WO2016138574A1 - Homologous recombination factors - Google Patents

Homologous recombination factors Download PDF

Info

Publication number
WO2016138574A1
WO2016138574A1 PCT/CA2016/000057 CA2016000057W WO2016138574A1 WO 2016138574 A1 WO2016138574 A1 WO 2016138574A1 CA 2016000057 W CA2016000057 W CA 2016000057W WO 2016138574 A1 WO2016138574 A1 WO 2016138574A1
Authority
WO
WIPO (PCT)
Prior art keywords
palb2
cell
brca1
keap1
brca2
Prior art date
Application number
PCT/CA2016/000057
Other languages
French (fr)
Inventor
Daniel Durocher
Alexandre Orthwein
Sylvie NOORDERMEER
Original Assignee
Sinai Health System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sinai Health System filed Critical Sinai Health System
Priority to CA2977685A priority Critical patent/CA2977685C/en
Priority to EP16758390.5A priority patent/EP3265560B1/en
Priority to CN201680017382.6A priority patent/CN109121418A/en
Priority to US15/554,042 priority patent/US11261466B2/en
Priority to JP2017546726A priority patent/JP6871169B2/en
Publication of WO2016138574A1 publication Critical patent/WO2016138574A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • C12N2310/141MicroRNAs, miRNAs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/065Modulators of histone acetylation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • BRCA2 promote DNA double-strand break (DSB) repair by homologous recombination (HR) [8-10].
  • BRCAl acts in this process minimally at two discrete steps. Firstly, it promotes DNA end resection [11 , 12], the initiating step in HR that involves the nucleolytic processing of breaks to produce the single-stranded (ss) DNA necessary for homology search and strand invasion [1]. Secondly, BRCAl interacts with PALB2 [13-15] to direct the recruitment of BRCA2 [13] and RAD51 [16, 17] to DSB sites.
  • USP1 1 ubiquitin-specific peptidase 1 1
  • USPl l status or the status of other HR-proteins in tumors may provide biomarkers for use of PARP inhibitors (Wiltshire et al, JBC 285(19), 14565-14571 , 2010).
  • the present inventors have found that the cell cycle tightly controls the interaction of BRCAl with PALB2-BRCA2 in order to constrain BRCA2 function to the S/G2 phases.
  • the BRCAl -interaction site on PALB2 is targeted by an E3 ubiquitin ligase composed of KEAP1 , a PALB2-interacting protein [6], in complex with c u ll in 3 ( CUL3)-RBX1 [7].
  • PALB2 ubiquitylation suppresses its interaction with BRCAl and is counteracted by the deubiquitylase USPl l , which is itself under cell cycle control.
  • USP1 1 is regulated by a cell cycle-CULLIN4-RING-ligase (CRL4) and DCAF10 acts as an adaptor for the USPl l E3 ligase.
  • CTL4 cell cycle-CULLIN4-RING-ligase
  • DCAF10 acts as an adaptor for the USPl l E3 ligase.
  • the invention provides a method for monitoring activity of USP1 1 in a sample by assaying the interaction of BRCAl and PALB2.
  • the invention provides a method for monitoring activity of USPl l in a sample by assaying the interaction of BRCAl , PALB2, and BRCA2.
  • the invention provides a method for monitoring activity of USP1 1 in a sample by assaying the interaction of USP1 1 and PALB2.
  • the invention provides a method for monitoring activity of USP1 1 in a sample by assaying DC AF 10.
  • the invention provides a method for monitoring activity or expression of USP1 1 in a sample by assaying for complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USP1 1 and PALB2; and/or (d) USP1 1 and DCAF10.
  • the invention provides a method for monitoring activity or expression of USPl l in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USPl l and PALB2; and/or (d) USPl l and DCAF10 in the sample; (ii) measuring the levels of the complexes; and (iii) detecting an increase or decrease in the activity or expression of the complexes as compared to a control as an indication of the activity or expression of USP1 1.
  • the invention provides a method for monitoring activity or expression of USP1 1 in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USPl l and PALB2; and/or (d) USPl l and DCAF10 in the sample by immunological purification; (ii) measuring the levels of the complexes; and (iii) detecting an increase or decrease in the activity or expression of the complexes as compared to a control as an indication of the activity or expression ofUSPl l .
  • the invention provides a method for monitoring activity or expression of USP11 in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USP1 1 and PALB2; and/or (d) USP1 1 and DCAF10 in the sample; (ii) preparing peptides or peptide fragments from the isolated complexes; and (iii) subjecting the peptides or peptide fragments to mass spectrometry to thereby monitor the activity or expression of USP1 1.
  • the invention provides a method for monitoring activity or expression of USP11 in a sample by assaying ubiquitylation of PALB2, in particular ubiquitylation of the N-terminus of PALB2.
  • the invention provides a method for monitoring activity or expression of USPl l in a sample by assaying ubiquitylation of PALB2 comprising measuring the amount of polyubiquitin bound to CRL3-KEAP1 E3 ligase in the sample and detecting an increase or decrease in polyubiquitin bound to CRL3-KEAP1 E3 ligase as compared to a control as an indication of the activity or expression of USP1 1.
  • the invention provides a method for monitoring activity or expression of USP1 1 in a sample by assaying ubiquitylation of PALB2 comprising measuring the activity of CRL3-KEAP1 E3 ligase, and detecting an increase or decrease in CRL3-KEAP1 E3 ligase activity as compared to a control as an indication of the activity or expression of USP1 1.
  • the methods of the invention may be performed in the presence or absence of a test compound or agent and detection of an increase or decrease in activity or expression of one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA 1 -PALB2 -BRC A2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF complex, CRL3-KEAP1 complex, CRL3-KEAP1 -PALB2 complex, and KEAP1 - PALB2 complex, as compared to a control in the absence of the test compound or agent indicates that the test compound or agent may be useful as a therapeutic agent, or for modulating homologous recombination.
  • the invention relates to a method of identifying or evaluating an agent for its ability to sensitize cells or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on KEAP1 , CRL3- KEAP1, KEAP1-PALB2 or CRL3-KEAP1.
  • the present invention provides methods of detecting an anticancer agent comprising performing a test assay comprising contacting an immortalized cell with a test compound and assaying USPl 1 activity or expression using a method of the invention.
  • the invention also provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination comprising determining the effect of a test compound or agent on one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 -PALB2-BRCA2 complex, PALB2-USP 1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 complex and CRL3-KEAP1 - PALB2 complex.
  • the invention provides a method of screening for a therapeutic agent for treatment of a disease associated with defects in HR (i.e., HR Disease), comprising identifying an agent that disrupts or modulates one or more of USPl 1 , PALB2, PALB2 ubiquitylation, DCAF10, BRCA1 -PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAPl , USP11-DCAF10 complex, CRL3-KEAP1 or CRL3-KEAP1 -PALB2 complex.
  • HR i.e., HR Disease
  • the screening methods of the invention may further comprise conducting therapeutic profiling of the identified agents or further analogs thereof, for efficacy and toxicity in animals; optionally formulating a pharmaceutical composition including one or more agents identified as having an acceptable therapeutic profile; and optionally administering the agent to a subject or individual.
  • the invention provides methods of treating a HR Disease in an individual comprising identifying an agent that modulates HR in accordance with a method of the invention and administering the agent to the individual.
  • the invention provides a method for sensitizing cells to PARP inhibitors in an individual comprising identifying an agent that sensitizes cells to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
  • the invention provides a method for reversing or delaying emergence of resistance to PARP inhibitors in an individual comprising identifying an agent that reverses or delays emergence of resistance to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
  • the present invention provides methods of treating cancer in an individual comprising identifying an anti-cancer agent identified in accordance with a method of the invention and administering the agent to the individual.
  • the invention also provides a method for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying one or more of USPl l , DCAFI O, BRCAl, BRCA2, PALB2, KEAP1 , CRL3, CRL3-KEAP1 , USPl l -DCAFIO complex, BRCAl -PALB2-BRCA2 complex, PALB2-USP1 1 complex and CRL3-KEAP1-PALB2 complex in a sample from the subject using a method of the invention.
  • a method for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying USPl l activity or expression in a sample from the subject using a method of the invention. In an aspect, a method is provided for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying PALB2 activity or expression in a sample from the subject using a method of the invention.
  • a subject is categorized as responsive to a PARP inhibitor if there is a decrease in one or more of USPl l, DCAFIO, BRCAl , BRCA2, PALB2, KEAP1 , CRL3, USP1 1 -DCAFI O, CRL3-KEAP1, BRCAl -PALB2 and BRCAl - PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • a subject is categorized as responsive to a PARP inhibitor if there is an increase in one or more of USPl l , DCAFI O, BRCAl , BRCA2, PALB2, KEAP1 , CRL3, USP1 1 -DCAF10, CRL3-KEAP1 , BRCA1-PALB2 and BRCA1 -PALB2- BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • a method of predicting responsiveness to a PARP inhibitor may further comprise administering the PARP inhibitor to the individual.
  • the invention provides a method for treating a patient in need of treatment with a PARP inhibitor comprising (a) requesting a test providing the results of an analysis to determine if the patient is sensitive or responsive to the PARP inhibitor by detecting one or more of USP11 , DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 , USP1 1-DCAF10, CRL3, CRL3-KEAP1 , BRCA1-PALB2 and BRCA1- PALB2-BRCA2, in a sample from the subject and comparing to a control to determine if the patient is sensitive or responsive to the PARP inhibitor; and (b) administering the PARP inhibitor to the patient if the patient is sensitive or responsive to the PARP inhibitor.
  • the patient has breast cancer.
  • the patient has ovarian cancer.
  • the invention provides a method for treating a patient in need of treatment with a PARP inhibitor comprising (a) requesting a test providing the results of an analysis to determine if the patient is sensitive to the PARP inhibitor by detecting USP1 1, DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 and/or CRL3 in a sample from the subject and comparing to a control to determine if the patient is sensitive to the PARP inhibitor; and (b) administering the PARP inhibitor to the patient if the patient is sensitive to the PARP inhibitor.
  • the patient has breast cancer.
  • the patient has ovarian cancer.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USP11 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, USP1 1 -DCAF complex, KEAP1 , CRL3-KEAP1 and/or CRL3- EAP1 -PALB2 complex in a sample from the subject using a method of the invention.
  • the invention provides a method for activating or modulating (e.g., promoting) homologous recombination in a cell comprising:
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in G l phase of the cell cycle (Gl) or GO phase of the cell cycle, comprising administering, or stimulating assembly of BRCA 1 -PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell.
  • the invention also provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1 - PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell.
  • the assembly of BRCA1-PALB2 or BRCA1- PALB2-BRCA2 complexes is promoted or stimulated by administering an agent that promotes or stimulates such assembly or an agent that promotes or stimulates such assembly identified using a method of the invention.
  • the agent is USP1 1 or an agonist of USP1 1.
  • the agent is an inhibitor of CRL- KEAPl . In an embodiment, the agent is an inhibitor of KEAPl . In an embodiment, the agent is a PALB2 mutant. In an embodiment, the agent is a PALB2 mutant that disrupts its interaction with KEAPl . In an embodiment, the agent is a PALB2 comprising mutations of its Lys20, Lys25 and Lys30 residues.
  • a method for activating or modulating homologous recombination in a cell may be performed in a cell wherein single strand DNA (ssDNA) generation pathways are activated.
  • ssDNA generation pathways in the cell are activated by DNA end resection.
  • the invention also provides a method for activating or modulating homologous recombination in a cell, in particular a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell.
  • the assembly of the complexes is promoted or stimulated by administering an agent that modulates HR.
  • the agent is an agent that modulates HR identified using a method of the invention.
  • the agent is USPl l or an agonist of USPl l .
  • the agent is an inhibitor of CRL-KEAPl . In an embodiment, the agent is an inhibitor of KEAPl . In an embodiment, the agent is a PALB2 mutant. In an embodiment, the agent is an inhibitor of DCAF10. In an embodiment, the agent is an inhibitor of a CULLIN4-PJNG-ligase.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising contacting the cell with BRCA1 -PALB2 or BRCA 1 -PALB2-BRCA2 complexes.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting KEAP1 or CRL3-KEAP1 or administering an inhibitor of KEAP1 or CRL3-KEAP1.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of blocking the degradation of USP1 1 or promoting or stimulating USP1 1 activity.
  • the method comprises administering USP1 1 or an agonist thereof.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting CRL-KEAPl or administering an inhibitor of KEAP1 or CRL3-KEAP1 and blocking the degradation of USP1 1 or promoting or stimulating USP1 activity.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in Gl or GO in which DNA end resection is or has been activated generating single-stranded DNA, the method comprising (a) inhibiting KEAPl or CRL3-KEAP1 ; (b) blocking the degradation of USP1 1 or promoting or stimulating USP1 1 activity; (c) administering USP1 1 or an agonist thereof; (d) administering an inhibitor of KEAPl or CRL3 -KEAPl ; (e) administering an inhibitor of DCAF10; and/or (e) inhibiting CRL-KEAPl and blocking the degradation of USP1 1.
  • a method for activating or modulating homologous recombination in a cell may further comprise activating or promoting single-strand DNA (ssDNA) generation pathways.
  • ssDNA generation pathways are activated by DNA end resection.
  • the invention also provides a method for suppressing homologous recombination in a cell, in particular a cell in Gl , comprising suppressing the assembly of BRCA1-PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell.
  • the interaction is suppressed by administering KEAPl or CRL3 -KEAPl or an agonist thereof.
  • the interaction is suppressed by administering a USPl l antagonist/inhibitor (e.g., mitoxantrone).
  • the interaction is suppressed by administering an agent that inhibits or suppresses HR identified using a method of the invention.
