAFAH1B1, PCNA, POLR2L, RPL9, and SF3A3). All positive and negative control sgRNAs are listed in Table 3. Procedures for virus production, cell infection, and performing the sgRNA screen were previously described (Tsherniak et al, Cell 170:564-576 (2017); Munoz et al, Cancer Discovery 6:900-913 (2016)). For each sgRNA, 50 counts were added to the sequencing counts and for each time point the resulting counts were normalized to the total number of counts. The log 2 of the ratio between the counts (defined as dropout ratio) at day 24 and day 1 post-infection was calculated. For negative control sgRNAs, the 2.5 and 97.5 percentile of the log 2 dropout ratio of all non-targeting sgRNAs was calculated and considered as background (grey box in the graph). Protein domains were obtained from PFAM regions defined for the UNIPROT identifier: Q9NZM4.


Results: As shown in FIG. 1, targeted inhibition of the GBAF complex component BICRA by sgRNA resulted in growth inhibition of the SYO1 synovial sarcoma cell line, respectively. sgRNAs against other components of the BAF complex resulted in increased proliferation of cells, inhibition of cell growth, or had no effect on SYO1 cells. These data show that targeting various subunits of the GBAF complex represents a therapeutic strategy for the treatment of synovial sarcoma.


Example 2—High Density Tiling sgRNA Screen Against Human BAF Complex Subunits in Synovial Sarcoma Cell Line HS-SY-II

The following example shows that BICRA sgRNA inhibits cell growth in synovial sarcoma cells.


Procedure: To perform high density sgRNA tiling screen, an sgRNA library against BAF complex subunits was custom synthesized at Cellecta (Mountain View, Calif.). All BICRA-targeting sgRNAs used in this screen are listed in Table 2. Negative and positive control sgRNA were included in the library. Negative controls consisted of 200 sgRNAs that do not target human genome. The positive controls are sgRNAs targeting essential genes (CDC16, GTF2B, HSPA5, HSPA9, PAFAH1B1, PCNA, POLR2L, RPL9, and SF3A3). All positive and negative control sgRNAs are listed in Table 3. Procedures for virus production, cell infection, and performing the sgRNA screen were previously described (Tsherniak et al, Cell 170:564-576 (2017); Munoz et al, Cancer Discovery 6:900-913 (2016)). For each sgRNA, 50 counts were added to the sequencing counts and for each time point the resulting counts were normalized to the total number of counts. The log 2 of the ratio between the counts (defined as dropout ratio) at day 24 and day 1 post-infection was calculated. For negative control sgRNAs, the 2.5 and 97.5 percentile of the log 2 dropout ratio of all non-targeting sgRNAs was calculated and considered as background (grey box in the graph). Protein domains were obtained from PFAM regions defined for the UNIPROT identifier: Q9NZM4.


Results: As shown in FIG. 2, targeted inhibition of the GBAF complex component BICRA by sgRNA resulted in growth inhibition of the HS-SY-II synovial sarcoma cell line. sgRNAs against other components of the BAF complex resulted in increased proliferation of cells, inhibition of cell growth, or had no effect on HS-SY-II cells. These data show that targeting various subunits of the GBAF complex represents a therapeutic strategy for the treatment of synovial sarcoma.