  • the invention also provides a system comprising: an assay for determining the level of USP 1 1 activity, complexes or biomarker levels in a sample obtained from the subject; a processor for processing the results; computer coded instructions for comparing the results with a database; and a user display for providing the results of the comparison.
  • the database may comprise reference values for USP1 1 activity or biomarker levels.
  • the invention also contemplates the use of methods, kits, and systems of the invention in genome modification or editing.
  • the invention further contemplates the use of methods, compositions, kits, and systems of the invention in genome modification or editing, provided that said use is not a method for treatment of the human or animal body by surgery or therapy, and provided that said use is not a process for modifying the germ line genetic identity of human beings.
  • Genome modification may comprise modifying a target polynucleotide sequence in a cell, modifying expression of a polynucleotide sequence in a cell, generating a model cell comprising a mutated disease gene, or knocking out a gene.
  • a use of the invention may further comprise repairing or editing a cleaved target polynucleotide by inserting an exogenous template polynucleotide, wherein the repair or editing results in a mutation comprising an insertion, deletion, or substitution of one or more nucleotides of the target polynucleotide.
  • FIG. 1 Ubiquitylation of PALB2 prevents BRCA1-PALB2 interaction
  • a Sequence of the PALB2 N terminus and mutants.
  • SEQ ID NOs: 1 -3 b, GFP immunoprecipitation (IP) of extracts derived from Gl - or S-phase synchronized 293T cells expressing the indicated GFP-PALB2 proteins
  • c In vitro ubiquitylation of the indicated HA-tagged PALB2 proteins by CRL3-KEAP1.
  • d Pulldown assay of ubiquitylated HA-PALB2 (1-103) incubated with MBP or MBP-BRCA1 -CC. I, input; FT, flow-through; PD, pulldown.
  • the asterisk denotes a fragment of HA-PALB2 competent for BRCA1 binding
  • b-d Numbers on left indicate kDa.
  • FIG. 1 opposes the activity of CRL3-KEAP1.
  • a Normal IgG or PALB2 immunoprecipitation (IP) of extracts derived from camptothecin (CPT)- treated 293T cells of the indicated genotypes transfected with GFP— USP1 1 constructs.
  • EV empty vector; CS, C318S; WT, wild type,
  • b Clonogenic survival assays of 293T cells of the indicated genotypes treated with olaparib (mean ⁇ s.d., N > 3).
  • c Normal IgG or PALB2 immunoprecipitation of extracts derived from CPT-treated 293T cells of the indicated genotypes
  • d Immunoblots of deubiquitylation reactions containing ubiquitylated HA-tagged PALB2 (1-103) and increasing concentrations of glutathione ⁇ -transferase (GST)-USP1 1 or its C270S (CS) mutant. USP2 was used as a control.
  • DUB deubiquitylase.
  • e Cell cycle- synchronized U20S cells were irradiated (20 Gy dose) and processed for immunoblotting.
  • IR ionizing radiation
  • f Immunoblots of extracts from irradiated U20S cells transfected with the indicated siRNAs.
  • CTRL control
  • Scale bars, 5 ⁇ m. a, c, d, f Numbers to left or right indicate kDa.
  • IR ionizing radiation
  • c Schematic of the gene-targeting assay
  • HR homologous recombination; sgRNA, single guide RNA.
  • f Model of the cell- cycle regulation of homologous recombination.
  • Figure 5 Suppression of PALB2-BRCA2 accumulation at DSB sites in Gl 53 ⁇ 1 ⁇ cells, a, Schematic representation of human 53BP1 gene organization and targeting sites of sgRNAs used. Boxes indicate exons (E: yellow, coding sequence; brown, untranslated regions (UTRs)). The indels introduced by CRISPR/Cas9 and their respective frequencies are indicated, b, Wild-type (WT) and 53BP1A and U20S cells were mock- or X-irradiated (10 Gy) before being processed for 53BP1 fluorescence microscopy.
  • WT Wild-type
  • 53BP1A and U20S cells were mock- or X-irradiated (10 Gy) before being processed for 53BP1 fluorescence microscopy.
  • FIG. 7 Inhibition of the BRCA1-PALB2 interaction in Gl depends on CRL3-KEAP1.
  • a Representative micrographs of the experiment shown in Figure I d.
  • b Schematic representation of human KEAPl gene organization and targeting sites of sgRNAs used as described in Figure 5a. The indels introduced by CRISPR/Cas9 and their respective frequencies are indicated
  • c Immunoprecipitation (IP) of PALB2 from extracts prepared from irradiated 293T cells. IP with normal IgG was performed as a control, d, 293T cells with the indicated genotypes were transfected with the indicated HA-KEAP1 constructs, synchronized in Gl or S phases and irradiated.
  • IP Immunoprecipitation
  • FIG. 9 Analysis of KEAP1- and USP1 1 -dependent modulation of PALB2 and homologous recombination, a, Site-specific chemical ubiquitylation of HA-PALB2 (1-103) at residue 20 (PALB2-KC20-Ub) and 45 (PALB2-KC45-Ub) was carried out by dichloroacetone linking. The resulting ubiquitylated PALB2 polypeptides along with their unmodified counterparts were subjected to pulldown with a fusion of MBP with the coiled-coil domain of BRCA1 (MBP-BRCA1 -CC). I, input; PD, pulldown.
  • b Wild-type and ⁇ 293T cells were treated with cycloheximide (CHX) for the indicated time and then processed for NRF2 and KEAP1 immunoblotting. Actin levels were also determined as a loading control
  • c Immunoprecipitation (IP) of USP1 1 from extracts prepared from 293T cells that were or were not treated with camptothecin (CPT; 200 nM). Immunoprecipitation with normal IgG was performed as a control
  • d U20S DR-GFP cells were transfected with the indicated siRNAs.
  • CRL non-targeting
  • FIG. 11 Characterization of USPl l protein stability a, U20S cells synchronized in Gl or S/G2 were treated with cyclohexamide (CHX) and processed at the indicated time points to monitor USPl l stability, b, Immunoprecipitation (IP) of PALB2 from extracts prepared from 293T cells that were synchronized in Gl or S phase and treated or not with IR (20 Gy). c, U20S cells were irradiated with a dose of 2 or 20 Gy and processed for USPl l immunoblotting at the indicated times post-IR.
  • CHX cyclohexamide
  • IP Immunoprecipitation
  • Actin was used as a loading control
  • ATMi ATM inhibitor KU55933
  • ATRi ATR inhibitor VE-821
  • DNAPKcs inhibitor NU7441 DNAPKcs inhibitor NU7441
  • FIG. 12 Reactivation of RAD51 loading and unscheduled DNA synthesis in Gl .
  • a 53BP1A U20S cells were transfected with the indicated siRNA, synchronized in Gl or S/G2 by release from a double-thymidine block and irradiated (20 Gy) before being processed for fluorescence microscopy.
  • DAPI was used to trace the nuclear boundary and cyclin A staining was used to determine cell cycle position.
  • FIG. 13 Analysis of homologous recombination in Gl .
  • a Quantitation of gene targeting efficiency at the LMNA locus in asynchronously dividing U20S cells transfected with increasing amount of donor template and with (grey) or without (white) sgRNAs. Gene-targeting events were detected by flow cytometry (mean ⁇ s.d., N > 3).
  • FIG. 14 Identification of DCAF10 as a regulator of USP1 1 stability in response to DNA damage, a. siRNA screen where U20S cells were transfected with siRNAs targeting known and predicted DCAFs along with other CUL4-interacting proteins. Cells were either irradiated with IR (20 Gy) or UV (50 J/m-2), let to recover for 3 h and then processed for USP1 1 immunofluorescence. Each point plotted corresponds to the percentage of USP1 1 left after irradiation. The red dots correspond to the siRNA non-targeting controls (CTRL) and targeting USP11, whereas the red dots correspond to core CRL4 factors, that include CUL4 itself. D.U20S cells were transfected with the indicated siRNAs and then irradiated with a dose of 20 Gy and processed for USP1 1 immunoblotting at the indicated times post-ionizing radiation. Actin was used as a loading control.
  • CRL siRNA non-targeting controls
  • FIG. 15 Validation of DCAF10 as a regulator of USP1 1.
  • DCAF10 interacts with USP1 1.
  • Whole cell extracts of mouse embryo fibroblasts (MEFs) of the indicated genotypes were processed for USP1 1 immunoblotting. Tubulin was used as a loading control
  • U20S DR-GFP cells were transfected with the indicated siRNAs or expression vectors. Twenty-four hours post-transfection, cells were transfected with an I-Scel expression vector. The percentage of GFP -positive cells was determined 48 h post-plasmid transfection for each condition and was normalized to the I-Scel plus non- targeting (CTRL) + empty vector (EV) condition.
  • FIG. 16 KEAP1 inhibition can activate HR in Gl cells.
  • Gene targeting at the LMNA locus in Gl -arrested cells transfected with the indicated siRNA and vectors expressing either the Rl KEAP1 inhibitor or its FN3 scaffold control (mean ⁇ s.d., N 3).
  • a “CRISPR system” generally refers to transcripts and other elements involved in the expression of, or directing the activity of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated (“Cas”) genes.
  • a CRISPR system may include without limitation, sequences encoding a Cas gene, a tracr (trans- activating CRISPR) sequence (e.g. tracrRNA or an active partial tracrRNA), a tracr- mate sequence, a guide sequence, or other sequences and transcripts from a CRISPR locus.
  • One or more elements of a CRISPR system may be derived from a type I, type II, or type III CRISPR system.
  • a CRISPR system promotes the formation of a CRISPR complex (comprising a guide sequence hybridized to a target sequence and complexed with one or more Cas proteins) at the site of a target sequence.
  • a "target sequence” or “target polynucleotide” refers to a sequence which is sufficiently complementary to a designed guide sequence that the target sequence hybridizes to the guide sequence promoting the formation of a CRISPR complex.
  • a target sequence may comprise any polynucleotide, such as DNA or RNA polynucleotides, and it may be located in the nucleus, cytoplasm, or an organelle, for example, mitochondria or chloroplast.
  • formation of a CRISPR complex in an endogenous CRISPR system results in cleavage of one or both strands in or near (e.g. within 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, or more base pairs from) the target sequence.
  • CRISPR systems also include the systems developed by or available from Editas Medicine (Cambridge, MA), Caribou Biosciences (Berkeley, CA), CRIPSR Therapeutics (Basel, Switzerland), Addgene (Cambridge, MA) and Intellia Therapeutics (Cambridge, MA).
  • DNA end resection generally refers to nucleolytic degradation of the 5'-terminated strand of a DNA double- stranded break leading to the formation of 3'- terminated single- stranded DNA.
  • DNA end resection in eukaryotes comprises two phases: a slow initial phase, catalyzed by the Mrel l-Rad50-Nbsl (MRN) complex in mammals, and a second and faster phase catalyzed by the exonuclease Exol or the helicase Bloom Syndrome Protein (BLM).
  • MRN Mrel l-Rad50-Nbsl
  • BBM helicase Bloom Syndrome Protein
  • Pathways involved in DNA end resection may be activated by stimulating or activating BRCA1 recruitment to DNA double- strand breaks by inhibiting TP53BP1 (53BP1) or RIF, or blocking recruitment of 53BP1 or RIF to DNA double-stranded break sites.
  • DNA end resection may be activated by inhibiting 53BP1 (or RIF) expression and/or activity and expressing a mutated form of CtIP that mimics constitutive phosphorylation, for example CtIP-Thr879Glu.
  • DNA end resection is reconstituted or activated using inhibitors of 53BP1 and a mutated form of CtIP that mimics constitutive phosphorylation, in particular CtIP-Thr879Glu.
  • DNA end resection may be reconstituted or activated using purified human proteins: Bloom helicase (BLM); DNA2 helicase/nuclease; Exonuclease 1 (EXOl ); the complex comprising MRE11 , RAD50, and NBSl (MRN); and Replication protein A (RPA.)
  • BBM Bloom helicase
  • EXOl Exonuclease 1
  • MRN NBSl
  • RPA Replication protein A
  • “Homologous recombination” and “HR” refer to a type of genetic recombination in which DNA strands of similar or identical nucleotide sequences are exchanged.
  • HR can be used by cells to repair DNA double-strand breaks (DSB) by the following general steps. HR is initiated when the DSB is resected by nucleases and helicases, generating 3' single-stranded DNA (ssDNA) overhangs onto which the RAD51 recombinase assembles as a nucleoprotein filament. This structure can invade homologous duplex DNA, which is used as a template for repair DNA synthesis.
  • ssDNA 3' single-stranded DNA
  • the resulting intermediates can be metabolized to yield non-crossover products thereby restoring the damaged DNA molecule as it existed before the double-strand break (San Filippo et al., Annu. Rev. Biochem. 2008. 77:229-57).
  • the terms also include recombination using single-stranded donor oligonucleotides (ssODNs), in particular recombination using single-stranded donor oligonucleotides (ssODNs) requiring resection and which may be activated by 53BP1 inhibitors.
  • HR Disease refers to any disorder, disease, condition, syndrome or combination of manifestations or symptoms recognized or diagnosed as a disorder which may be associated with or characterized by a HR defect.
  • exemplary diseases include, for example, cancer, cardiovascular diseases including heart failure, hypertension and atherosclerosis, respiratory diseases, renal diseases, gastrointestinal diseases including inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, hepatic, gallbladder and bile duct diseases, including hepatitis and cirrhosis, hematologic diseases, metabolic diseases, endocrine and reproductive diseases, including diabetes, bone and bone mineral metabolism diseases, immune system diseases including autoimmune diseases such as rheumatoid arthritis, lupus erythematosus, and other autoimmune diseases, musculoskeletal and connective tissue diseases, including arthritis, achondroplasia infectious diseases and neurological diseases such as Alzheimer's disease, Huntington's disease and Parkinson's disease.
  • Methods of the invention may be used to monitor or treat a disease caused by a defect in a gene that mediates homologous recombination, for example, BRCA1 , BRCA2, PALB2, PARP-1 , USP11 , RAD51 , and/or DCAF10.
  • the invention provides for monitoring or treatment of cancer with BRCA-1 defects, BRCA-2 defects, dual BRCA-l/BRCA-2 defects, and Fanconi anemia.
  • the cancer is breast cancer, in particular invasive ductal carcinoma and invasive lobular carcinoma.
  • the cancer is ovarian cancer, in particular epithelial ovarian tumors, germ cell ovarian tumors, and sex cord stromal tumors.
  • Methods of the invention for activating or modulating homologous recombination may be used to genetically modify polynucleotides associated with a genetic disorder.
  • the genetic disorder is a monogenetic disorder.
  • the genetic disorder is a multigenetic disorder.
  • the genetic disorder is associated with one or more SNPs.
  • the genomic modification corrects a point mutation.
  • Examples of genetic disorders and polynucleotide sequences associated with the genetic disorders may be found on the World Wide Web (see for example, the National Center for Biotechnology Information, National Library of Medicine (Bethesda, MA) or the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, Md)), listed in published patents and applications (see, for example, US Published Application No. 2015/0247150), and in publications (see for example, Turitz Cox D.B. et al, Nature Medicine 21 , 121 -131 , 2015; and O'Connor T.P. and R.G. Crystal, Nature Reviews/Genetics Volume 7, April 2006, pages 261 -276 including Supplementary Information, and publications cited therein).
  • the genetic disorder is Wilson disease which depends on mutations in the gene encoding the ATP7B Cu translocase, a protein mainly expressed by the hepatocyte that regulates the levels of copper in the liver.
  • the genetic disorder is a genetic disorder of the lungs.
  • the genetic disorder is cystic fibrosis, an autosomal recessive disease caused by mutations of the Cystic Fibrosis Transmembrane Regulator (CFTR) protein, a member of the ATP -binding cassette superfamily of transmembrane proteins.
  • CFTR Cystic Fibrosis Transmembrane Regulator
  • the genetic disorder is sickle cell anemia and a method of the invention comprises correcting the mutated HBB hemoglobin gene by gene conversion with its paralog HBD.
  • PARP Inhibitor refers to an inhibitor of the nuclear enzyme poly(adenosine 5'-diphospho-ribose) polymerase ["poly(ADP-ribose) polymerase” or “PARP”, which is also referred to as ADPRT (NAD:protein (ADP-ribosyl transferase (polymerising)) and PARS (poly(ADP-ribose) synthetase).
  • ADPRT NAD:protein (ADP-ribosyl transferase (polymerising)
  • PARS poly(ADP-ribose) synthetase
  • the PARP inhibitor is Olaparib
  • the PARP inhibitor is Veliparib (AbbVie Inc, Chicago, IL). In aspects of the invention, the PARP inhibitor is Rucaparib (Clovis Oncology, Inc., Boulder, CO). In aspects of the invention, the PARP inhibitor is ⁇ - 1001 (Inotek Pharmaceuticals Corp, Lexington, MA). In aspects of the invention, the PARP inhibitor is MK-4827 (niraparib) (Tesaro, Waltham, MA, also see Montoni et al, Frontiers in Pharmacology, [4], Article 18, pages 1 -7). In aspects of the invention, the PARP inhibitor is talazoparib (Medivation, Inc, San Francisco CA).
  • sample is a sample derived from any biological source, such as tissues, extracts, or cell cultures, including cells (e.g. tumor cells), cell lines, cell lysates, and physiological fluids, such as, for example, blood or subpopulations thereof (e.g. white blood cells, erythrocytes), plasma, serum, saliva, ocular lens fluid, cerebrospinal fluid, sweat, urine, fecal matter, tears, bronchial lavage, swabbings, milk, ascites fluid, nipple aspirate, needle aspirate, synovial fluid, peritoneal fluid, lavage fluid, and the like.
  • the sample can be obtained from animals, preferably mammals, most preferably humans.
  • Samples can be from a single individual or pooled prior to analysis.
  • the sample can be treated prior to use, such as preparing plasma from blood, diluting viscous fluids, and the like.
  • Methods of treating samples can involve filtration, distillation, extraction, centrifugation, concentration, inactivation of interfering components, the addition of reagents, and the like.
  • the sample is a mammalian tissue sample.
  • the sample is a cell lysate.
  • the sample is a cell.
  • the sample is a human physiological fluid.
  • the sample is human serum.
  • the sample is white blood cells or erythrocytes.
  • the term also includes animals bred for food, as pets, or for study including horses, cows, sheep, poultry, fish, pigs, cats, dogs, and zoo animals goats, apes (e.g. gorilla or chimpanzee), and rodents such as rats and mice.
  • animals bred for food, as pets, or for study including horses, cows, sheep, poultry, fish, pigs, cats, dogs, and zoo animals goats, apes (e.g. gorilla or chimpanzee), and rodents such as rats and mice.
  • NCBI Accession Numbers for USPl l , PALB2, BRCA1 , BRCA2, KEAP1 , 53BP1 , DCAFIO, RBX1 , CUL3 and CtIP are in Table 1 and the human sequences for same are in the Sequence Listing.
  • the invention provides a method for monitoring activity or expression of USPl l by assaying the interaction of BRCA1 and PALB2, the interaction of BRCA1 , PALB2 and BRCA2, the interaction of USPl l and DCAFIO, and/or the interaction of USP11 and PALB2.
  • Routine methods known to persons skilled in the art can be used to assay protein interactions in a sample.
  • BRCA1-PALB2, BRCA1-PALB2- BRCA2, USPl l-DCAFlO, or USP11 -PALB2 complexes may be isolated using affinity techniques such as for example immunologically-based purification (e.g.
  • peptides may be prepared from the isolated complexes using conventional methods (e.g. gel electrophoresis, liquid chromatography, capillary electrophoresis, nano-reversed phase liquid chromatography, high performance liquid chromatography, or reverse phase high perfonnance liquid chromatography), and the peptides or peptide fragments may be subjected to mass spectrometry (e.g., quantitative mass spectrometry such as selected reaction monitoring mass spectrometry (sMRM), high resolution data independent analyses (SWATH), high resolution multiple reaction monitoring (MRM HR ) or MSI based quantitation).
  • mass spectrometry e.g., quantitative mass spectrometry such as selected reaction monitoring mass spectrometry (sMRM), high resolution data independent analyses (SWATH), high resolution multiple reaction monitoring (MRM HR ) or MSI based quantitation.
  • the invention also provides a method for monitoring activity of USPl l by assaying ubiquitylation of the N-terminus of PALB2.
  • Routine methods known to persons skilled in the art can be used to assay ubiquitination in a sample.
  • ubiquitination or PALB2 may be assayed by measuring changes in PALB2 (e.g., weight; see US Patent No. 6,413,725), the amount of poly-ubiquitin bound to CRL3- KEAP1 E3 ligase (see for example, EP 1268847), and/or the activity of CRL3-KEAP1 E3 ligase (see for example, US Publication No. 2013/01 16152).
  • the invention provides a method for assaying ubiquitylation of PALB2 polypeptides in a sample, the method comprising digesting ubiquitinated PALB2 polypeptides in the sample with a protease, thereby generating a plurality of test peptides; determining the presence of at least one isopeptide bond between ubiquitin and a lysine residue of the test peptides by mass spectrometry to determine the numbers of ubiquitination sites and thereby the amount of ubiquitination of PALB2 polypeptides in the sample.
  • the test peptides are from the PALB2 N- terminus.
  • the lysine residue corresponds to Lysl4, Lysl6, Lys20, Lys25, Lys30, Lys43 or Lys45.
  • the method may utilize peptide internal standards corresponding to different peptide subsequences of PALB2 to provide for controls in a quantitative assay.
  • different synthetic peptide internal standards corresponding to PALB2 are generated and differentially labeled.
  • Proximity ligation assays may also be used to assay activity of USP1 1 by assaying the interaction between BRCA1 and PALB2 and/or PALB2- interacting proteins such as BRCA2, using DNA-based detection.
  • primary antibodies against binding partners of an interaction e.g., PALB2 and BRCA1
  • a second set of antibodies termed PLA probes or proximity probes, recognize the first set of primary antibodies.
  • the PLA probes contain DNA strands that assemble into an assay-specific DNA molecule when in close proximity. This DNA molecule can then be amplified and detected using, for example, fluorescent probes. [See, for example, Soderberg O. et al., Nat. Methods., 2006 December; 3(12):995-1000; Jarvius M. et al., Mol. Cell. Proteomics, 2007 September; 6(9): 1500- 9)] ⁇
  • the invention provides a method for assaying BRCA1- PALB2 or BRCA1-PALB2-BRCA2 interactions in a sample comprising: contacting the sample with primary antibodies to each binding partner in the interaction; contacting the sample with proximity probes comprising a secondary antibody that binds to a corresponding primary antibody, wherein each proximity probe has an oligonucleotide conjugated thereto; wherein when the oligonucleotides of the proximity probes are in sufficient proximity to each other, the oligonucleotides of the proximity probes interact to form circular products that are amplified by rolling circle replication producing amplification products; and, measuring the amplification products to thereby assay or measure the interactions.
  • Assays that monitor PALB2 (or associated proteins such as BRCA2) in situ co-localization with BRCA1 can also provide a method for monitoring USP1 1 activity (see Example herein). For example, PALB2 localization at sites of DNA damage (marked by BRCA1 or other markers such as ⁇ - ⁇ 2 ⁇ ) is dependent on USP1 1 activity.
  • cells are fixed, permeabilized and then incubated with antibodies that detect PALB2, BRCA2 or their associated proteins (e.g., BRCA1).
  • Addition of labeled secondary antibodies enable the in situ visualization of protein accumulation at DNA damage sites in subnuclear structures termed foci.
  • Addition of a genotoxic insult such as ionizing radiation or other clastogenic treatments) increases the number of "foci" and can be included to augment the dynamic range of the assay.
  • proximity ligation assays and in situ co- localization assays may be used to assay any of the interactions disclosed herein.
  • the methods of the invention may be performed in the presence or absence of a test compound or agent and detection of an increase or decrease in activity or expression of one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAFlO complex, CRL3-KEAP1 complex, CRL3-KEAP1-PALB2 complex, and KEAP1- PALB2 complex, as compared to a control in the absence of the test compound or agent indicates that the test compound or agent may be useful as a therapeutic agent, or for modulating homologous recombination.
  • the invention also relates to a method of identifying or evaluating an agent for its ability to sensitize cells or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on KEAP1 , CRL3-KEAP1 , or KEAP1-PALB2.
  • a negative effect on KEAP1 indicates that the agent is a poor sensitizer of cells to PARP inhibitors.
  • a positive effect on KEAP1 or CRL3-KEAP1 activity indicates that the agent is a sensitizer of cells to PARP inhibitors or can reverse or delay emergence of resistance to PARP inhibitors.
  • the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by increased levels of KEAP1 , CRL3-KEAP1 , KEAP1-PALB2 or CRL3-KEAP1 activity or expression when compared to such levels obtained from a control. In an aspect, the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by decreased levels of KEAP1 , CRL3-KEAP1 , KEAP1-PALB2 or CRL3- KEAP1 activity or expression when compared to such levels obtained from a control.
  • the present invention contemplates methods of detecting an anti-cancer agent comprising performing a test assay comprising contacting an immortalized cell with a test compound and measuring USP11 activity or CRL3- KEAP1 using a method of the invention and comparing to a control test assay in the absence of the test compound.
  • USP1 1 activity is assayed by measuring USP1 1 , PALB2, DCAF10, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP11-DCAF10 complex, CRL3-KEAP1 and/or CRL3-KEAP1 -PALB2 complex in the cell.
  • detecting a negative effect of the agent on USP11 activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer.
  • detecting a negative effect of the agent on BRCA1-PALB2-BRCA2 complex and/or PALB2- USP1 1 complex activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer.
  • detecting a positive effect of the agent on KEAP1 , CRL3-KEAP1 and/or CRL3 -KEAP 1 -P ALB2 complex activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer.
  • decreased levels of USP11 activity when compared to such levels obtained from a control are indicative that agent has anticancer activity or is a PARP inhibitor sensitizer.
  • the invention provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination comprising determining the effect of a test compound or agent on one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 -PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 complex and CRL3-KEAP1-PALB2 complex.
  • the invention provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination in a cell comprising (i) assaying in a sample USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 - PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 and/or CRL3-KEAP1 -PALB2 complex in the cell in the presence or absence of the agent, and (ii) detecting an increase or decrease in USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2- USP1 1 complex, KEAP1, USP11 -DCAF10 complex, CRL3-KEAP1 complex and/or CRL3-KEAP1 -PALB2 complex in the sample compared to a control as an indication of the ability of the agent
  • Test compounds used in the methods of the invention can be any product in isolated form or in a mixture.
  • the test compound may be defined by structure or function or it may be undefined. Examples of undefined test compounds include without limitation tissue samples, biological fluids, cell supernatants, vegetal preparations; etc.
  • Test compounds may be peptides such as soluble peptides including Ig-tailed fusion peptides, members of random peptide libraries and combinatorial chemistry-derived molecular libraries made of D- and/or L-configuration amino acids, carbohydrates, nucleic acids, antisense molecules, phosphopeptides (including members of random or partially degenerate, directed phosphopeptide libraries), antibodies [e.g.
  • test compound may be an endogenous physiological compound or natural or synthetic compounds.
  • the methods of the present invention for identifying agents, in particular anti-cancer agents comprises the step of contacting a cell in the presence of a test compound.
  • the cells can then be observed to determine if the test compound(s) effects USPl l activity, DCAFI O, PALB2, ubiquitination of PALB2, KEAPl , CRL-KEAPl activity, the interaction of BRCAl and PALB2, the interaction of USP1 1 and PALB2, the interaction of USP1 1 and DCAFI O, and/or the interaction of BRCAl , PALB2 and BRCA2.