TABLE 2







BICRA sgRNA Library










SEQ ID

SEQ ID



NO
Nucleic Acid Sequence
NO
Nucleic Acid Sequence





125
GGAGGAGGCCACAGCAGAGG
354
GGCGTGTTGAGCGCCATGAC





126
GGTGGAGGATGGAGGGAGGT
355
GTTCAGGAGGTGGACGCTCA





127
CTTGGCCAGGAGCTGGGAGG
356
TCCGAGGGTCATCGGCTTCC





128
GCTGCCTGGGAAGAGGGCTT
357
CGGAAGAGGCTGCGATGGGG





129
ACTGAGCCTGGGCCCCGTGT
358
TGGCCACATTCAGGGTCGGG





130
CTTGGTGGGCAGGTGTGGAG
359
CTGCGTCTGTGCTGGTCAGT





131
CCACCCTCCAGTCACAACGG
360
CTGCTTGGCCAGGAGCTGGG





132
TACCTCACCAGGCCCCTCTG
361
GGTGAGCCCCTCAGCGGAGA





133
TCTGCAGGGAGTCCTGAGTG
362
CACGGGCCAGATGAACGGCA





134
GGTTGGGCTCTGGGTTCAGG
363
AACTCTGTGTTCGGAGGCGC





135
GCCCGGAGAAGATCGTCCTG
364
TGCTGCCTTGGTTCAGGAGG





136
ATCCTCCACCTCCTCTGCTG
365
GGAGGGCGCCCTGGTAGACA





137
AAGGCACTTTGGGCCTCCCC
366
GCCATGAACGTCAGGTTCTG





138
AAGTGGACGAGGCCACCAGC
367
TTGTTGACCACGTCCTGGGG





139
GACCCCCTGGAGGACAGTGG
368
TAAATATCGGCTCCTGCTCC





140
AGGGCCGAGGCCAGCTCCTT
369
CCCGACCCTGAATGTGGCCA





141
AGGGTGCAAGGGTGGCTCTG
370
CCCTCGGAAGAGGCTGCGAT





142
GGAGGCCACAGCAGAGGAGG
371
TACCCACCCTCCAGTCACAA





143
TAGCCCGGAGAAGATCGTCC
372
ACCCCCCGCTACCCTCAAGG





144
AAAGATGCCAGGCAGAGTTG
373
GGGGGCCCTTCAGCATACGC





145
GTGGAGGGTTGGAGGGGGAG
374
GCCCAAAGTGCCTTCTATGA





146
CCTCGGAAGAGGCTGCGATG
375
TCAGGGACCAGGTGGAGGGT





147
CATCCTCCAGAACAAGGCTG
376
GTCAGCTGGTAGAGCTTGGG





148
CCTGGAGGACGTCTCAGAGG
377
CTGCCTGGGAAGAGGGCTTG





149
TGTGCGATGCAGGATCACGT
378
TGCTCCTGGAGGAGTCCCGG





150
TGCCCACCAAGCTTGTGATC
379
CAGGCAGTAGGAACTGGTTC





151
GAAGCTGGCCTGGTCCACGT
380
GATGGCGCTCTGCAGGTGCT





152
GTCCAAGGAGGAGGCGGCAG
381
CTTCCTGGAAGATGACATCC





153
GGACCAGGTGGAGGGTTGGA
382
ACAGCTGGCCAAGGAGAAGC





154
ATCATTACCATCTCCGCTGA
383
CTGACCAGCACAGACGCAGC





155
TGGAGGATGGAGGGAGGTGG
384
GAAGTCTAGGTCCACACTGG





156
TGGAAGGTCCGAGGAGGCGG
385
ATGGGTTTGCTCAGGGCCCC





157
TGGGCAGCTGGAAGGGACCC
386
GATGACCTCCTGGATAATCC





158
GATCATTACCATCTCCGCTG
387
GTGCCTTCTATGAAGGTCCT





159
ACAGCAGAGGAGGTGGAGGA
388
CCCCACGGCCATCCTCACTC





160
TCCAGTTCCCACCCAGCCAG
389
AGCTTGTGATCCGGCACGGC





161
CTGAGTGAGGATGGCCGTGG
390
CCTCCTGGATAATCCCGGGG





162
TGACCTCCTGGATAATCCCG
391
CTACCTCACCAGGCCCCTCT





163
CTCTGCTGGAGGATGTCACA
392
CTCTGGGTTCAGGTGGTTGC





164
GCAAGGGTGGCTCTGAGGGA
393
TGCAAGGGTGGCTCTGAGGG





165
TGCCCCAGGACGATCTTCTC
394
CAAACGGCTTGGGCGTCAGC





166
CTCCTCTGCCGCCTCCTCCT
395
TTGCTGGGCTGAAGCGTGGG





167
GCCTCCTCGGACCTTCCAGA
396
TGGTCAACGTCCTGGCGCCG





168
AGAGGTTCTTCTGGATGACC
397
GGCGTCAGCTGGTAGAGCTT





169
TCTTCTCGAGAGATTTCACC
398
GTGCCGGATCACAAGCTTGG





170
AGCGTCCACCTCCTGAACCA
399
TGCAGGGCGTCCTCAAAGGA





171
TCTGCTTCCAGCCGAGAACA
400
GATCGACCGAATGTTCATTC





172
GGGCCTCCCCGGGATTATCC
401
CCTGGGGCAGGAACATCTGC





173
TGCCGGATCACAAGCTTGGT
402
CAGCGTCTGCTCCGTGATGT





174
AGAGTTCCCATTGAGCGTGG
403
TCCATGCAAGAAGTCATTGA





175
GGTGGCCTCGTCCACTTTGG
404
CAGGAAGTCTAGGTCCACAC





176
TTCCTGGAAGATGACATCCT
405
CCCCAGTGACTACCACAAAG





177
CTGCCCTACCATGTCTACCA
406
TGGCCTCTTGGAGGCTCTGC





178
TCCTGGGCTCTCCTGCGACA
407
TTCCTTGCTGGGCTGAAGCG





179
GCCCCAGGACGATCTTCTCC
408
GGGGGTGGTCACCATCTGGA





180
CTCGAGAAGAGCCTTCGGCT
409
CAGAACCAGTTCCTACTGCC





181
GGTAGTGACGGGGACGAAGA
410
CTGTGGCCACCACGCTCAAT





182
CGGTGAGCGTGTCATCCTGC
411
CAGCAAGGTCGTGCACAACA





183
GAAGAACCTCTCGGCCGCTG
412
GACGTTCATGGCGGCGGGGA





184
CTGCAGGGTGGGCAGCTGGA
413
AGTGGACGAGGAGTTTGAGA





185
TTGGCCAGGAGCTGGGAGGT
414
GGGACCAGGTGGAGGGTTGG





186
AGTGCCTTCTATGAAGGTCC
415
CTGTACCAGCGTATGCTGAA





187
TCCTGCTCCTGGAGGAGTCC
416
TCAGTGGGCCCGCCAGGTTC





188
GCCGTGCCGGATCACAAGCT
417
GGGCGCCAGGCAGTAGGAAC





189
CGGCTGGAAGCAGAGGAGGG
418
GGGGGTGTTGAGCATGGCCA





190
ACCCCCGTCGCCAAAGGAGC
419
GACCTTCATAGAAGGCACTT





191
GTCAGTGGGCAGGCCCCATC
420
TCTCGGCTGGAAGCAGAGGA





192
ACCCTCGGAAGAGGCTGCGA
421
GTCTGTGCTGGTCAGTGGGC





193
GGTGACCCCCTGGAGGACAG
422
GCTGGAAGTCGGATGGCGTA





194
GCAGGGCGTCCTCAAAGGAG
423
GTTGAGCATGGCCACGGCGC





195
CGGAGAGGATGTGCGCGCTG
424
AAGCTTGTGATCCGGCACGG





196
ACTACCTCACCAGGCCCCTC
425
GGTAGGGCAGGAGGCGATGC





197
TCTTGAGCTTGAGCCCGATG
426
GTCGGTGCTGCCTGGGAAGA





198
ATATCGGCTCCTGCTCCTGG
427
CATGGGTTTGCTCAGGGCCC





199
GTTGTGACTGGAGGGTGGGT
428
TGGGAACTGGAGCTGGAAGT





200
AGCGGCCTTGGCCACATTCA
429
GAGGCCAGCTCCTTTGGCGA





201
AGATGCCAGGCAGAGTTGGG
430
GTCACCATCTGGAAGGTCCG





202
GGCCTCCAAGGCCTGCCCAA
431
AATCTCTCGAGAAGAGCCTT





203
GGAAGGGGGTGGTCACCATC
432
AGTGAGGATGGCCGTGGGGG





204
ATGCAGGGCGTCCTCAAAGG
433
AAGATGCCAGGCAGAGTTGG





205
ATCCTCCAGAACAAGGCTGG
434
CCTGCAGATGTTCCTGCCCC





206
TTCTCGGCTGGAAGCAGAGG
435
GCAGGAAGCCGGGCTCAGCA





207
CAGCCCTTCCTGCAGCCTGT
436
CGGGCCTCGTAGGTCTTGAT





208
TGACCGAGGCAGGCACGGAG
437
GTCCTGAGTGAGGATGGCCG





209
CCTCGTAGGTCTTGATGGGC
438
ACCTACCGCGAGAACGTGGG





210
GCTGCAGTGTCACATTGCCC
439
CTCCAGTTCCCACCCAGCCA





211
ACGGGCATCTGGAAGAGCGC
440
AACTGGAGCTGGAAGTCGGA





212
GGGCCTTGTTGACCACGTCC
441
GGGAAGGCCGTCTTGTAGTC





213
AGCTTGGTGGGCAGGTGTGG
442
GGGGCCTGGTGAGGTAGTGA





214
GCTTGACAGTGATGACCTCC
443
ACCTGACGTTCATGGCGGCG





215
GCCTTGTTGACCACGTCCTG
444
GCCGTCGGGGGTGTTGAGCA





216
ACCAGGTGGAGGGTTGGAGG
445
GGTAGTCACTGGGGGAGGGG





217
GTCCTCGTCGGCGTCCAAGG
446
CTCCTCCTTGGACGCCGACG





218
ACCAAGCTTGTGATCCGGCA
447
ACTCTGTGTTCGGAGGCGCG





219
ATGACCTCCTGGATAATCCC
448
CGTAGGGGCTGGCAACCTGG





220
TCATCCTCCAGAACAAGGCT
449
CTTGTAGTCGGGGTGCAGGA





221
GGTGCGTTTCAGCAGCTGCG
450
CGTGCCGTTCATCTGGCCCG





222
TGCTGCTGCCTTGGTTCAGG
451
ATGACCAGGCCAGCCCCCTG





223
GCGATGCAGGGCGTCCTCAA
452
GTCCCCGTCACTACCTCACC





224
GGTCATCCAGAAGAACCTCT
453
AGAAGTCATTGAGGGCCTGT





225
CTTGGCCAGCTGTTTATCCA
454
AGCCCAGGATGTCATCTTCC





226
CGGGGCGCTGACTATGACCG
455
CTGAGAGCTGCTGCGGGAGC





227
CCTCCTCTGAGACGTCCTCC
456
AGCTGGAAGTCGGATGGCGT





228
AAGCCGATGACCCTCGGAAG
457
GTGGCCTCGTCCACTTTGGC





229
TCTAGGTCCACACTGGGGGC
458
GGTCGGTGCTGCCTGGGAAG





230
TCGGTGAGCGTGTCATCCTG
459
GGTTCTGGTTTGTGAGGATG





231
CCCATCGCAGCCTCTTCCGA
460
ACTGGAGGGTGGGTAGGCCT





232
GTGACACTGCAGCCCATCCC
461
GAACCCAGAGCCCAACCAGC





233
CAAGTCCGAGTCGCCCGACG
462
CCTGAGTGAGGATGGCCGTG





234
TCCCCCTCCAACCCTCCACC
463
ATTCAGGGTCGGGAGGTTGC





235
GCTCCGTGATGTTGGCCTCT
464
GCCTACCGTGCTGACCCACC





236
GCTGGTGGCCTCGTCCACTT
465
CAGTATGAGAGCAAACTGAG