  • Positive and negative controls may be performed in which known amounts of test compound and no compound, respectively, are added to the assay.
  • One skilled in the art can select and perform the appropriate controls.
  • the activity of a test compound(s) may be unknown, and the methods of the invention may be used to identify compounds exhibiting the selected property (e.g., PARP inhibitor sensitizer).
  • the activity or type of activity of the test compound(s) is known or expected, and the methods of the invention can be used to further characterize or optimize the activity (e.g., specificity, efficacy, etc).
  • a method of the invention may also comprise assaying PARP activity in the presence of the test compound.
  • PARP activity may be assayed by measuring a change of poly(ADP-ribose) polymers (PAR), and measuring NAD levels and/or ATP levels using methods routine to one of ordinary skill in the art.
  • the levels of NAD are depleted in the presence of the test compound.
  • the levels of ATP are depleted in the presence of the test compound.
  • the levels of NAD are increased in the presence of the test compound.
  • the levels of ATP are increased in the presence of the test compound.
  • a method of the invention may comprise the step of determining if a cell has undergone necrosis following administration of a test compound.
  • the physical characteristics of the cell can be analyzed using routine methods known to those skilled in the art to determine if a cell has undergone necrosis. For example, necrosis may be determined by measuring organelle swelling, plasma membrane disintegration, intracellular vacuole formation, and nuclear degradation without condensation.
  • the screening methods of the invention may further comprise conducting therapeutic profiling of the identified agents or further analogs thereof, for efficacy and toxicity in animals; optionally formulating a pharmaceutical composition including one or more agents identified as having an acceptable therapeutic profile; and optionally administering the agent to a subject or individual.
  • the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USPl l , DCAF10, BRCAl, BRCA2, PALB, KEAPl and CRL3 in a sample from the subject using a method of the invention.
  • significantly different levels of USP1 1 , DCAF10, BRCAl , BRCA2, PALB, KEAPl and CRL3 compared to a control indicate responsiveness (e.g., sensitivity) to the PARP inhibitor.
  • a subject is categorized as responsive to a PARP inhibitor if there is a decrease in one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl, CRL3, CRL3 -KEAPl, BRCAl -PALB 2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • a subject is categorized as responsive to a PARP inhibitor if there is an increase in one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3 -KEAPl , BRCA1- PALB2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • significantly different levels e.g., lower levels
  • USPl l activity compared to a control indicate sensitivity to the PARP inhibitor.
  • the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3 and CRL3- KEAPl activity or expression or PALB2 ubiquitylation in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
  • the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USP1 1 , BRCAl , BRCA2, PALB2 and KEAPl activity or expression in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
  • the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl and CRL3 activity in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
  • the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising assaying USP1 1 activity or expression in a sample from the individual using a method of the invention.
  • significantly different levels (e.g., lower levels) of USP1 1 activity or expression compared to a control indicate sensitivity to the PARP inhibitor.
  • the invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting CRL3-KEAP1 activity in a sample from the individual and comparing to a control to determine if the individual will be sensitive to a PARP inhibitor.
  • significantly different levels (e.g., higher levels) of CRL3-KEAP1 compared to a control indicate sensitivity to the PARP inhibitor.
  • the invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting PALB2 ubiquitylation in a sample from the individual and comparing to a control to determine if the individual will be sensitive to a PARP inhibitor.
  • significantly different levels of PALB2 ubiquitylation compared to a control indicate sensitivity to the PARP inhibitor.
  • the invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting complexes of BRCA1 , PALB2 and BRCA2 in a sample from the subject and comparing to a control to determine if the individual will be responsive (e.g., sensitive) to a PARP inhibitor.
  • significantly different levels (e.g., absence or low levels) of complexes of BRCA1 , PALB2 and BRCA2 indicate sensitivity to the PARP inhibitor.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USPl l , DCAFIO, PALB2, PALB2 ubiquitylation, BRCAl -PALB2-BRCA2 complex, PALB2-USP1 1 complex, USP1 1 -DCAF10 complex, KEAPl , CRL3-KEAP1 or CRL3-KEAP1 -PALB2 complex in a sample from the individual using a method of the invention.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USPl l activity in a sample from the individual using a method of the invention.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying CRL3-KEAP activity in a sample from the individual.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating USPl l , BRCAl , BRCA2, PALB2 and KEAPl in a sample from the individual.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating USPl l , DCAFIO, BRCAl , BRCA2, PALB2 and KEAPl in a sample from the individual.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating BRCAl -PALB2-BRCA2 complex in a sample from the individual.
  • the invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating PALB2 ubiquitylation in a sample from the individual.
  • an individual is assigned to a category for a clinical trial for a PARP inhibitor based on a decrease in one or more of USPl l , DCAFIO, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3 -KEAPl , BRCAl -PALB2 and BRCAl - PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • an individual is assigned to a category for a clinical trial for a PARP inhibitor based on an increase in one or more of USPl l , DCAFIO, BRCAl , BRCA2, PALB2, KEAPl, CRL3, CRL3-KEAP1 , BRCA1-PALB2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
  • a variety of routine methods known to a person skilled in the art can be employed for detecting or assaying the biomarkers USP1 1 , DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 , CRL3 and/or complexes thereof in a sample.
  • Biomarker levels present in a sample may be determined by any suitable assay, which may comprise the use of any of the group comprising or consisting of immunoassays, spectrometry, mass spectrometry, Matrix Assisted Laser Desorption/Ionization Time-of-Flight (MALDI- TOF) Mass Spectrometry, microscopy, northern blot, isoelectric focussing, SDS- PAGE, PCR, quantitative RT-PCR, gel electrophoresis, DNA microarray, and antibody microarray, or combinations thereof.
  • any suitable assay may comprise the use of any of the group comprising or consisting of immunoassays, spectrometry, mass spectrometry, Matrix Assisted Laser Desorption/Ionization Time-of-Flight (MALDI- TOF) Mass Spectrometry, microscopy, northern blot, isoelectric focussing, SDS- PAGE, PCR, quantitative RT-PCR, gel electrophoresis, DNA microarray, and antibody micro
  • the invention also provides a system comprising: an assay for determining the level of USP11 activity, complexes or biomarker levels in a sample obtained from the subject; a processor for processing the results; computer coded instructions for comparing the results with a database; and a user display for providing the results of the comparison.
  • the database may comprise reference values for USP1 1 activity or biomarker levels.
  • a method of the invention for predicting or characterizing responsiveness to a PARP inhibitor may further comprise administering the PARP inhibitor to the individual or subject.
  • the invention relates to a method of treating a subject with a PARP inhibitor comprising:
  • the invention further provides a method for treating cancer in a subject the method comprising: (i) selecting a subject who is responsive to a PARP inhibitor using a method of the invention, and (ii) administering to said subject the PARP inhibitor in an effective amount to treat the cancer.
  • the cancer is breast cancer. In an embodiment, the cancer is ovarian cancer.
  • Agents identified using the methods of the present invention have numerous therapeutic applications related to, for example, cancer, ischemia reperfusion injury, inflammatory diseases, degenerative diseases, protection from adverse effects of cytotoxic compounds, and potentiation of cytotoxic cancer therapy.
  • Agents identified using the methods of the invention may be used to potentiate radiation and chemotherapy by increasing apoptosis of cancer cells, limiting tumor growth, decreasing metastasis, and prolonging the survival of tumor-bearing subjects.
  • the agents can be used to treat leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer and cervical carcinomas.
  • the agents can be used to treat, without limitation, retroviral infection, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, uveitis, diabetes, Parkinson's disease, myocardial infarction, stroke, other neural trauma, organ transplantation, reperfusion of the eye, reperfusion of the kidney, reperfusion of the gut, reperfusion of skeletal muscle, liver toxicity following acetaminophen overdose, cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents, and skin damage secondary to sulfur mustards.
  • the invention provides a method for sensitizing an individual to treatment with PARP inhibitors comprising identifying an agent that sensitizes cells to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
  • the invention provides a method for treating an individual being treated with a PARP inhibitor comprising administering to the individual an agent that sensitizes cells to the PARP inhibitor identified using a method of the invention.
  • the invention provides a method for reversing or delaying emergence of resistance to PARP inhibitors in an individual comprising identifying an agent that reverses or delays emergence of resistance to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
  • the present invention provides methods of treating cancer in an individual comprising identifying an anti-cancer agent in accordance with a method of the invention and administering the agent to the individual.
  • the present invention provides a method of treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer or cervical carcinomas in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of an agent identified using a method of the invention or a therapeutically acceptable salt thereof.
  • the present invention provides a method of potentiation of cytotoxic cancer therapy in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of an agent identified using a method of the invention that potentiates cytotoxic cancer therapy or a therapeutically acceptable salt thereof.
  • the invention provides methods of treating a disease associated with defects in HR (i.e., HR Disease) in an individual comprising identifying an agent that modulates HR in accordance with a method of the invention and administering the agent to the individual.
  • a disease associated with defects in HR i.e., HR Disease
  • the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer or cervical carcinomas in a mammal in need of such treatment.
  • the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for potentiation of cytotoxic cancer therapy in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of the agent.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an agent identified using a method of the invention, or a therapeutically acceptable salt thereof, in combination with a therapeutically acceptable carrier.
  • Pharmaceutical compositions can be prepared by per se known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.
  • Suitable carriers are described, for example, in Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., USA 1985).
  • the compositions include, albeit not exclusively, solutions of the active agents in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
  • compositions are indicated as therapeutic agents either alone or in conjunction with other therapeutic agents or other forms of treatment.
  • compositions of the invention may be administered concurrently, separately, or sequentially with other therapeutic agents or therapies, for example PARP inhibitors.
  • the invention provides a method for activating or modulating homologous recombination in a cell comprising:
  • the cell is in the Gl phase of the cell cycle (Gl). In an aspect the cell is a non-dividing cell or a causing cell in Gl . In an aspect, the cell is in the GO phase of the cell cycle (GO). In an aspect, the methods of the invention are used in vitro to activate or modulate homologous recombination in cells.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in the Gl phase of the cell cycle (Gl ) or GO phase of the cell cycle (GO), comprising (a) promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell; and/or (b) contacting the cell with BRCA1 -PALB2 or BRCA1 - PALB2-BRCA2 complexes.
  • the assembly of BRCA1-PALB2 or BRCA1 -PALB2- BRCA2 complexes is promoted or stimulated by administering an agent that promotes or stimulates BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes identified using a method of the invention.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting KEAP1 or CRL3-KEAP1.
  • the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of blocking the degradation of USP1 1 or promoting USP1 1 activity.
  • the method comprises administering USP1 1 or an agonist thereof.
  • Methods of the invention may be performed in a cell, in particular a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA.
  • Gl Gl phase of the cell cycle
  • GO GO phase of the cell cycle
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1- PALB2 or BRC A 1 -PALB2-BRC A2 complexes in the cell.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell.
  • the assembly of the complexes is promoted or stimulated by administering an agent that promotes or stimulates the assembly or occurrence of BRCA1-PALB2 or BRCA1-PALB2-BRCA2 complexes identified using a method of the invention.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising contacting the cell with BRCA1 -PALB2 or BRC A 1 -P ALB2-BRC A2 complexes.
  • the invention also provides a method for repairing DNA double-strand breaks in a cell in Gl or GO in which DNA end resection is or has been activated generating single-stranded DNA, the method comprising (a) inhibiting KEAP1 and/or CRL3-KEAP1 ; (b) blocking the degradation of USPl l or promoting or stimulating USPl l activity; (c) administering USPl l or an agonist thereof; and/or (d) inhibiting CRL3-KEAP1 and blocking the degradation of USP1 1.
  • a method for activating homologous recombination in a cell may further comprise activating or promoting single strand DNA (ssDNA) generation pathways.
  • ssDNA generation pathways are activated by DNA end resection.
  • a method for activating homologous recombination in a cell further comprises activating DNA end resection.
  • DNA end resection is activated or promoted by inhibiting 53BP1 (or RIF1) expression or activity (e.g., recruitment of 53BP1 to DSB sites) and/or upregulating or expressing CtlP.
  • DNA end resection is activated or promoted by inhibiting 53BP1 (or RIF1 ) expression or activity (e.g., recruitment of 53BP1 to DSB sites) and upregulating or expressing CtlP.
  • the method involves inhibiting 53BP1 using antagonists, including without limitation short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs) or an inhibitor of the histone deacetylase (HDAC) family of enzymes (for example, trichostatin A), and using an analog of CtlP that mimics constitutive phosphorylation, for example CtIP-Thr879Glu.
  • antagonists including without limitation short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs) or an inhibitor of the histone deacetylase (HDAC) family of enzymes (for example, trichostatin A), and using an analog of CtlP that mimics constitutive phosphorylation, for example CtIP-Thr879Glu.
  • a method for activating homologous recombination in a cell may comprise activating BRCA1 recruitment to DNA double-strand break (DSB) sites.
  • BRCA1 recruitment is activated by inhibiting expression of 53BP1 (TP53BP1 ) or RIF1 , or inhibiting the recruitment of 53BP1 or RIF1 to DSB sites.
  • 53BP1 or RIF1 may be inhibited using antagonists, including without limitation short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs).
  • a method for activating or stimulating homologous recombination in a cell further comprises a gene editing system.
  • the gene editing system comprises contacting the cell with a nuclease.
  • nucleases include without limitation, zinc finger nucleases (ZFNs), engineered meganucleases, transcription activator like effector nucleases (TALENS), mega or homing endonucleases, clustered regularly interspaced short palindromic repeats (CRISPR)- associated (Cas) protein, Ttago nucleases, and fusions between nucleases, such as mega-TALs and compact TALENs.