237
ACCATCTGGAAGGTCCGAGG
466
AGGCCATGCTCAATAAATAT





238
CCGGGCCTCGTAGGTCTTGA
467
GGGCCTGGTGAGGTAGTGAC





239
GACCTACCGCGAGAACGTGG
468
CAGCATCCTGAACCTGCAGC





240
CCTCCCCGGGATTATCCAGG
469
GCCCCAGGACGTGGTCAACA





241
GGAGAGGATGTGCGCGCTGT
470
ATGAGCTGTCCACCTGTGTG





242
GGGTGCAAGGGTGGCTCTGA
471
TCTTGATGGGCGGGCGGTTG





243
TGAGCTGTCCACCTGTGTGG
472
CAGCCTCTTCCGAGGGTCAT





244
GCAGCTGCTGAAACGCACCC
473
CCATGAACGTCAGGTTCTGC





245
AGCCCGGAGAAGATCGTCCT
474
AAGTCGGATGGCGTAGGGGC





246
TCAGTGGGCAGGCCCCATCT
475
CGATGCTGCTGCCTTGGTTC





247
ACCTTCATAGAAGGCACTTT
476
TGGGCGTGGGTGTGCGATGC





248
CCTCCAAGAGGCCAACATCA
477
GGTAGGTCTTGCGCAGTGGC





249
ACCCAGGTCCAGCTCAGCCT
478
GGATCACAAGCTTGGTGGGC





250
TCCTTGCTGGGCTGAAGCGT
479
CTGGTACAGCTCGTCCTCCA





251
TGTCATCTTCCAGGAAGTCT
480
CCCCACGCTTCAGCCCAGCA





252
TTTGTCATCCAAAACCAGCT
481
ACCTTGAGGGTAGCGGGGGG





253
AGGCCCACAGGCTGCAGGAA
482
ACAAAGATGCCAGGCAGAGT





254
AGCCCGACAGCACCACGTTC
483
GACGGCCTTCCCCTCCTTTG





255
GCTGTGGCCACCACGCTCAA
484
CTTGCTGGGCTGAAGCGTGG





256
CAGGATCTGCCCGCCCACGT
485
TCTTCTCCGGGCTAGACGCC





257
GGTAGACATGGTAGGGCAGG
486
AGGCCAGCTCCTTTGGCGAC





258
CAACGTGGGCGGGCAGATCC
487
GGCGTCCTCAAAGGAGGGGA





259
CCTTGCCTTGGATAAACAGC
488
AGGAAGTCTAGGTCCACACT





260
TCGAGAAGAGCCTTCGGCTG
489
GGAGGGCGGGACACACTCTG





261
CCTTGCTGGGCTGAAGCGTG
490
CAGTGTGGACCTAGACTTCC





262
GGCCTGGTGAGGTAGTGACG
491
GTCACATTGCCCAGGCCCAC





263
TCTCGAGAAGAGCCTTCGGC
492
GCTTGGTGGGCAGGTGTGGA





264
ATTGAGCGTGGTGGCCACAG
493
GCCCTCAATGACTTCTTGCA





265
CTCGTCGGCGTCCAAGGAGG
494
CCACCGTTGTGACTGGAGGG





266
GCTGGCAAAAGCCTTGTTCT
495
TGACCAGCACAGACGCAGCG





267
CAGGAAGGGCTGCACACTCA
496
CAGCTGTTTATCCAAGGCAA





268
GATGAGCTGTCCACCTGTGT
497
TGACGGGCACCTGCTTGGCC





269
CAACATCACGGAGCAGACGC
498
CACAGAGTTCCCATTGAGCG





270
CAGGCCCACAGGCTGCAGGA
499
GGTCGTGCACAACACGGCCC





271
TGCAGGGTGGGCAGCTGGAA
500
TGCCATTGGGCAGGCCTTGG





272
CCTGCCCTACCATGTCTACC
501
CACCTGCTTGGCCAGGAGCT





273
AAAGTGGACGAGGCCACCAG
502
CAGGGAGTCCTGAGTGAGGA





274
CTCAATGGGAACTCTGTGTT
503
GGGCGTCAGCTGGTAGAGCT





275
TCGCGGTAGGTCTTGCGCAG
504
CTGTTTATCCAAGGCAAGGG





276
GGAAGGCCGTCTTGTAGTCG
505
GAAGGCACTTTGGGCCTCCC





277
GCCTTGGCCACATTCAGGGT
506
TTCTGGAGGATGATTCCGGA





278
GCAGAACCTGACGTTCATGG
507
AGTAGGAACTGGTTCTGGCC





279
GTCATCCTGCGGGCTGTCGC
508
AGGCCACCGTTGTGACTGGA





280
CCTCACAAACCAGAACCTGG
509
GGATGACGATGCTGCTGCCT





281
AACACGGGGCCCAGGCTCAG
510
CGGGCTGTCGCTGGAGAAGC





282
CATCCTCACAAACCAGAACC
511
GTGCACGACCTTGCTGAGCC





283
CTCCATGTGCAAGAAGCTTC
512
CTGGGGCAGGAACATCTGCA





284
AGGGGGAGTGGGGGACTTGT
513
GTCGCCAAAGGAGCTGGCCT





285
GCGCTGACTATGACCGAGGC
514
TCAAGATCAAGCAGGAAGCC





286
AAGCGGCCTTGGCCACATTC
515
ATCTGGAAGGTCCGAGGAGG





287
TTCAGGAGGTGGACGCTCAT
516
CTCGGCCACCTTGAGGGTAG





288
TTGTTCTCGGCTGGAAGCAG
517
CATGAACGTCAGGTTCTGCG





289
CTTCTCCAGCGACAGCCCGC
518
GGATGATTCCGGACGGCACC





290
CCTGGTAGACATGGTAGGGC
519
CCTCTTGGAGGCTCTGCTGG





291
GCTGGAGGATGTCACAGGGC
520
GAACTCTGTGTTCGGAGGCG





292
GGCGCCCTGGTAGACATGGT
521
ATATTTATTGAGCATGGCCT





293
GATGCCAGGCAGAGTTGGGG
522
CGACTCGGACTTGCGCCGCT





294
GGTCCACCGTGCCGTTCATC
523
CTCACAAACCAGAACCTGGC





295
CCTTGGCCACATTCAGGGTC
524
TTTGTGGTAGTCACTGGGGG





296
GCCATGCTCAACACCCCCGA
525
TCATTACCATCTCCGCTGAG





297
TGCTGTCGATGGCGCTCTGC
526
GAGCATTTGCACAAACACCA





298
GCCTCGTAGGTCTTGATGGG
527
TCGTCCACTTTGGCGGGCAG





299
GCAGGAAGGGCTGCACACTC
528
ATCCTGGGCTCTCCTGCGAC





300
CTGGAAGTCGGATGGCGTAG
529
GCATCTGGAAGAGCGCAGGC





301
GTAGGGCAGGAGGCGATGCA
530
CTCGCCCTGGATGGTGAGCG





302
TGGCGTAGGGGCTGGCAACC
531
TTGAGCATGGCCACGGCGCT





303
GCTCTGCTGGAGGATGTCAC
532
GGGCGTGTTGAGCGCCATGA





304
TCTGGCCCGGCAGCATGTGC
533
ATCCATGCAAGAAGTCATTG





305
GAGGGGGAGTGGGGGACTTG
534
TCGGCCACCTTGAGGGTAGC





306
AGGTAGTGACGGGGACGAAG
535
CCAGCTGTTTATCCAAGGCA





307
CAACCTCCCGACCCTGAATG
536
CTCCCAGCTCCTGGCCAAGC





308
GCTGGTTGGGCTCTGGGTTC
537
ACAAGCTTGGTGGGCAGGTG





309
GGGCCAGAACGTGGTGCTGT
538
GCTGTACCAGCGTATGCTGA





310
CACCGTTGTGACTGGAGGGT
539
CTTGGATAAACAGCTGGCCA





311
TCTCCTCCTGAATGAACATT
540
CGTAGGTCTTGATGGGCGGG





312
GCAGCCCTTCCTGCAGCCTG
541
TCCCCGCCGCCATGAACGTC





313
TCCTTGGACGCCGACGAGGA
542
GAGCCGATATTTATTGAGCA





314
GACCAGGTGGAGGGTTGGAG
543
GAGGCCACCGTTGTGACTGG





315
CAACCTGGAGGACGTCTCAG
544
AAGAAGTCATTGAGGGCCTG





316
CCGTGATGTTGGCCTCTTGG
545
CTCAAGATCAAGCAGGAAGC





317
TCCTGAGTGAGGATGGCCGT
546
CAGGTTCTGGTTTGTGAGGA





318
GGACACACTCTGTGGCCGGG
547
GTCAGGTTCTGCGGGGCCTT





319
ACTGACCAGCACAGACGCAG
548
CAAAGATGCCAGGCAGAGTT





320
TCCTCCAGAACAAGGCTGGG
549
GCAGGCTGCACCGTGAGGAC





321
GGAGCATTTGCACAAACACC
550
TGTTGAGCGCCATGACGGGC





322
ATCATCCTCCAGAACAAGGC
551
GTGCAAATGCTCCAGGAAAC





323
GGCCTTGTTGACCACGTCCT
552
AACCTGACGTTCATGGCGGC





324
GACACACTCTGTGGCCGGGA
553
CTTCCAGATGCCCGTGTCGC





325
GGGCCAGATGAACGGCACGG
554
TGCTGCCTGGGAAGAGGGCT





326
AGGTTCTGGTTTGTGAGGAT
555
ACACTCTGTGGCCGGGAGGG





327
AATGGGAACTCTGTGTTCGG
556
TCGGACTTGCGCCGCTTGGC





328
GCTCAAGCTCAAGATCAAGC
557
CTTGTTCTGGAGGATGATTC





329
GCACCTGCTTGGCCAGGAGC
558
GCAAAAGCCTTGTTCTCGGC





330
TTGTGGTAGTCACTGGGGGA
559
CGATGAGCTGTCCACCTGTG





331
GAAGCGTGGGGGGCTTCTTC
560
GCTGCCCGCCAAAGTGGACG





332
GTGCCGTTCATCTGGCCCGT
561
CCCCCCCAGCCTTGTTCTGG





333
CAGCGGCCGAGAGGTTCTTC
562
GCCGAGGCCACCGTTGTGAC





334
GACTATGACCGAGGCAGGCA
563
CGACCGAATGTTCATTCAGG





335
AGCTCCTTTGGCGACGGGGG
564
CTTCCTGCAGCCTGTGGGCC





336
GGACGCTCATGGGTTTGCTC
565
CCGCCAGGTTCTGGTTTGTG





337
CCGTCTTGTAGTCGGGGTGC
566
TCCAGTCACAACGGTGGCCT





338
CCCCATCGCAGCCTCTTCCG
567
GATATTTATTGAGCATGGCC





339
TGTGACACTGCAGCCCATCC
568
GGCTGCCATTGGGCAGGCCT





340
GGGGAAGGCCGTCTTGTAGT
569
TGGTGGGTCAGCACGGTAGG





341
CCCGCAGAACCTGACGTTCA
570
TTCCTGCAGCCTGTGGGCCT





342
CATCACGGAGCAGACGCTGG
571
GCGCCCTGGTAGACATGGTA





343
AAGCTGGCCTGGTCCACGTC
572
TGCCGGAAGCTTCTTGCACA





344
TGTGGTAGTCACTGGGGGAG
573
CCTCCAGCAGAGCCTCCAAG





345
GCTGCGTCTGTGCTGGTCAG
574
GTAGTCACTGGGGGAGGGGA





346
GGTGTTTGTGCAAATGCTCC
575
CGTCAGGTTCTGCGGGGCCT





347
GGAAGTCTAGGTCCACACTG
576
CAGGTTCAGGATGCTGTCGA





348
GAACCTGACGTTCATGGCGG
577
TGTCGCTGGAGAAGCTGGCC





349
GTAGTGACGGGGACGAAGAG
578
GGCCAGAACGTGGTGCTGTC





350
GACGCTCATGGGTTTGCTCA
579
GAGCTGTCCACCTGTGTGGG





351
CCTCCAGAACAAGGCTGGGG
580
GCTCCAGTTCCCACCCAGCC





352
CGGAATCATCCTCCAGAACA
581
GGCTGGGTGGGAACTGGAGC





353
GCCTGGTGGGTCAGCACGGT
















TABLE 3







Control sgRNA Library










SEQ





ID





NO.
gRNA Label
Gene
Nucleic Acid Sequence





582
1|sg_Non_Targeting_Human_0001|
Non-Targeting_Human
GTAGCGAACGTGTCCGGCGT



Non_Targeting_Human







583
1|sg_Non_Targeting_Human_0002|
Non-Targeting_Human
GACCGGAACGATCTCGCGTA



Non_Targeting_Human







584
1|sg_Non_Targeting_Human_0003|
Non-Targeting_Human
GGCAGTCGTTCGGTTGATAT



Non_Targeting_Human







585
1|sg_Non_Targeting_Human_0004|
Non-Targeting_Human
GCTTGAGCACATACGCGAAT



Non_Targeting_Human







586
1|sg_Non_Targeting_Human_0005|
Non-Targeting_Human
GTGGTAGAATAACGTATTAC



Non_Targeting_Human







587
1|sg_Non_Targeting_Human_0006|
Non-Targeting_Human
GTCATACATGGATAAGGCTA



Non_Targeting_Human







588
1|sg_Non_Targeting_Human_0007|
Non-Targeting_Human
GATACACGAAGCATCACTAG



Non_Targeting_Human







589
1|sg_Non_Targeting_Human_0008|
Non-Targeting_Human
GAACGTTGGCACTACTTCAC



Non_Targeting_Human







590
1|sg_Non_Targeting_Human_0009|
Non-Targeting_Human
GATCCATGTAATGCGTTCGA



Non_Targeting_Human







591
1|sg_Non_Targeting_Human_0010|
Non-Targeting_Human
GTCGTGAAGTGCATTCGATC



Non_Targeting_Human







592
1|sg_Non_Targeting_Human_0011|
Non-Targeting_Human
GTTCGACTCGCGTGACCGTA



Non_Targeting_Human







593
1|sg_Non_Targeting_Human_0012|
Non-Targeting_Human
GAATCTACCGCAGCGGTTCG



Non_Targeting_Human







594
1|sg_Non_Targeting_Human_0013|
Non-Targeting_Human
GAAGTGACGTCGATTCGATA



Non_Targeting_Human







595
1|sg_Non_Targeting_Human_0014|
Non-Targeting_Human
GCGGTGTATGACAACCGCCG



Non_Targeting_Human







596
1|sg_Non_Targeting_Human_0015|
Non-Targeting_Human
GTACCGCGCCTGAAGTTCGC



Non_Targeting_Human







597
1|sg_Non_Targeting_Human_0016|
Non-Targeting_Human
GCAGCTCGTGTGTCGTACTC



Non_Targeting_Human







598
1|sg_Non_Targeting_Human_0017|
Non-Targeting_Human
GCGCCTTAAGAGTACTCATC



Non_Targeting_Human







599
1|sg_Non_Targeting_Human_0018|
Non-Targeting_Human
GAGTGTCGTCGTTGCTCCTA



Non_Targeting_Human







600
1|sg_Non_Targeting_Human_0019|
Non-Targeting_Human
GCAGCTCGACCTCAAGCCGT



Non_Targeting_Human







601
1|sg_Non_Targeting_Human_0020|
Non-Targeting_Human
GTATCCTGACCTACGCGCTG



Non_Targeting_Human







602
1|sg_Non_Targeting_Human_0021|
Non-Targeting_Human
GTGTATCTCAGCACGCTAAC



Non_Targeting_Human







603
1|sg_Non_Targeting_Human_0022|
Non-Targeting_Human
GTCGTCATACAACGGCAACG



Non_Targeting_Human







604
1|sg_Non_Targeting_Human_0023|
Non-Targeting_Human
GTCGTGCGCTTCCGGCGGTA



Non_Targeting_Human







605
1|sg_Non_Targeting_Human_0024|
Non-Targeting_Human
GCGGTCCTCAGTAAGCGCGT



Non_Targeting_Human







606
1|sg_Non_Targeting_Human_0025|
Non-Targeting_Human
GCTCTGCTGCGGAAGGATTC



Non_Targeting_Human







607
1|sg_Non_Targeting_Human_0026|
Non-Targeting_Human
GCATGGAGGAGCGTCGCAGA



Non_Targeting_Human







608
1|sg_Non_Targeting_Human_0027|
Non-Targeting_Human
GTAGCGCGCGTAGGAGTGGC



Non_Targeting_Human







609
1|sg_Non_Targeting_Human_0028|
Non-Targeting_Human
GATCACCTGCATTCGTACAC



Non_Targeting_Human







610
1|sg_Non_Targeting_Human_0029|
Non-Targeting_Human
GCACACCTAGATATCGAATG



Non_Targeting_Human







611
1|sg_Non_Targeting_Human_0030|
Non-Targeting_Human
GTTGATCAACGCGCTTCGCG



Non_Targeting_Human







612
1|sg_Non_Targeting_Human_0031|
Non-Targeting_Human
GCGTCTCACTCACTCCATCG



Non_Targeting_Human







613
1|sg_Non_Targeting_Human_0032|
Non-Targeting_Human
GCCGACCAACGTCAGCGGTA



Non_Targeting_Human







614
1|sg_Non_Targeting_Human_0033|
Non-Targeting_Human
GGATACGGTGCGTCAATCTA



Non_Targeting_Human







615
1|sg_Non_Targeting_Human_0034|
Non-Targeting_Human
GAATCCAGTGGCGGCGACAA



Non_Targeting_Human







616
1|sg_Non_Targeting_Human_0035|
Non-Targeting_Human
GCACTGTCAGTGCAACGATA



Non_Targeting_Human







617
1|sg_Non_Targeting_Human_0036|
Non-Targeting_Human
GCGATCCTCAAGTATGCTCA



Non_Targeting_Human







618
1|sg_Non_Targeting_Human_0037|
Non-Targeting_Human
GCTAATATCGACACGGCCGC



Non_Targeting_Human







619
1|sg_Non_Targeting_Human_0038|
Non-Targeting_Human
GGAGATGCATCGAAGTCGAT



Non_Targeting_Human







620
1|sg_Non_Targeting_Human_0039|
Non-Targeting_Human
GGATGCACTCCATCTCGTCT



Non_Targeting_Human







621
1|sg_Non_Targeting_Human_0040|
Non-Targeting_Human
GTGCCGAGTAATAACGCGAG



Non_Targeting_Human







622
1|sg_Non_Targeting_Human_0041|
Non-Targeting_Human
GAGATTCCGATGTAACGTAC



Non_Targeting_Human







623
1|sg_Non_Targeting_Human_0042|
Non-Targeting_Human
GTCGTCACGAGCAGGATTGC



Non_Targeting_Human







624
1|sg_Non_Targeting_Human_0043|
Non-Targeting_Human
GCGTTAGTCACTTAGCTCGA



Non_Targeting_Human







625
1|sg_Non_Targeting_Human_0044|
Non-Targeting_Human
GTTCACACGGTGTCGGATAG



Non_Targeting_Human







626
1|sg_Non_Targeting_Human_0045|
Non-Targeting_Human
GGATAGGTGACCTTAGTACG



Non_Targeting_Human







627
1|sg_Non_Targeting_Human_0046|
Non-Targeting_Human
GTATGAGTCAAGCTAATGCG



Non_Targeting_Human







628
1|sg_Non_Targeting_Human_0047|
Non-Targeting_Human
GCAACTATTGGAATACGTGA



Non_Targeting_Human







629
1|sg_Non_Targeting_Human_0048|
Non-Targeting_Human
GTTACCTTCGCTCGTCTATA



Non_Targeting_Human







630
1|sg_Non_Targeting_Human_0049|
Non-Targeting_Human
GTACCGAGCACCACAGGCCG



Non_Targeting_Human







631
1|sg_Non_Targeting_Human_0050|
Non-Targeting_Human
GTCAGCCATCGGATAGAGAT



Non_Targeting_Human







632
1|sg_Non_Targeting_Human_0051|
Non-Targeting_Human
GTACGGCACTCCTAGCCGCT



Non_Targeting_Human







633
1|sg_Non_Targeting_Human_0052|
Non-Targeting_Human
GGTCCTGTCGTATGCTTGCA



Non_Targeting_Human







634
1|sg_Non_Targeting_Human_0053|
Non-Targeting_Human
GCCGCAATATATGCGGTAAG



Non_Targeting_Human







635
1|sg_Non_Targeting_Human_0054|
Non-Targeting_Human
GCGCACGTATAATCCTGCGT



Non_Targeting_Human







636
1|sg_Non_Targeting_Human_0055|
Non-Targeting_Human
GTGCACAACACGATCCACGA



Non_Targeting_Human







637
1|sg_Non_Targeting_Human_0056|
Non-Targeting_Human
GCACAATGTTGACGTAAGTG



Non_Targeting_Human







638
1|sg_Non_Targeting_Human_0057|
Non-Targeting_Human
GTAAGATGCTGCTCACCGTG