  • the gene editing system comprises contacting the cell with a clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of a target polynucleotide sequence associated with a genetic disorder, wherein the target polynucleotide sequence is cleaved.
  • CRISPR regularly interspaced short palindromic repeats
  • the invention provides a method for altering a genetic disorder associated with a target polynucleotide sequence in a cell comprising: (1) contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 or agents that maintain the BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions throughout the cell cycle; and (2) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved.
  • the method may reduce expression of the target polynucleotide sequence, knock out the target polynucleotide
  • the invention provides a method for altering a genetic disorder associated with a target polynucleotide sequence in a cell comprising: (1) contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises a kit, vector(s) or composition of the invention, in particular the system comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation, preferably the system comprises a KEAP1 inhibitor, an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP- Thr879Glu; and (2) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein
  • a method for treating or preventing a genetic disorder in a subject comprising (a) altering a target polynucleotide sequence associated with the genetic disorder in a cell by contacting the cell with a system which activates homologous recombination in the cell
  • the system comprises a kit, vector(s) or composition of the invention, in particular the system comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation, preferably the system comprises a KEAP1 inhibitor, an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu; and (b) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids
  • a method for treating or preventing a genetic disorder may comprise introducing the cell into the subject, thereby treating or preventing the genetic disorder associated with the target polynucleotide sequence.
  • the method may comprise repairing the cleaved target polynucleotide sequence by inserting an exogenous template polynucleotide, wherein said repair results in a mutation comprising an insertion, deletion, or substitution of one or more nucleotides of said target polynucleotide sequence.
  • the target polynucleotide sequence is associated with a genetic disorder of the lung.
  • the target polynucleotide sequence is associated with cystic fibrosis, in particular the polynucleotide sequence is the cystic fibrosis transmembrane conductor receptor (CFTR) locus. Mutations in the CFTR (e.g., deletion of phenylalanine at position 508 in exon 1 1 ) cause cystic fibrosis.
  • CFTR cystic fibrosis transmembrane conductor receptor
  • the target polynucleotide sequence is associated with a genetic disorder of muscle.
  • the target polynucleotide sequence is associated with muscular dystrophies.
  • the target polynucleotide sequence is associated with Duchenne muscular dystrophy (DMD) (mutations in the dystrophin gene).
  • the target polynucleotide sequence is associated with Becker muscular dystrophy (mutations in the dystrophin gene).
  • the target polynucleotide is associated with myotonic dystrophy type 1 (mutations in the DMPK gene) or myotonic dystrophy type 2 (mutations in the CNBP gene).
  • the target polynucleotide sequence is associated with sickle cell anemia (mutated HBB hemoglobin).
  • the targeted polynucleotide sequence is associated with a genetic disorder of the liver.
  • the target polynucleotide sequence is associated with alpha- 1 antitrypsin deficiency (mutations in the SERPINA1 gene).
  • the targeted polynucleotide sequence is associated with Wilson disease (mutations in the gene encoding the ATP7B Cu translocase).
  • the methods of the invention further comprise providing a functional protein with enhanced characteristics as compared to its naturally occurring counterpart, in particular a functional protein lacking or deficient in a subject, for example for treating genetic disorders.
  • the methods comprise integrating a sequence encoding a functional protein in a cell in a subject in need thereof by sequential administration of a gene editing system and one or more transgene encoding a non-naturally occurring protein with enhanced properties as compared to its naturally occurring counterpart.
  • the methods comprise administering to the subject a genetically modified cell expressing a functional version of one or more proteins aberrantly expressed in a subject.
  • an isolated cell may be introduced into the subject (ex vivo cell therapy) or a cell may be modified when it is part of the subject (in vivo).
  • transgene(s) are delivered using a viral vector, a non-viral vector and/or combinations thereof.
  • the invention also provides a method for suppressing homologous recombination in a cell, in particular a cell in Gl , comprising suppressing the assembly of BRCA1-PALB2-BRCA2 complexes in the cell.
  • the interaction is suppressed by administering KEAP1 or CRL3-KEAP1 or an agonist thereof.
  • the interaction is suppressed by administering a USP1 1 antagonist/inhibitor (e.g., mitoxantrone).
  • the interaction is suppressed by administering an agent that suppresses homologous recombination identified using a method of the invention.
  • Components of the methods of the invention may be delivered by delivery systems known in the art, including without limitation viral based systems or non-viral based systems.
  • Conventional viral based systems may comprise, for example, retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer.
  • the expression vector is selected from the group consisting of a plasmid vector, a lentiviral vector, an adenoviral vector, and an adeno-associated virus vector.
  • the viral based system is an adenoviral vector or adeno-associated viral vector.
  • non-viral based systems examples include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virons and agent-enhanced uptake of DNA.
  • the invention provides vectors comprising activators or modulators of homologous recombination, and optionally activators or modulators of DNA end resection.
  • a vector e.g. viral vector
  • an activator of DNA end resection for example, an inhibitor of 53BP1 (or RTF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation
  • a factor that activates homologous recombination for example, a factor that maintains BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions during the cell cycle
  • components of a gene editing system in particular components of a CRISPR system, a TALEN system or a zinc finger nuclease system.
  • the components of the gene editing system are encoded in one or more separate expression vectors.
  • the invention provides a composition comprising activators or modulators of homologous recombination, and optionally activators or modulators of DNA end resection.
  • the invention provides a composition comprising one or more of the following components: 1) an activator of DNA end resection, for example, an inhibitor of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) a factor that activates homologous recombination, for example, a factor that maintains BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system.
  • the components of the gene editing system are in one or more separate compositions.
  • Examples of activators of DNA end-resection include without limitation, the coding sequence of CtIP-Thr847Glu, a shRNA against the TP53BP1 mRNA, and a shRNA against KEAP1.
  • the shRNA against TP53BP1 may be substituted with a shRNA against RIF1 or agents that block 53BP1 recruitment to DSB sites including a dominant-negative 53BP1 protein.
  • the shRNA against KEAP1 may be substituted with a the coding sequence of a PALB2 mutant that contains mutations of its Lys20, Lys25 and Lys30 residues or that contains a mutation that disrupts its interaction with KEAP1.
  • Examples of factors that maintain BRCA1 -PALB2 or BRCA1-PALB2- BRCA2 interactions during the cell cycle include without limitation, inhibitors of KEAP1 , inhibitors of DCAF10, RNA interference agents that maintain USP1 1 expression in GO and Gl cells or a mutated form of PALB2 that is insensitive to ubiquitylation by KEAP1 -CUL3-RBX1 which involves the mutation of one or more of the Lys20, Lys25 or Lys30 residues.
  • An example of a KEAP1 inhibitor is the monobody that is a potent competitive inhibitor of the KEAP1-NRF2 interaction disclosed in Guntas, G. et al, [44].
  • KEAP1 inhibitors are also described, for example in Canning P. et al, Acta Pharm Sin B., 2015 (4):285-99 and Wells, G., Biochem Soc Trans. 2015,43(4): 674-9.
  • a vector of the invention comprises sequences encoding an inhibitor of 53BP1, a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation.
  • a vector of the invention comprises sequences encoding a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu.
  • a vector of the invention comprises sequences encoding a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
  • a KEAP1 inhibitor e.g., Rl KEAP1 inhibitor; see Example 3
  • an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
  • a composition of the invention comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation.
  • a composition of the invention comprises a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu.
  • kits of the invention further provides a kit for performing an assay or method disclosed herein or comprising compositions or vectors disclosed herein.
  • a kit of the invention comprises at least one reagent for determining USP1 1 activity in a sample.
  • a kit of the invention comprises at least one reagent for determining BRCA1 -PALB2-BRCA2, PALB2-KEAP1 , BRCA1- PALB2, USP11-DCAF10, or USP1 1 -PALB2 complexes in a sample.
  • a kit of the invention comprises at least one reagent for determining BRCA1-PALB2-BRCA2, PALB2-KEAP1 or USP11-PALB2 complexes in a sample.
  • a kit of the invention comprises reagents for determining the levels of BRCA1 , BRCA2, PALB2, USP1 1 , DCAF10 and KEAP1 in a sample.
  • a kit of the invention comprises at least one reagent for determining ubiquitylation of PALB2, in particular ubiquitylation of the N-terminus of PALB2, in a sample.
  • the reagent is an antibody or a nucleic acid or primers for use in a PCR reaction.
  • a kit may also comprise instructions for suitable operational parameters in the form of an insert.
  • the instructions may inform a consumer about how to collect the sample.
  • the kit may comprise samples, to be used as standard(s) for calibration and comparison.
  • the kit may also comprise instructions to compare the level of activity or biomarkers detected in a sample with a calibration sample or chart.
  • the kit may also include instructions indicating what level of activity or biomarkers is diagnostic of a disease disclosed herein.
  • kits of the invention provides a kit comprising one or more of the components of a method of the invention for activating homologous recombination and optionally components of a gene editing system.
  • a kit of the invention may also include or be used in combination with a CRISPR system, a TALEN system or zinc finger nuclease system.
  • a kit of the invention includes or is used in combination with a CRISPR system.
  • a kit of the invention includes or is used in combination with a TALEN system.
  • a kit of the invention includes or is used in combination with a zinc finger nuclease system.
  • kits of the invention comprises vector systems and instructions for using the kit.
  • the kit comprises a vector comprising activators of DNA end resection and activators of homologous recombination discussed herein.
  • the kit comprises one or more vectors (e.g.
  • viral vectors comprising one or more of the following components: 1 ) activators of DNA end resection, for example, inhibitors of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) factors that activate homologous recombination, for example, factors that maintain BRCA1-PALB2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system.
  • activators of DNA end resection for example, inhibitors of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation
  • factors that activate homologous recombination for example, factors that maintain BRCA1-PALB2 interactions during the cell cycle
  • components of a gene editing system in particular components of a CRISPR system, TALEN system or zinc finger nuclease system.
  • factors that maintain BRCA1-PALB2 interactions during the cell cycle include without limitation, inhibitors of KEAP1 , for example, RNA interference agents that maintain USP1 1 expression in GO and G l cells or a mutated form of PALB2 that is insensitive to ubiquitylation by KEAP1 - CUL3-RBX1 which involves the mutation of one or more of the Lys20, Lys25 or Lys30 residues.
  • activators of DNA end resection include without limitation, the coding sequence of CtIP-Thr847Glu, a shRNA against the TP53BP1 inRNA, and a shRNA against KEAP1.
  • the shRNA against TP53BP1 may be substituted with a shRNA against RIF1 or agents that block 53BP1 recruitment to DSB sites including a dominant-negative 53BP1 protein.
  • the shRNA against KEAP1 may be substituted with a coding sequence of a PALB2 mutant that contains mutations of its Lys20, Lys25 and Lys30 residues or that contains a mutation that disrupts its interaction with KEAP1.
  • a kit of the invention comprises one or more vectors comprising sequences encoding an inhibitor of 53BP1 , a KEAPl inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation.
  • a kit of the invention comprises one or more vectors comprising sequences encoding a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu.
  • a kit of the invention comprises one or more vectors comprising sequences encoding a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
  • kits of the invention comprises a composition of the invention and instructions for using the kit.
  • the kit comprises a composition comprising activators or modulators of DNA end- resection and activators or modulators of homologous recombination discussed herein.
  • the kit comprises a composition comprising one or more of the following components: 1 ) an activator of DNA end resection, for example, an inhibitor of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) a factor that activates homologous recombination, for example, a factor that maintains BRCA1-PALB2 or BRCA1-PALB2-BRCA2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system.
  • the components of the gene editing system are in separate kit(s).
  • a kit of the invention comprises a composition comprising an inhibitor of 53BP1 , a KEAPl inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation.
  • a kit of the invention comprises a composition comprising a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu.
  • a kit of the invention comprises a composition comprising a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
  • a KEAPl inhibitor e.g., Rl KEAPl inhibitor; see Example 3
  • an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
  • a kit of the invention is used in combination with a gene editing kit, in particular a kit for a CRISPR system, a TALEN system or zinc finger nuclease system.
  • Gene editing kits are commercially available, for example from Addgene (Cambridge, MA), ThermoFisher Scientific, System Biosciences Inc., and OriGene Technologies (MD), Clontech.
  • the cDNA of PALB2 was obtained from the Mammalian Gene Collection (MGC). Full length PALB2 and BRCA1 were amplified by PCR, subcloned into pDONR221 and delivered into the pDEST-GFP, pDEST-Flag and the mCherry- LacR vectors using Gateway cloning technology (Invitrogen). Similarly, the coiled-coil domain of BRCA1 (residues 1363-1437) was amplified by PCR, subcloned into the pDONR221 vector and delivered into both mCherryLacR and pDEST-GFP vectors.
  • the USPl 1 cDNA was a gift of David Cortez and was amplified by PCR and cloned into the pDsRed2-C l vector using the EcoRI/Sall sites.
  • the bacterial codon-optimized coding sequence of pig USP1 1 (USP1 1 ) was subcloned into the 6 x His-GST vector pETM-30-Htb using the BamHI/EcoRI sites.
  • siRNA-resistant versions of PALB2, BRCA1 and USPl 1 constructs were generated as previously described [14].
  • Full-length CUL3 and RBX1 were amplified by PCR from a human pancreas cDNA library (Invitrogen) as previously described [32] and cloned into the dual expression pFBDM vector using Nhel/Xmal and BssHII/NotI respectively.
  • the NEDD8 cDNA was a gift of Dmitris Xirodimas and was fused to a double StrepII tag at its C-terminus in the pET17b vector (Millipore).
  • Human DEN1 was amplified from a vector supplied by Aude Echalier and fused to a non-cleavable N-terminal StrepII2 x tag by PCR and inserted into a pET17b vector.