Non_Targeting_Human







639
1|sg_Non_Targeting_Human_0058|
Non-Targeting_Human
GTCGGTGATCCAACGTATCG



Non_Targeting_Human







640
1|sg_Non_Targeting_Human_0059|
Non-Targeting_Human
GAGCTAGTAGGACGCAAGAC



Non_Targeting_Human







641
1|sg_Non_Targeting_Human_0060|
Non-Targeting_Human
GTACGTGGAAGCTTGTGGCC



Non_Targeting_Human







642
1|sg_Non_Targeting_Human_0061|
Non-Targeting_Human
GAGAACTGCCAGTTCTCGAT



Non_Targeting_Human







643
1|sg_Non_Targeting_Human_0062|
Non-Targeting_Human
GCCATTCGGCGCGGCACTTC



Non_Targeting_Human







644
1|sg_Non_Targeting_Human_0063|
Non-Targeting_Human
GCACACGACCAATCCGCTTC



Non_Targeting_Human







645
1|sg_Non_Targeting_Human_0064|
Non-Targeting_Human
GAGGTGATCGATTAAGTACA



Non_Targeting_Human







646
1|sg_Non_Targeting_Human_0065|
Non-Targeting_Human
GTCACTCGCAGACGCCTAAC



Non_Targeting_Human







647
1|sg_Non_Targeting_Human_0066|
Non-Targeting_Human
GCGCTACGGAATCATACGTT



Non_Targeting_Human







648
1|sg_Non_Targeting_Human_0067|
Non-Targeting_Human
GGTAGGACCTCACGGCGCGC



Non_Targeting_Human







649
1|sg_Non_Targeting_Human_0068|
Non-Targeting_Human
GAACTGCATCTTGTTGTAGT



Non_Targeting_Human







650
1|sg_Non_Targeting_Human_0069|
Non-Targeting_Human
GATCCTGATCCGGCGGCGCG



Non_Targeting_Human







651
1|sg_Non_Targeting_Human_0070|
Non-Targeting_Human
GGTATGCGCGATCCTGAGTT



Non_Targeting_Human







652
1|sg_Non_Targeting_Human_0071|
Non-Targeting_Human
GCGGAGCTAGAGAGCGGTCA



Non_Targeting_Human







653
1|sg_Non_Targeting_Human_0072|
Non-Targeting_Human
GAATGGCAATTACGGCTGAT



Non_Targeting_Human







654
1|sg_Non_Targeting_Human_0073|
Non-Targeting_Human
GTATGGTGAGTAGTCGCTTG



Non_Targeting_Human







655
1|sg_Non_Targeting_Human_0074|
Non-Targeting_Human
GTGTAATTGCGTCTAGTCGG



Non_Targeting_Human







656
1|sg_Non_Targeting_Human_0075|
Non-Targeting_Human
GGTCCTGGCGAGGAGCCTTG



Non_Targeting_Human







657
1|sg_Non_Targeting_Human_0076|
Non-Targeting_Human
GAAGATAAGTCGCTGTCTCG



Non_Targeting_Human







658
1|sg_Non_Targeting_Human_0077|
Non-Targeting_Human
GTCGGCGTTCTGTTGTGACT



Non_Targeting_Human







659
1|sg_Non_Targeting_Human_0078|
Non-Targeting_Human
GAGGCAAGCCGTTAGGTGTA



Non_Targeting_Human







660
1|sg_Non_Targeting_Human_0079|
Non-Targeting_Human
GCGGATCCAGATCTCATTCG



Non_Targeting_Human







661
1|sg_Non_Targeting_Human_0080|
Non-Targeting_Human
GGAACATAGGAGCACGTAGT



Non_Targeting_Human







662
1|sg_Non_Targeting_Human_0081|
Non-Targeting_Human
GTCATCATTATGGCGTAAGG



Non_Targeting_Human







663
1|sg_Non_Targeting_Human_0082|
Non-Targeting_Human
GCGACTAGCGCCATGAGCGG



Non_Targeting_Human







664
1|sg_Non_Targeting_Human_0083|
Non-Targeting_Human
GGCGAAGTTCGACATGACAC



Non_Targeting_Human







665
1|sg_Non_Targeting_Human_0084|
Non-Targeting_Human
GCTGTCGTGTGGAGGCTATG



Non_Targeting_Human







666
1|sg_Non_Targeting_Human_0085|
Non-Targeting_Human
GCGGAGAGCATTGACCTCAT



Non_Targeting_Human







667
1|sg_Non_Targeting_Human_0086|
Non-Targeting_Human
GACTAATGGACCAAGTCAGT



Non_Targeting_Human







668
1|sg_Non_Targeting_Human_0087|
Non-Targeting_Human
GCGGATTAGAGGTAATGCGG



Non_Targeting_Human







669
1|sg_Non_Targeting_Human_0088|
Non-Targeting_Human
GCCGACGGCAATCAGTACGC



Non_Targeting_Human







670
1|sg_Non_Targeting_Human_0089|
Non-Targeting_Human
GTAACCTCTCGAGCGATAGA



Non_Targeting_Human







671
1|sg_Non_Targeting_Human_0090|
Non-Targeting_Human
GACTTGTATGTGGCTTACGG



Non_Targeting_Human







672
1|sg_Non_Targeting_Human_0091|
Non-Targeting_Human
GTCACTGTGGTCGAACATGT



Non_Targeting_Human







673
1|sg_Non_Targeting_Human_0092|
Non-Targeting_Human
GTACTCCAATCCGCGATGAC



Non_Targeting_Human







674
1|sg_Non_Targeting_Human_0093|
Non-Targeting_Human
GCGTTGGCACGATGTTACGG



Non_Targeting_Human







675
1|sg_Non_Targeting_Human_0094|
Non-Targeting_Human
GAACCAGCCGGCTAGTATGA



Non_Targeting_Human







676
1|sg_Non_Targeting_Human_0095|
Non-Targeting_Human
GTATACTAGCTAACCACACG



Non_Targeting_Human







677
1|sg_Non_Targeting_Human_0096|
Non-Targeting_Human
GAATCGGAATAGTTGATTCG



Non_Targeting_Human







678
1|sg_Non_Targeting_Human_0097|
Non-Targeting_Human
GAGCACTTGCATGAGGCGGT



Non_Targeting_Human







679
1|sg_Non_Targeting_Human_0098|
Non-Targeting_Human
GAACGGCGATGAAGCCAGCC



Non_Targeting_Human







680
1|sg_Non_Targeting_Human_0099|
Non-Targeting_Human
GCAACCGAGATGAGAGGTTC



Non_Targeting_Human







681
1|sg_Non_Targeting_Human_0100|
Non-Targeting_Human
GCAAGATCAATATGCGTGAT



Non_Targeting_Human







682
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACGGAGGCTAAGCGTCGCAA



0101|Non_Targeting_Human







683
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCTTCCGCGGCCCGTTCAA



0102|Non_Targeting_Human







684
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ATCGTTTCCGCTTAACGGCG



0103|Non_Targeting_Human







685
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTAGGCGCGCCGCTCTCTAC



0104|Non_Targeting_Human







686
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CCATATCGGGGCGAGACATG



0105|Non_Targeting_Human







687
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TACTAACGCCGCTCCTACAG



0106|Non_Targeting_Human







688
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TGAGGATCATGTCGAGCGCC



0107|Non_Targeting_Human







689
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GGGCCCGCATAGGATATCGC



0108|Non_Targeting_Human







690
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TAGACAACCGCGGAGAATGC



0109|Non_Targeting_Human







691
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACGGGCGGCTATCGCTGACT



0110|Non_Targeting_Human







692
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCGGAAATTTTACCGACGA



0111|Non_Targeting_Human







693
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTTACAATCGTCGGTCCAAT



0112|Non_Targeting_Human







694
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GCGTGCGTCCCGGGTTACCC



0113|Non_Targeting_Human







695
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGGAGTAACAAGCGGACGGA



0114|Non_Targeting_Human







696
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGAGTGTTATACGCACCGTT



0115|Non_Targeting_Human







697