  • the pCOOL-mKEAPl plasmid was a gift from Dr. Feng Shao.
  • the pcDNA3-HA2-KEAPl and pcDNA3 -H A2-KE AP 1 ⁇ were gifts from Dr. Yue Xiong (Addgene plasmids #21556 and 21593).
  • gRNAs were synthesized and processed as described previously [33].
  • Annealed gRNAs were cloned into the Cas9- expressing vectors pSpCas9(BB)-2A-Puro (PX459) or pX330-U6- Chimeric_BB-CBh- hSpCas9, a gift from Feng Zhang (Addgene plasmids #48139 and 42230).
  • the gRNAs targeting the LMNA or the PML locus and the mClover-tagged LMNA or PML are previously described [45]
  • the lentiviral packaging vector psPAX2 and the envelope vector VSV-G were a gift from Didier Trono (Addgene plasmids #12260 and 12259).
  • rabbit anti-53BPl (A300- 273A, Bethyl), rabbit anti-53BPl (sc-22760, Santa Cruz), mouse anti-53BPl (#612523, BD Biosciences), mouse anti-y-H2AX (clone JBW301 , Millipore), rabbit anti-y-H2AX (#2577, Cell Signaling Technologies), rabbit anti-KEAPl (ab66620, Abeam), rabbit anti-NRF2 (ab62352, Abeam), mouse anti-Flag (clone M2, Sigma), mouse anti-tubulin (CP06, Calbiochem), mouse anti-GFP (#1 1814460001 , Roche), mouse anti-CCNA (MONX10262, Monosan), rabbit anti-BRCA2 (ab9143, Abeam), mouse anti-BRCA2 (OP95, Calbiochem), rabbit anti-BRCAl (#07-434, Millipore), rabbit anti-USPl l (abl09232, Abeam
  • Alexa Fluor 488 donkey anti-rabbit IgG Alexa Fluor 488 donkey anti-goat IgG
  • Alexa Fluor 555 donkey anti- mouse IgG Alexa Fluor 555 donkey anti-rabbit IgG
  • Alexa Fluor 647 donkey anti- mouse IgG Alexa Fluor 647 donkey anti-human IgG
  • Alexa Fluor 647 donkey anti- goat IgG Molecular Probes.
  • RNA interference (RNAi) transfections were performed using Lipofectamine RNAiMax (Invitrogen) in a forward transfection mode.
  • siRNA duplexes were: BRCA1 (D-003461 -05), PALB2 (D-012928- 04), USP11 (D-006063-01), CUL1 (M-004086-01), CUL2 (M-007277-00), CUL3 (M- 010224-02), CUL4A (M-012610-01), CUL4B (M-017965-01), CUL5 (M-019553-01), KEAP1 (D-12453-02), RAD 51 (M-003530-04), QIP/RBBP8 (M-001376-00), BRCA2 (D-003462-04), 53BP1 (D-003549-01) and non-targeting control siRNA (D-001210- 02). Except when stated otherwise, siRNAs were transfected 48 h prior to cell processing.
  • cycloheximide (CHX; Sigma) at 100 ng/mL "1 , camptothecin (CPT; Sigma) at 0.2 ⁇ , ATM inhibitor (KU55933; Selleck Chemicals) at 10 ⁇ , ATR inhibitor (VE-821 ; gift of Philip Reaper) at 10 ⁇ , DNA-PKcs inhibitor (NU7441 ; Genetex) at 10 ⁇ , proteasome inhibitor MG132 (Sigma) at 2 ⁇ , Lovastatin (S2061 ; Selleck Chemicals) at 40 ⁇ , Doxycycline (#8634-1 ; Clontech), Nedd8-activating enzyme inhibitor (MLN4929; Active Biochem) at 5 ⁇ and olaparib (Selleck) at the indicated concentrations.
  • CHX cycloheximide
  • CPT camptothecin
  • ATM inhibitor (KU55933; Selleck Chemicals) at 10 ⁇
  • ATR inhibitor (VE-821 ; gift of Philip Reaper) at 10 ⁇
  • 293T and U20S cells were transiently transfected with 3 distinct sgRNAs targeting either 53BP1 , USP1 1 or KEAP1 and expressed from the pX459 vector containing Cas9 followed by the 2A-Puromycin cassette. The next day, cells were selected with puromycin for 2 days and subcloned to form single colonies or subpopulations. Clones were screened by immunoblot and/or immunofluorescence to verify the loss of 53BP1 , USP1 1 or EAP1 expression and subsequently characterized by PCR and sequencing.
  • the genomic region targeted by the CRISPR/Cas9 was amplified by PCR using Turbo Pfu polymerase (Agilent) and the PCR product was cloned into the pCR2.1 TOPO vector (Invitrogen) before sequencing.
  • MBP proteins expressed in Escherichia coli were purified on amylose resin (New England Biolabs) according to the batch method described by the manufacturer and stored in IX PBS, 5% glycerol.
  • GST proteins expressed in E. coli were purified on glutathione sepharose 4B (GE Healthcare) resin in 50 mM Tris HC1 pH 7.5, 300 mM NaCl, 2 mM dithiothreitol (DTT), 1 mM EDTA, 15 ⁇ g/mL "1 AEBSF and lx Complete protease inhibitor cocktail (Roche).
  • the His 6 -GST tag was cleaved off using His-tagged TEV protease (provided by F. Sicheri) in 50 mM Tris HC1 pH 7.5, 150 mM NaCl, 10 mM glutathione, 10% glycerol, 2 mM sodium citrate and 2 niM ⁇ -mercaptoethanol.
  • His 6 -tagged proteins were depleted using Ni-NTA-agarose beads (Qiagen) in 50 mM Tris HC1 pH 7.5, 300 mM NaCl, 20 mM imidazole, 5 mM glutathione, 10% glycerol, ImM sodium citrate and 2 mM ⁇ -mercaptoethanol followed by centrifugal concentration (Amicon centrifugal filters, Millipore).
  • GST-mKEAPl was purified as described previously [38], with an additional anion exchange step on a HiTrap Q HP column (GE Healthcare). The GST tag was left on the protein for in vitro experiments. Purification of CUL3 and RBX1 was performed as previously described [32].
  • Nedd8 and Denl were expressed in E.coli BL21 grown in Terrific broth media and induced overnight with 0.5 mM isopropyl-p-D-thiogalactoside (IPTG) at 16°C.
  • Cells were harvested and resuspended in wash buffer (400 mM NaCl, 50 mM Tris-HCl, pH 8, 5% glycerol, 2 mM DTT), supplemented with Iysozyme, universal nuclease (Pierce), benzamidine, leupeptin, pepstatin, PMSF and Complete protease inhibitor cocktail (Roche), except for DEN 1 -expressing cells where the protease inhibitors were omitted.
  • DEN1 was further purified over a Superdex 75 size exclusion column, buffer exchanged into 150 mM NaCl, HEPES, pH 7.6, 2% glycerol and 1 mM DTT.
  • the C- terminal pro-peptide and StrepII2x-tag were removed by incubation with StrepII2x- DEN1 in 1 :20 molar ratio for 1 hour at room temperature.
  • the DEN1 cleavage reaction was buffer exchanged on a Zeba MWCO desalting column (Pierce), to remove the desthiobiotin, and passed through a Strep-Tactin Cartridge, which retains the C- terminal pro-peptide and DENl .
  • HA-tagged N-terminal fragments of PALB2 (1-103) (1 ⁇ ) were in vitro ubiquitylated using 50 ⁇ wild-type (Ubi WT, Boston Biochem) or a lysine-less ubiquitin (Ubi K0, Boston Biochem), 100 nM human UBA1 (El), 500 nM CDC34 (provided by F. Sicheri and D.
  • Ceccarelli 250 nM neddylated CUL3/RBX1 , 375 nM GST-mKEAPl and 1.5 mM ATP in a buffer containing 50 mM Tris HC1 pH 7.5, 20 mM NaCl, 10 mM MgCl 2 and 0.5 mM DTT. Ubiquitylation reactions were carried out at 37°C for 1 hour, unless stated otherwise.
  • HA-PALB2 (1 -103) was first ubiquitylated using lysine-less ubiquitin with enzyme concentrations as described above in 50 reactions in a buffer containing 25mM HEPES pH 8, 150 mM NaCl, 10 mM MgCl 2 , 0.5 mM DTT and 1.5 mM ATP for 1.5 h at 37°C. Reactions were stopped by the addition of 1 unit Apyrase (New England Biolabs). Reaction products were mixed at a 1 : 1 ratio with wild-type or catalytically inactive (C270S) USP11 , or USP2 (provided by Dr. F. Sicheri and E.
  • PALB2 in vitro ubiquitylation reaction products were diluted in a buffer at final concentration of 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl 2 , 0.25 mM DTT and 0.1% NP-40. 20 ⁇ g MBP or MBP-BRCAl -CC was coupled to amylose resin (New England Biolabs) in the above buffer supplemented with 0.1% BSA prior to addition of the ubiquitylation products. Pulldown reactions were performed at 4°C for 2 h, followed by extensive washing.
  • Flag-PALB2 For MS analysis of Flag-PALB2, 150 x 10 6 transiently transfected HEK293T cells were lysed in high-salt lysis buffer (50 mM Tris-HCl pH 7.5, 300 mM NaCl, 1 mM EDTA, 1 % Triton X100, 3 mM MgCl 2 , 3 mM CaCl 2 ), supplemented with Complete protease inhibitor cocktail (Roche), 4 mM 1 , 10-Phenantroline, 50 U benzonase and 50 U micrococcal nuclease. Cleared lysates were incubated with Flag- M2 agarose (Sigma), followed by extensive washing in lysis buffer and 50 mM ammoniumbicarbonate.
  • high-salt lysis buffer 50 mM Tris-HCl pH 7.5, 300 mM NaCl, 1 mM EDTA, 1 % Triton X100, 3 mM MgCl
  • Tandem MS spectra were acquired in a data-dependent mode for the top two most abundant multiply charged peptides and included targeted scans for five specific N- terminal PALB2 tryptic digest peptides (charge state 1+, 2+, 3+), either in non- modified form or including a diGly-ubiquitin trypsin digestion remnant. Tandem MS spectra were acquired using collision-induced dissociation. Spectra were searched against the human Refseq_V53 database using Mascot, allowing up to 4 missed cleavages and including carbamidomethyl (C), deamidation (NQ), oxidation (M), GlyGly (K) and LeuArgGlyGly (K) [SEQ ID NO: 4] as variable modifications.
  • Digested peptides were mixed with 20 ⁇ of a mix of 10 unique heavy isotope-labeled N-terminal PALB2 (AQUA) peptides (covering full or partial tryptic digests of regions surrounding Lys 16, 25, 30 or 43, either in non- modified or diG-modified form; 80-1 ,200 fmol ⁇ ⁇ 1 per peptide, based on individual peptide sensitivity testing) before loading 6 ⁇ onto a 12 cm fused silica column with pulled tip packed in-house with 3.5 ⁇ Zorbax C18. Samples were measured on an Orbitrap ELITE (Thermo Scientific, MA, USA) coupled to an Eksigent nanoLC ultra (AB SCIEX, CA, USA).
  • Tandem MS spectra were acquired in a data-dependent mode for the top two most abundant multiply charged ions and included targeted scans for ten specific N-terminal PALB2 tryptic digest peptides (charge states 1 +, 2+, 3+), either in light or heavy isotope-labeled form. Tandem MS spectra were acquired using collision induced dissociation.
  • Spectra were searched against the human Refseq_V53 database using Mascot, allowing up to 2 missed cleavages and including carbamidomethyl (C), deamidation (NQ), oxidation (M), GlyGly (K) and LeuArgGlyGly (K) [SEQ ID NO:4] as variable modifications, after which spectra were manually validated.
  • Lysates were sonicated on ice twice for 10 sec with 1 min break and centrifuged at 15 000 g for 10 min at 4°C. The supernatant was incubated with Ni- NTA-agarose beads (Qiagen) for 4 h at 4°C.
  • Beads were collected by centrifugation, washed once with denaturating lysis buffer, once with wash buffer (8 M Urea, 0.1 M Na 2 HP0 4 /NaH 2 P0 4 , 10 mM Tris-HCl, 5 mM imidazole, 0.01 M ⁇ -mercaptoethanol, complete protease inhibitor cocktail), and twice with wash buffer supplemented with 0.1% Triton X-100, and eluted in elution buffer (0.2 M imidazole, 0.15 M Tris-HCl, 30% glycerol, 0.72 M ⁇ - mercaptoethanol, 5% SDS) before analysis by SDS-PAGE and immunoblotting.
  • Purified PALB2 cysteine mutant (final concentration of 600 ⁇ ) was mixed with Hise-TEV-Ubiquitin G76C (350 ⁇ ) in 300 mM Tris pH 8.8, 120 mM NaCl and 5% glycerol.
  • TCEP Tris(2-carboxyethyl)phosphine
  • the bi-reactive cysteine cross-linker 1 ,3-dichloroacetone (Sigma-Aldrich), was dissolved in dimethylformamide and added to the protein mix to a final concentration of 5.25 mM. The reaction was allowed to proceed on ice for 1 h, before being quenched by the addition of 5 mM ⁇ -mercaptoethanol. His 6 -TEV-Ubiquitin-conjugated PALB2 was enriched by passing over Ni-NTA-agarose beads (Qiagen).
  • PALB2 ubiquitylation suppresses its interaction with BRCAl and is counteracted by the deubiquitylase USP1 1 , which is itself under cell cycle control.
  • Restoration of the BRCAl -PALB2 interaction combined with the activation of DNA-end resection is sufficient to induce HR in Gl , as measured by RAD51 recruitment, unscheduled DNA synthesis and a CRISPR/Cas9-based gene targeting assay.