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGACTAACCGGAAACTTTTT



0116|Non_Targeting_Human







698
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CAACGGGTTCTCCCGGCTAC



0117|Non_Targeting_Human







699
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CAGGAGTCGCCGATACGCGT



0118|Non_Targeting_Human







700
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TTCACGTCGTCTCGCGACCA



0119|Non_Targeting_Human







701
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTGTCGGATTCCGCCGCTTA



0120|Non_Targeting_Human







702
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CACGAACTCACACCGCGCGA



0121|Non_Targeting_Human







703
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCTAGTACGCTCCTCTATA



0122|Non_Targeting_Human







704
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCGCGCTTGGGTTATACGCT



0123|Non_Targeting_Human







705
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTATCTCGAGTGGTAATGCG



0124|Non_Targeting_Human







706
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AATCGACTCGAACTTCGTGT



0125|Non_Targeting_Human







707
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CCCGATGGACTATACCGAAC



0126|Non_Targeting_Human







708
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACGTTCGAGTACGACCAGCT



0127|Non_Targeting_Human







709
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCGACGACTCAACCTAGTC



0128|Non_Targeting_Human







710
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GGTCACCGATCGAGAGCTAG



0129|Non_Targeting_Human







711
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTCAACCGACCGTATGGTCA



0130|Non_Targeting_Human







712
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGTATTCGACTCTCAACGCG



0131|Non_Targeting_Human







713
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTAGCCGCCCAGATCGAGCC



0132|Non_Targeting_Human







714
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GAATCGACCGACACTAATGT



0133|Non_Targeting_Human







715
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACTTCAGTTCGGCGTAGTCA



0134|Non_Targeting_Human







716
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTGCGATGTCGCTTCAACGT



0135|Non_Targeting_Human







717
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCCTAATTTCCGGATCAAT



0136|Non_Targeting_Human







718
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGTGGCCGGAACCGTCATAG



0137|Non_Targeting_Human







719
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACCCTCCGAATCGTAACGGA



0138|Non_Targeting_Human







720
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AAACGGTACGACAGCGTGTG



0139|Non_Targeting_Human







721
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACATAGTCGACGGCTCGATT



0140|Non_Targeting_Human







722
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GATGGCGCTTCAGTCGTCGG



0141|Non_Targeting_Human







723
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ATAATCCGGAAACGCTCGAC



0142|Non_Targeting_Human







724
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCCGGGCTGACAATTAACG



0143|Non_Targeting_Human







725
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGTCGCCATATGCCGGTGGC



0144|Non_Targeting_Human







726
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGGGCCTATAACACCATCGA



0145|Non_Targeting_Human







727
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCCGTTCCGAGATACTTGA



0146|Non_Targeting_Human







728
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGGGACGTCGCGAAAATGTA



0147|Non_Targeting_Human







729
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCGGCATACGGGACACACGC



0148|Non_Targeting_Human







730
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AGCTCCATCGCCGCGATAAT



0149|Non_Targeting_Human







731
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ATCGTATCATCAGCTAGCGC



0150|Non_Targeting_Human







732
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCGATCGAGGTTGCATTCGG



0151|Non_Targeting_Human







733
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTCGACAGTTCGTCCCGAGC



0152|Non_Targeting_Human







734
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGGTAGTATTAATCGCTGAC



0153|Non_Targeting_Human







735
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TGAACGCGTGTTTCCTTGCA



0154|Non_Targeting_Human







736
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGACGCTAGGTAACGTAGAG



0155|Non_Targeting_Human







737
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CATTGTTGAGCGGGCGCGCT



0156|Non_Targeting_Human







738
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CCGCTATTGAAACCGCCCAC



0157|Non_Targeting_Human







739
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AGACACGTCACCGGTCAAAA



0158|Non_Targeting_Human







740
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TTTACGATCTAGCGGCGTAG



0159|Non_Targeting_Human







741
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TTCGCACGATTGCACCTTGG



0160|Non_Targeting_Human







742
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GGTTAGAGACTAGGCGCGCG



0161|Non_Targeting_Human







743
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CCTCCGTGCTAACGCGGACG



0162|Non_Targeting_Human







744
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TTATCGCGTAGTGCTGACGT



0163|Non_Targeting_Human







745
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TACGCTTGCGTTTAGCGTCC



0164|Non_Targeting_Human







746
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCGGCCCACGCGTCATCGC



0165|Non_Targeting_Human







747
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AGCTCGCCATGTCGGTTCTC



0166|Non_Targeting_Human







748
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AACTAGCCCGAGCAGCTTCG



0167|Non_Targeting_Human







749
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCAAGGTGTCGGTAACCCT



0168|Non_Targeting_Human







750
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CTTCGACGCCATCGTGCTCA



0169|Non_Targeting_Human







751
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCCTGGATACCGCGTGGTTA



0170|Non_Targeting_Human







752
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ATAGCCGCCGCTCATTACTT



0171|Non_Targeting_Human