  • the mechanism prohibiting HR in Gl minimally consists of the suppression of DNA-end resection coupled with a multi-step block of the recruitment of BRCA2 to DNA damage sites that involves the inhibition of BRCAl -PALB2-BRCA2 complex assembly.
  • the ability to induce HR in Gl cells with defined factors may be used in gene targeting applications in non- dividing cells.
  • BRCAl promotes DNA-end resection to produce the single-stranded (ss) DNA necessary for homology search and strand invasion [1], and it interacts with PALB2 [13-15] to direct the recruitment of BRCA2 [13] and RAD51 [16, 17] to DSB sites.
  • the accumulation of BRCAl on the chromatin that flanks DSB sites is suppressed in Gl cells [18], reminiscent of the potent inhibition of HR in this phase of the cell cycle. Since the inhibition of BRCAl recruitment in Gl is dependent on the 53BP1 and RIF1 proteins [18, 19], two inhibitors of end-resection [18-22], this regulation of BRCAl was originally viewed in light of its function in stimulating DNA-end processing.
  • KEAPl is a substrate adaptor for a CULLIN 3 -RING ubiquitin (Ub) ligase (CRL3) that targets the antioxidant regulator NRF2 for proteasomal degradation [24] and recognizes an "ETGE" motif on both PALB2 and NRF2 through its KELCH domain [6].
  • Ub CULLIN 3 -RING ubiquitin
  • CCL3 CULLIN 3 -RING ubiquitin (Ub) ligase
  • CUL3 also interacts with PALB2 (Figure 7c) and its depletion in 53BP1A U20S cells de-repressed PALB2 IR-induced foci in Gl ( Figure I d and Figure 7a). Furthermore, in Gl -synchronized KEAP1A cells, expression of a CUL3-binding deficient KEAPl protein that lacks its BTB domain ( ⁇ ) failed to suppress the BRCA1 -PALB2 interaction, unlike its wild type counterpart (Figure 7d). These results suggest that KEAPl recruits CUL3 to PALB2 to suppress its interaction with BRCA1.
  • ubiquitylation of the N-terminus of PALB2 could be readily reconstituted by recombinant CRL3-KEAP1 , in a manner that depended on the KEAP1 -interaction domain of PALB2 ( Figure 2c) and Lys25 and Lys30 were unambiguously identified as being ubiquitylated by KEAP1 in vitro by mass spectrometry.
  • a CRL4 E3 Ub ligase is most likely responsible for controlling the stability of USPl l as treatment with MLN4924, a pan-CRL inhibitor [28] ( Figure l l g), or depletion of CUL4 (Figure 3f> protected USPl l from DNA damage-induced degradation.
  • CUL4 depletion led to BRCA2 and PALB2 IR-induced focus formation in Gl 53BP1A cells ( Figure 3g and Figure 12a), consistent with the regulation of USP1 1 by a CRL4 complex acting as the upstream signal that ultimately controls BRCA1 -PALB2-BRCA2 complex assembly.
  • CUL4-RING-Ligase (CRL4) complexes are composed of CULLIN4 (CUL4), RBXl , DDB1 , DDA1 and a substrate adaptor called a DCAF [42].
  • CUL4-RING-Ligase (CRL4) complexes are composed of CULLIN4 (CUL4), RBXl , DDB1 , DDA1 and a substrate adaptor called a DCAF [42].
  • a focused siRNA library was assembled that depletes known and predicted DCAFs along with other CUL4-interacting proteins. This library was screened in a high content microscopy assay where USPl l levels were evaluated by immunofluorescence microscopy. Cells were treated either with ultraviolet light (UV) or ionizing radiation (IR) to induce USPl l degradation.
  • UV ultraviolet light
  • IR ionizing radiation
  • DCAF10 is indeed involved in the regulation of USP1 1
  • mouse embryo fibroblasts (MEFs) were generated from congenic wild type (Deaf 1 CT /+ ), heterozygote (Deaf 10 "/+ ) and Deaf 10 _/" mice and immunoblotted for USP1 1.
  • Loss of DCAF10 resulted in higher steady-state levels of USP11 in mouse cells ( Figure 15b), consistent with DCAF10 being the adaptor of a CRL4 complex targeting USP11.
  • DCAF10 over-expression can suppress homologous recombination in a dominant manner using the direct repeat (DR)-GFP assay [43].
  • a genetically-encoded inhibitor of KEAP1 can promote homologous recombination in Gl cells.
  • the LMNA gene targeting assay [45] was carried out in 53 ⁇ 1 ⁇ U20S cells synchronized in Gl phase and it was found that transfection of the Rl KEAP1 inhibitor, but not its FN3 control, led to a robust activation of gene-targeting, albeit less, and KEAP1 depletion (Figure 16). Inhibition of KEAP1 can be a propitious route for the activation of HR in non-dividing cells.
  • PALB2 partner and localizer of BRCA2
  • KEAP1 (kelch-like ECH-associated protein 1) NP_036421.2 9
  • DCAF 10 (DDB l - and CUL4-associated factor 10 NP_077321.3 13 isoform a)
  • DCAF DDBl - and CUL4-associated factor 10 isoform NP_001273739.1 14 b
  • CtIP/RBBP8 Homo sapiens retinoblastoma binding NG_012121.1 17 protein 8)
  • CULLIN3-RING ubiquitin ligases CULLIN3-RING ubiquitin ligases
  • Ubiquitin-specific Protease 1 1 (USP11 ) Deubiquitinates Hybrid Small Ubiquitin-like Modifier (SUMO)-Ubiquitin Chains to Counteract RING Finger Protein 4 (RNF4). J Biol Chem 290, 15526-15537, doi: 10.1074/jbc.Ml 14.618132 (2015).
  • Nonhydrolyzable diubiquitin analogues are inhibitors of ubiquitin conjugation and deconjugation. Biochemistry 39, 10001 -10010 (2000).
  • CRL4s the CUL4-RING E3 ubiquitin ligases. Trends Biochem Sci 34, 562-570.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention relates to factors that influence or regulate homologous recombination, methods to monitor these factors, the use of these factors to screen for agents that modulate homologous recombination, and methods to activate or modulate homologous recombination.

Description

Title: HOMOLOGOUS RECOMBINATION FACTORS
FIELD OF THE INVENTION
[0001] The invention relates to factors that influence or regulate homologous recombination, methods to monitor these factors, the use of these factors to screen for agents that modulate homologous recombination, and methods to modulate homologous recombination.
BACKGROUND OF THE INVENTION
[0002] The breast and ovarian tumour suppressors BRCAl , PALB2 and
BRCA2 promote DNA double-strand break (DSB) repair by homologous recombination (HR) [8-10]. BRCAl acts in this process minimally at two discrete steps. Firstly, it promotes DNA end resection [11 , 12], the initiating step in HR that involves the nucleolytic processing of breaks to produce the single-stranded (ss) DNA necessary for homology search and strand invasion [1]. Secondly, BRCAl interacts with PALB2 [13-15] to direct the recruitment of BRCA2 [13] and RAD51 [16, 17] to DSB sites. The accumulation of BRCAl on the chromatin that flanks DSB sites is strikingly suppressed in Gl cells [18], reminiscent of the potent inhibition of homologous recombination in this phase of the cell cycle. The inhibition of BRCAl recruitment in Gl is dependent on the 53BP1 and RIF1 proteins [18, 19], two inhibitors of end-resection [18-22]. BRCAl is also involved in promoting the recruitment of BRCA2 through its interaction with PALB2 [13-15].
[0003] Tumors with compromised ability to repair double-strand DNA breaks by HR, including those with defects in BRCAl and BRCA2, have been shown to be highly sensitive to poly ADP-ribose polymerase (PARP) inhibitors. PARP inhibitors have also been proposed for treating other conditions such as stroke, myocardial infarction, inflammatory bowel disorders, head trauma, and neurodegenerative diseases. Inhibition of ubiquitin-specific peptidase 1 1 (USP1 1) has been shown to hypersensitize cells to PARP inhibitors and it has been proposed that USPl l status or the status of other HR-proteins in tumors may provide biomarkers for use of PARP inhibitors (Wiltshire et al, JBC 285(19), 14565-14571 , 2010).
[0004] Identification and assessment of factors that influence or regulate homologous recombination repair proteins and the identification of events that are both necessary and sufficient to suppress HR in Gl cells is desirable. In addition, identification and assessment of factors that influence or regulate USP1 1 may facilitate the selection and monitoring of PARP inhibitor treatments, and in particular selection of treatments that reverse or delay emergence of PARP inhibitor resistance.
SUMMARY OF THE INVENTION
[0005] The present inventors have found that the cell cycle tightly controls the interaction of BRCAl with PALB2-BRCA2 in order to constrain BRCA2 function to the S/G2 phases. The BRCAl -interaction site on PALB2 is targeted by an E3 ubiquitin ligase composed of KEAP1 , a PALB2-interacting protein [6], in complex with c u ll in 3 ( CUL3)-RBX1 [7]. PALB2 ubiquitylation suppresses its interaction with BRCAl and is counteracted by the deubiquitylase USPl l , which is itself under cell cycle control. Restoration of the BRCAl -PALB2 interaction combined with the activation of DNA end resection was sufficient to induce HR in Gl -phase cells, as measured by RAD51 recruitment, unscheduled DNA synthesis and a CRISPR/Cas9- based gene targeting assay. The mechanism prohibiting HR in Gl minimally consists of the suppression of DNA end resection coupled to a multi-step block to BRCA2 recruitment to DNA damage sites that involves the inhibition of BRCAl -PALB2- BRCA2 complex assembly. The ability to induce HR in Gl cells with defined factors may be used in gene targeting applications in non-dividing cells or cells that are dormant in Gl phase. The findings also provide a basis for targeting USPl l in combination with poly(ADP-ribose) polymerase (PARP) inhibitors.
[0006] The present inventors have also found that USP1 1 is regulated by a cell cycle-CULLIN4-RING-ligase (CRL4) and DCAF10 acts as an adaptor for the USPl l E3 ligase.
[0007] The invention provides a method for monitoring activity of USP1 1 in a sample by assaying the interaction of BRCAl and PALB2.
[0008] The invention provides a method for monitoring activity of USPl l in a sample by assaying the interaction of BRCAl , PALB2, and BRCA2.
[0009] The invention provides a method for monitoring activity of USP1 1 in a sample by assaying the interaction of USP1 1 and PALB2.
[00010] The invention provides a method for monitoring activity of USP1 1 in a sample by assaying DC AF 10.
[00011] The invention provides a method for monitoring activity or expression of USP1 1 in a sample by assaying for complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USP1 1 and PALB2; and/or (d) USP1 1 and DCAF10. [00012] In an aspect, the invention provides a method for monitoring activity or expression of USPl l in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USPl l and PALB2; and/or (d) USPl l and DCAF10 in the sample; (ii) measuring the levels of the complexes; and (iii) detecting an increase or decrease in the activity or expression of the complexes as compared to a control as an indication of the activity or expression of USP1 1.
[00013] In an aspect, the invention provides a method for monitoring activity or expression of USP1 1 in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USPl l and PALB2; and/or (d) USPl l and DCAF10 in the sample by immunological purification; (ii) measuring the levels of the complexes; and (iii) detecting an increase or decrease in the activity or expression of the complexes as compared to a control as an indication of the activity or expression ofUSPl l .
[00014] In an aspect, the invention provides a method for monitoring activity or expression of USP11 in a sample comprising (i) isolating complexes of (a) BRCAl and PALB2; (b) BRCAl , PALB2, and BRCA2; (c) USP1 1 and PALB2; and/or (d) USP1 1 and DCAF10 in the sample; (ii) preparing peptides or peptide fragments from the isolated complexes; and (iii) subjecting the peptides or peptide fragments to mass spectrometry to thereby monitor the activity or expression of USP1 1.
[00015] The invention provides a method for monitoring activity or expression of USP11 in a sample by assaying ubiquitylation of PALB2, in particular ubiquitylation of the N-terminus of PALB2.
[00016] In an aspect, the invention provides a method for monitoring activity or expression of USPl l in a sample by assaying ubiquitylation of PALB2 comprising measuring the amount of polyubiquitin bound to CRL3-KEAP1 E3 ligase in the sample and detecting an increase or decrease in polyubiquitin bound to CRL3-KEAP1 E3 ligase as compared to a control as an indication of the activity or expression of USP1 1.
[00017] In another aspect, the invention provides a method for monitoring activity or expression of USP1 1 in a sample by assaying ubiquitylation of PALB2 comprising measuring the activity of CRL3-KEAP1 E3 ligase, and detecting an increase or decrease in CRL3-KEAP1 E3 ligase activity as compared to a control as an indication of the activity or expression of USP1 1.
[00018] The methods of the invention may be performed in the presence or absence of a test compound or agent and detection of an increase or decrease in activity or expression of one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA 1 -PALB2 -BRC A2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF complex, CRL3-KEAP1 complex, CRL3-KEAP1 -PALB2 complex, and KEAP1 - PALB2 complex, as compared to a control in the absence of the test compound or agent indicates that the test compound or agent may be useful as a therapeutic agent, or for modulating homologous recombination.
[00019] In an aspect, the invention provides a method for identifying or evaluating an agent for its ability to sensitize or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on USPl 1 activity or expression using a method of the invention.
|00020] In an aspect, the invention relates to a method of identifying or evaluating an agent for its ability to sensitize cells or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on KEAP1 , CRL3- KEAP1, KEAP1-PALB2 or CRL3-KEAP1.
[00021] In an aspect, the present invention provides methods of detecting an anticancer agent comprising performing a test assay comprising contacting an immortalized cell with a test compound and assaying USPl 1 activity or expression using a method of the invention.
[00022] The invention also provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination comprising determining the effect of a test compound or agent on one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 -PALB2-BRCA2 complex, PALB2-USP 1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 complex and CRL3-KEAP1 - PALB2 complex.