753
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTCGTCCGGGATTACAAAAT



0172|Non_Targeting_Human







754
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TAATGCTGCACACGCCGAAT



0173|Non_Targeting_Human







755
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TATCGCTTCCGATTAGTCCG



0174|Non_Targeting_Human







756
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTACCATACCGCGTACCCTT



0175|Non_Targeting_Human







757
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TAAGATCCGCGGGTGGCAAC



0176|Non_Targeting_Human







758
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTAGACGTCGTGAGCTTCAC



0177|Non_Targeting_Human







759
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCGCGGACATAGGGCTCTAA



0178|Non_Targeting_Human







760
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AGCGCAGATAGCGCGTATCA



0179|Non_Targeting_Human







761
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTTCGCTTCGTAACGAGGAA



0180|Non_Targeting_Human







762
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GACCCCCGATAACTTTTGAC



0181|Non_Targeting_Human







763
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACGTCCATACTGTCGGCTAC



0182|Non_Targeting_Human







764
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTACCATTGCCGGCTCCCTA



0183|Non_Targeting_Human







765
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TGGTTCCGTAGGTCGGTATA



0184|Non_Targeting_Human







766
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCTGGCTTGACACGACCGTT



0185|Non_Targeting_Human







767
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGCTAGGTCCGGTAAGTGCG



0186|Non_Targeting_Human







768
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AGCACGTAATGTCCGTGGAT



0187|Non_Targeting_Human







769
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AAGGCGCGCGAATGTGGCAG



0188|Non_Targeting_Human







770
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ACTGCGGAGCGCCCAATATC



0189|Non_Targeting_Human







771
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGTCGAGTGCTCGAACTCCA



0190|Non_Targeting_Human







772
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TCGCAGCGGCGTGGGATCGG



0191|Non_Targeting_Human







773
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
ATCTGTCCTAATTCGGATCG



0192|Non_Targeting_Human







774
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TGCGGCGTAATGCTTGAAAG



0193|Non_Targeting_Human







775
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CGAACTTAATCCCGTGGCAA



0194|Non_Targeting_Human







776
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GCCGTGTTGCTGGATACGCC



0195|Non_Targeting_Human







777
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
TACCCTCCGGATACGGACTG



0196|Non_Targeting_Human







778
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
CCGTTGGACTATGGCGGGTC



0197|Non_Targeting_Human







779
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
GTACGGGGCGATCATCCACA



0198|Non_Targeting_Human







780
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AAGAGTAGTAGACGCCCGGG



0199|Non_Targeting_Human







781
1|sg_Non_Targeting_Human_GA_
Non-Targeting_Human
AAGAGCGAATCGATTTCGTG



0200|Non_Targeting_Human







782
3|sg_hCDC16_CC_1|CDC16
CDC16
TCAACACCAGTGCCTGACGG





783
3|sg_hCDC16_CC_2|CDC16
CDC16
AAAGTAGCTTCACTCTCTCG





784
3|sg_hCDC16_CC_3|CDC16
CDC16
GAGCCAACCAATAGATGTCC





785
3|sg_hCDC16_CC_4|CDC16
CDC16
GCGCCGCCATGAACCTAGAG





786
3|sg_hGTF2B_CC_1|GTF2B
GTF2B
ACAAAGGTTGGAACAGAACC





787
3|sg_hGTF2B_CC_2|GTF2B
GTF2B
GGTGACCGGGTTATTGATGT





788
3|sg_hGTF2B_CC_3|GTF2B
GTF2B
TTAGTGGAGGACTACAGAGC





789
3|sg_hGTF2B_CC_4|GTF2B
GTF2B
ACATATAGCCCGTAAAGCTG





790
3|sg_hHSPA5_CC_1|HSPA5
HSPA5
CGTTGGCGATGATCTCCACG





791
3|sg_hHSPA5_CC_2|HSPA5
HSPA5
TGGCCTTTTCTACCTCGCGC





792
3|sg_hHSPA5_CC_3|HSPA5
HSPA5
AATGGAGATACTCATCTGGG





793
3|sg_hHSPA5_CC_4|HSPA5
HSPA5
GAAGCCCGTCCAGAAAGTGT





794
3|sg_hHSPA9_CC_1|HSPA9
HSPA9
CAATCTGAGGAACTCCACGA





795
3|sg_hHSPA9_CC_2|HSPA9
HSPA9
AGGCTGCGGCGCCCACGAGA





796
3|sg_hHSPA9_CC_3|HSPA9
HSPA9
ACTTTGACCAGGCCTTGCTA





797
3|sg_hHSPA9_CC_4|HSPA9
HSPA9
ACCTTCCATAACTGCCACGC





798
3|sg_hPAFAH1B1_CC_1|PAFAH1B1
PAFAH1B1
CGAGGCGTACATACCCAAGG





799
3|sg_hPAFAH1B1_CC_2|PAFAH1B1
PAFAH1B1
ATGGTACGGCCAAATCAAGA





800
3|sg_hPAFAH1B1_CC_3|PAFAH1B1
PAFAH1B1
TCTTGTAATCCCATACGCGT





801
3|sg_hPAFAH1B1_CC_4|PAFAH1B1
PAFAH1B1
ATTCACAGGACACAGAGAAT





802
3|sg_hPCNA_CC_1|PCNA
PCNA
CCAGGGCTCCATCCTCAAGA





803
3|sg_hPCNA_CC_2|PCNA
PCNA
TGAGCTGCACCAAAGAGACG





804
3|sg_hPCNA_CC_3|PCNA
PCNA
ATGTCTGCAGATGTACCCCT





805
3|sg_hPCNA_CC_4|PCNA
PCNA
CGAAGATAACGCGGATACCT





806
3|sg_hPOLR2L_CC_1|POLR2L
POLR2L
GCTGCAGGCCGAGTACACCG





807
3|sg_hPOLR2L_CC_2|POLR2L
POLR2L
ACAAGTGGGAGGCTTACCTG





808
3|sg_hPOLR2L_CC_3|POLR2L
POLR2L
GCAGCGTACAGGGATGATCA





809
3|sg_hPOLR2L_CC_4|POLR2L
POLR2L
GCAGTAGCGCTTCAGGCCCA





810
3|sg_hRPL9_CC_1|RPL9
RPL9
CAAATGGTGGGGTAACAGAA





811
3|sg_hRPL9_CC_2|RPL9
RPL9
GAAAGGAACTGGCTACCGTT





812
3|sg_hRPL9_CC_3|RPL9
RPL9
AGGGCTTCCGTTACAAGATG





813
3|sg_hRPL9_CC_4|RL9
RPL9
GAACAAGCAACACCTAAAAG





814
3|sg_hSF3A3_CC_1|SF3A3
SF3A3
TGAGGAGAAGGAACGGCTCA





815
3|sg_hSF3A3_CC_2|SF3A3
SF3A3
GGAAGAATGCAGAGTATAAG





816
3|sg_hSF3A3_CC_3|SF3A3
SF3A3
GGAATTTGAGGAACTCCTGA





817
3|sg_hSF3A3_CC_4|SF3A3
SF3A3
GCTCACCGGCCATCCAGGAA





818
3|sg_hSF3B3_CC_1|SF3B3
SF3B3
ACTGGCCAGGAACGATGCGA





819
3|sg_hSF3B3_CC_2|SF3B3
SF3B3
GCAGCTCCAAGATCTTCCCA





820
3|sg_hSF3B3_CC_3|SF3B3
SF3B3
GAATGAGTACACAGAACGGA





821
3|sg_hSF3B3_CC_4|SF3B3
SF3B3
GGAGCAGGACAAGGTCGGGG









Other Embodiments

All publications, patents, and patent applications mentioned in this specification are incorporated herein by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Where a term in the present application is found to be defined differently in a document incorporated herein by reference, the definition provided herein is to serve as the definition for the term.


While the invention has been described in connection with specific embodiments thereof, it will be understood that invention is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims.


Other embodiments are in the claims.

Claims
  • 1. A method of treating soft tissue sarcoma in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the sarcoma.
  • 2. A method of reducing tumor growth of a soft tissue sarcoma in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the tumor.
  • 3. A method of inducing apoptosis in a soft tissue sarcoma cell, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell.
  • 4. A method of reducing the level and/or activity of BICRA in a soft tissue sarcoma cell, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell.
  • 5. The method of claim 3 or 4, wherein the soft tissue sarcoma cell is in a subject.
  • 6. The method of any one of claims 1 to 5, wherein the subject or cell has been identified as expressing SS18-SSX fusion protein or BICRA fusion protein.
  • 7. The method of any one of claims 1 to 6, wherein the effective amount of the agent reduces the level and/or activity of BICRA by at least 5% as compared to a reference.
  • 8. The method of any one of claims 1 to 7, wherein the effective amount of the agent reduces the level and/or activity of BICRA by at least 5% as compared to a reference for at least 12 hours.
  • 9. The method of any one of claims 1 to 8, wherein the level and/or activity of SS18-SSX or BICRA fusion protein is reduced in the subject or cell.
  • 10. The method of any one of claims 1 to 9, wherein the soft tissue sarcoma is adult soft tissue sarcoma.
  • 11. The method of claim 10, wherein the adult soft tissue sarcoma is synovial sarcoma.
  • 12. A method of modulating the activity of an SS18-SSX fusion protein, SS18 wild-type protein, or SSX wild-type protein in a cell, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell.
  • 13. A method of modulating the level and/or activity of an SS18-SSX fusion protein, SS18 wild-type protein, or SSX wild-type protein in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in a cell or subject.
  • 14. The method of claim 12 or 13, wherein the cell is in a subject.
  • 15. A method of treating a disorder related to an SS18-SSX fusion protein, SS18 wild-type protein, or SSX wild-type protein in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in an SS18-SSX fusion protein-expressing cell in the subject.
  • 16. The method of any one of claims 12 to 15, wherein the subject has cancer.
  • 17. The method of claim 16, wherein the cancer expresses SS18-SSX fusion protein and/or the cell or subject has been identified as expressing SS18-SSX fusion protein.
  • 18. The method of any one of claims 15 to 17, wherein the disorder is synovial sarcoma or Ewing's sarcoma.
  • 19. The method of claim 18, wherein the disorder is synovial sarcoma.
  • 20. A method of modulating the activity of a BAF complex in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 21. A method of increasing the level and/or activity of BAF47 in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 22. A method of decreasing Wnt/β-catenin signaling in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 23. A method treating a disorder related to BAF47 in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the subject.
  • 24. The method of claim 23, wherein the disorder related to BAF47 is a cancer or viral infection.
  • 25. The method of claim 24, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer.
  • 26. The method of claim 24, wherein the viral infection is an infection with a virus of the Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or Togaviridae family.
  • 27. A method for treating cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a cancer cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.
  • 28. A method of reducing tumor growth of a cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a tumor cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.
  • 29. A method of inducing apoptosis in a cancer cell, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.
  • 30. A method of reducing the level and/or activity of BICRA in a cancer cell, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.
  • 31. The method of any one of claims 27 to 30, wherein the cancer is a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer.
  • 32. The method of any one of claims 27 to 31, wherein the cancer is non-small cell lung cancer, stomach cancer, breast cancer, malignant rhabdoid tumor, multiple myeloma, or atypical teratoid rhabdoid tumor.
  • 33. A method of modulating the activity of a BICRA fusion protein in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 34. A method of modulating the level and/or activity of a BICRA fusion protein in a cell or subject, the method comprising contacting the cell with an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 35. The method of claim 33 or 34, wherein the cell is in a subject.
  • 36. A method of treating a disorder related to a BICRA fusion protein in a subject in need thereof, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a BICRA fusion protein-expressing cell.
  • 37. The method of any one of claims 33 to 36, wherein the subject has cancer.
  • 38. The method of claim 37, wherein the cancer expresses a BICRA fusion protein and/or the cell or subject has been identified as expressing a BICRA fusion protein.
  • 39. The method of any one of claims 36 to 38, wherein the disorder related to a BICRA fusion protein is Ewing's sarcoma, lung cancer, or renal cancer.
  • 40. The method of any one of claims 1 to 39, wherein the method further comprises administering to the subject or contacting the cell with an anticancer therapy.
  • 41. The method of claim 40, wherein the anticancer therapy is a chemotherapeutic or cytotoxic agent or radiotherapy.
  • 42. The method of claim 41, wherein the chemotherapeutic or cytotoxic agent is doxorubicin or ifosfamide.
  • 43. The method of claim 41 or 42, wherein the anticancer therapy and the agent that reduces the level and/or activity of BICRA in a cell are administered within 28 days of each other and each in an amount that together are effective to treat the subject.
  • 44. The method of any one of claims 1 to 43, wherein the subject or cancer has been identified as having an elevated level of an SS18-SSX fusion protein or a BICRA fusion protein as compared to a reference.
  • 45. The method of any one of claims 1 to 44, wherein the subject or cancer has been identified as having a decreased level of SS18 wild-type protein or SSX wild-type protein as compared to a reference.
  • 46. A method of treating a viral infection, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in a cell of the subject.
  • 47. The method of claim 46, wherein the viral infection is an infection with a virus of the Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or Togaviridae family.
  • 48. The method of any one of claims 1 to 47, wherein the agent that reduces the level and/or activity of BICRA in a cell is a small molecule compound, an antibody, an enzyme, and/or a polynucleotide.
  • 49. The method of claim 48, wherein the agent that reduces the level and/or activity of BICRA in a cell is an enzyme.
  • 50. The method of claim 49, wherein the enzyme is a clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein, a zinc finger nuclease (ZFN), a transcription activator-like effector nuclease (TALEN), or a meganuclease.
  • 51. The method of claim 50, wherein the CRISPR-associated protein is CRISPR-associated protein 9 (Cas9).
  • 52. The method of claim 48, wherein the agent that reduces the level and/or activity of BICRA in a cell is a polynucleotide.
  • 53. The method of claim 52, wherein the polynucleotide is an antisense nucleic acid, a short interfering RNA (siRNA), a short hairpin RNA (shRNA), a micro RNA (miRNA), a CRISPR/Cas 9 nucleotide, or a ribozyme.
  • 54. The method of claim 52, wherein the polynucleotide comprises a sequence having at least 85% sequence identity to the nucleic acid sequence of any one of SEQ ID NOs: 3-124.
  • 55. The method of claim 54, wherein the polynucleotide comprises a sequence having at least 85% sequence identity to the nucleic acid sequence of any one of SEQ ID NOs: 3-68.
  • 56. The method of claim 48, wherein the agent that reduces the level and/or activity of BICRA in a cell is a small molecule compound.
  • 57. The method of claim 56, wherein the small molecule compound is a small molecule BICRA inhibitor.
  • 58. The method of claim 56 or 57, wherein the small molecule compound is a degrader.
  • 59. The method of claim 58, wherein the degrader has the structure of Formula I: A-L-B   Formula I
  • 60. The method of claim 59, wherein the degradation moiety is a ubiquitin ligase binding moiety.
  • 61. The method of claim 60, wherein the ubiquitin ligase binding moiety comprises Cereblon ligands, IAP (Inhibitors of Apoptosis) ligands, mouse double minute 2 homolog (MDM2), or von Hippel-Lindau ligands, or derivatives or analogs thereof.
  • 62. The method of claim 60 or 61, wherein the ubiquitin ligase binding moiety has the structure:
  • 63. The method of any one of claims 59 to 62, wherein the linker has the structure of Formula II: A1-(B1)f—(C1)g—(B2)h-(D)-(B3)i—(C2)j—(B4)k-A2   Formula II
  • 64. A method of treating cancer in a subject determined to have an elevated level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein, the method comprising administering to the subject an effective amount of an agent that reduces the level and/or activity of BICRA in the cell or subject.
  • 65. The method of claim 64, wherein the level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein in the subject is measured in one or more cancer cells.
  • 66. The method of claim 64 or 65, wherein the level of SS18-SSX fusion protein, SS18 wild-type protein, SSX wild-type protein, or a BICRA fusion protein in the subject is measured systemically.
  • 67. A composition comprising an adult soft tissue sarcoma cell and an agent that reduces the level and/or activity of BICRA in a cell.
PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/038296 6/20/2019 WO 00
Provisional Applications (1)
Number Date Country
62688274 Jun 2018 US