[00023] The invention provides a method of screening for a therapeutic agent for treatment of a disease associated with defects in HR (i.e., HR Disease), comprising identifying an agent that disrupts or modulates one or more of USPl 1 , PALB2, PALB2 ubiquitylation, DCAF10, BRCA1 -PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAPl , USP11-DCAF10 complex, CRL3-KEAP1 or CRL3-KEAP1 -PALB2 complex.
[00024] The screening methods of the invention may further comprise conducting therapeutic profiling of the identified agents or further analogs thereof, for efficacy and toxicity in animals; optionally formulating a pharmaceutical composition including one or more agents identified as having an acceptable therapeutic profile; and optionally administering the agent to a subject or individual. [00025] The invention provides methods of treating a HR Disease in an individual comprising identifying an agent that modulates HR in accordance with a method of the invention and administering the agent to the individual.
[00026] In some embodiments, the invention provides a method for sensitizing cells to PARP inhibitors in an individual comprising identifying an agent that sensitizes cells to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
[00027] In some embodiments, the invention provides a method for reversing or delaying emergence of resistance to PARP inhibitors in an individual comprising identifying an agent that reverses or delays emergence of resistance to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
[00028] In some embodiments, the present invention provides methods of treating cancer in an individual comprising identifying an anti-cancer agent identified in accordance with a method of the invention and administering the agent to the individual.
[00029] The invention also provides a method for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying one or more of USPl l , DCAFI O, BRCAl, BRCA2, PALB2, KEAP1 , CRL3, CRL3-KEAP1 , USPl l -DCAFIO complex, BRCAl -PALB2-BRCA2 complex, PALB2-USP1 1 complex and CRL3-KEAP1-PALB2 complex in a sample from the subject using a method of the invention. In an aspect, a method is provided for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying USPl l activity or expression in a sample from the subject using a method of the invention. In an aspect, a method is provided for predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying PALB2 activity or expression in a sample from the subject using a method of the invention.
[00030] In an aspect, a subject is categorized as responsive to a PARP inhibitor if there is a decrease in one or more of USPl l, DCAFIO, BRCAl , BRCA2, PALB2, KEAP1 , CRL3, USP1 1 -DCAFI O, CRL3-KEAP1, BRCAl -PALB2 and BRCAl - PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control. In an aspect, a subject is categorized as responsive to a PARP inhibitor if there is an increase in one or more of USPl l , DCAFI O, BRCAl , BRCA2, PALB2, KEAP1 , CRL3, USP1 1 -DCAF10, CRL3-KEAP1 , BRCA1-PALB2 and BRCA1 -PALB2- BRCA2 activity or expression or PALB2 ubiquitylation compared to a control.
[00031] A method of predicting responsiveness to a PARP inhibitor may further comprise administering the PARP inhibitor to the individual.
[00032] The invention provides a method for treating a patient in need of treatment with a PARP inhibitor comprising (a) requesting a test providing the results of an analysis to determine if the patient is sensitive or responsive to the PARP inhibitor by detecting one or more of USP11 , DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 , USP1 1-DCAF10, CRL3, CRL3-KEAP1 , BRCA1-PALB2 and BRCA1- PALB2-BRCA2, in a sample from the subject and comparing to a control to determine if the patient is sensitive or responsive to the PARP inhibitor; and (b) administering the PARP inhibitor to the patient if the patient is sensitive or responsive to the PARP inhibitor. In an aspect of this method of the invention, the patient has breast cancer. In an aspect of this method of the invention, the patient has ovarian cancer.
[00033] In an aspect, the invention provides a method for treating a patient in need of treatment with a PARP inhibitor comprising (a) requesting a test providing the results of an analysis to determine if the patient is sensitive to the PARP inhibitor by detecting USP1 1, DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 and/or CRL3 in a sample from the subject and comparing to a control to determine if the patient is sensitive to the PARP inhibitor; and (b) administering the PARP inhibitor to the patient if the patient is sensitive to the PARP inhibitor. In an aspect of this method of the invention, the patient has breast cancer. In an aspect of this method of the invention, the patient has ovarian cancer.
[00034] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USP11 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, USP1 1 -DCAF complex, KEAP1 , CRL3-KEAP1 and/or CRL3- EAP1 -PALB2 complex in a sample from the subject using a method of the invention.
[00035] The invention also provides pharmacogenetic methods for determining suitable treatment regimens for diseases, in particular cancer, and methods for treating patients, based around selection of patients based on PARP responsiveness, in particular USP 11 activity. [00036] The methods of the invention, in particular methods for assaying USP1 1 activity or CRL3-KEAP1 activity, may be used as a read out in animal model based screening methods for new therapeutic approaches and compounds. In an aspect, a method of the invention is utilized to predict the efficacy of potential new treatments in animal models for disease states.
[00037] The invention provides a method for activating or modulating (e.g., promoting) homologous recombination in a cell comprising:
(a) promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell;
(b) activating or stimulating BRCA1 recruitment to DNA double-strand break
(DSB) sites;
(c) contacting the cell with BRCA1-PALB2 or BRCA1 -PALB2-BRCA2 complexes;
(d) inhibiting or removing KEAP1 or CRL3-KEAP1 ;
(e) inhibiting the degradation of USP1 1 or promoting USP11 activity; and/or
(f) inhibiting or removing DCAF10.
[00038] The invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in G l phase of the cell cycle (Gl) or GO phase of the cell cycle, comprising administering, or stimulating assembly of BRCA 1 -PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell.
[00039] The invention also provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell.
[00040] The invention also provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising administering to the cell or contacting the cell with BRCA1-PALB2 or BRCA 1 -PALB2-BRCA2 complexes.
[00041] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1 - PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell. [00042] In aspects of the invention, the assembly of BRCA1-PALB2 or BRCA1- PALB2-BRCA2 complexes is promoted or stimulated by administering an agent that promotes or stimulates such assembly or an agent that promotes or stimulates such assembly identified using a method of the invention. In an embodiment, the agent is USP1 1 or an agonist of USP1 1. In an embodiment, the agent is an inhibitor of CRL- KEAPl . In an embodiment, the agent is an inhibitor of KEAPl . In an embodiment, the agent is a PALB2 mutant. In an embodiment, the agent is a PALB2 mutant that disrupts its interaction with KEAPl . In an embodiment, the agent is a PALB2 comprising mutations of its Lys20, Lys25 and Lys30 residues.
[00043] A method for activating or modulating homologous recombination in a cell may be performed in a cell wherein single strand DNA (ssDNA) generation pathways are activated. In an aspect, ssDNA generation pathways in the cell are activated by DNA end resection.
[00044] The invention also provides a method for activating or modulating homologous recombination in a cell, in particular a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell.
[00045] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell. In an embodiment, the assembly of the complexes is promoted or stimulated by administering an agent that modulates HR. In an embodiment, the agent is an agent that modulates HR identified using a method of the invention. In an embodiment, the agent is USPl l or an agonist of USPl l . In an embodiment, the agent is an inhibitor of CRL-KEAPl . In an embodiment, the agent is an inhibitor of KEAPl . In an embodiment, the agent is a PALB2 mutant. In an embodiment, the agent is an inhibitor of DCAF10. In an embodiment, the agent is an inhibitor of a CULLIN4-PJNG-ligase.
[00046] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising contacting the cell with BRCA1 -PALB2 or BRCA 1 -PALB2-BRCA2 complexes.
[00047] In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting KEAP1 or CRL3-KEAP1 or administering an inhibitor of KEAP1 or CRL3-KEAP1. In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of blocking the degradation of USP1 1 or promoting or stimulating USP1 1 activity. In an embodiment, the method comprises administering USP1 1 or an agonist thereof. In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting CRL-KEAPl or administering an inhibitor of KEAP1 or CRL3-KEAP1 and blocking the degradation of USP1 1 or promoting or stimulating USP1 activity.
[00048] The invention also provides a method for repairing DNA double-strand breaks in a cell in Gl or GO in which DNA end resection is or has been activated generating single-stranded DNA, the method comprising (a) inhibiting KEAPl or CRL3-KEAP1 ; (b) blocking the degradation of USP1 1 or promoting or stimulating USP1 1 activity; (c) administering USP1 1 or an agonist thereof; (d) administering an inhibitor of KEAPl or CRL3 -KEAPl ; (e) administering an inhibitor of DCAF10; and/or (e) inhibiting CRL-KEAPl and blocking the degradation of USP1 1.
[00049] A method for activating or modulating homologous recombination in a cell may further comprise activating or promoting single-strand DNA (ssDNA) generation pathways. In an aspect, ssDNA generation pathways are activated by DNA end resection.
[00050] A method for activating or modulating homologous recombination in a cell may further comprise a gene editing system. In an aspect the gene editing steps comprise contacting the cell with a nuclease. In aspects of the invention the gene editing system may correct a genomic modification.
[00051] The invention also provides a method for suppressing homologous recombination in a cell, in particular a cell in Gl , comprising suppressing the assembly of BRCA1-PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell. In an embodiment, the interaction is suppressed by administering KEAPl or CRL3 -KEAPl or an agonist thereof. In an embodiment, the interaction is suppressed by administering a USPl l antagonist/inhibitor (e.g., mitoxantrone). In an embodiment, the interaction is suppressed by administering an agent that inhibits or suppresses HR identified using a method of the invention.
[00052] The invention further provides kits for performing methods of the invention.
[00053] The invention also provides a system comprising: an assay for determining the level of USP 1 1 activity, complexes or biomarker levels in a sample obtained from the subject; a processor for processing the results; computer coded instructions for comparing the results with a database; and a user display for providing the results of the comparison. The database may comprise reference values for USP1 1 activity or biomarker levels.
[00054] The invention also contemplates the use of methods, kits, and systems of the invention in genome modification or editing.
[00055] In an aspect, the invention further contemplates the use of methods, compositions, kits, and systems of the invention in genome modification or editing, provided that said use is not a method for treatment of the human or animal body by surgery or therapy, and provided that said use is not a process for modifying the germ line genetic identity of human beings. Genome modification may comprise modifying a target polynucleotide sequence in a cell, modifying expression of a polynucleotide sequence in a cell, generating a model cell comprising a mutated disease gene, or knocking out a gene. A use of the invention may further comprise repairing or editing a cleaved target polynucleotide by inserting an exogenous template polynucleotide, wherein the repair or editing results in a mutation comprising an insertion, deletion, or substitution of one or more nucleotides of the target polynucleotide.
[00056] Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
DESCRIPTION OF THE DRAWINGS
[00057] The invention will now be described in relation to the drawings in which: [00058] Figure 1. Inhibition of the BRCA1-PALB2 interaction in Gl is CRL3- KEAP1 -dependent, a, Micrographs of irradiated (2 Gy) Gl -synchronized U20S cells processed for γ-Η2ΑΧ, BRCA1 and BRCA2 immunofluorescence. DAPI, 4' ,6- diamidino-2-phenylindole; IR, ionizing radiation; WT, wild type, b, Quantitation of the experiment shown in a and Figure 5d. ASN, asynchronously dividing cells. WT, wild type (Mean ± standard deviation (s.d.), N=3). c, Immunoprecipitation (IP) of PALB2 from extracts prepared from mock- or X-irradiated 293T cells synchronized in S or Gl phases. A normal immunoglobulin (Ig)G immunoprecipitation was performed as control. Cyclin A staining ascertains cell cycle synchronization. Numbers on left indicate kDa. For gel source data see Figure 5. d, Quantitation of the experiment shown in Figure 7a. 53BPJA U20S cells transfected with the indicated GFP-PALB2 vectors and short interfering (si)RNAs were irradiated (20 Gy) before being processed for microscopy, (mean ± s.d., N=3). e, Normal IgG and PALB2 immunoprecipitations from extracts prepared from synchronized and irradiated 293T cells of the indicated genotypes. Numbers on the left indicate kDa.
[00059] Figure 2. Ubiquitylation of PALB2 prevents BRCA1-PALB2 interaction, a, Sequence of the PALB2 N terminus and mutants. [SEQ ID NOs: 1 -3] b, GFP immunoprecipitation (IP) of extracts derived from Gl - or S-phase synchronized 293T cells expressing the indicated GFP-PALB2 proteins, c, In vitro ubiquitylation of the indicated HA-tagged PALB2 proteins by CRL3-KEAP1. d, Pulldown assay of ubiquitylated HA-PALB2 (1-103) incubated with MBP or MBP-BRCA1 -CC. I, input; FT, flow-through; PD, pulldown. The asterisk denotes a fragment of HA-PALB2 competent for BRCA1 binding, b-d, Numbers on left indicate kDa.
[00060] Figure 3. USP1 1 opposes the activity of CRL3-KEAP1. a, Normal IgG or PALB2 immunoprecipitation (IP) of extracts derived from camptothecin (CPT)- treated 293T cells of the indicated genotypes transfected with GFP— USP1 1 constructs. EV, empty vector; CS, C318S; WT, wild type, b, Clonogenic survival assays of 293T cells of the indicated genotypes treated with olaparib (mean ± s.d., N > 3). c, Normal IgG or PALB2 immunoprecipitation of extracts derived from CPT-treated 293T cells of the indicated genotypes, d, Immunoblots of deubiquitylation reactions containing ubiquitylated HA-tagged PALB2 (1-103) and increasing concentrations of glutathione ^-transferase (GST)-USP1 1 or its C270S (CS) mutant. USP2 was used as a control. DUB, deubiquitylase. e, Cell cycle- synchronized U20S cells were irradiated (20 Gy dose) and processed for immunoblotting. IR, ionizing radiation, f, Immunoblots of extracts from irradiated U20S cells transfected with the indicated siRNAs. CTRL, control, g, Fluorescence micrographs of Gl -synchronized and irradiated (20 Gy) 53ΒΡ1Δ U20S cells transfected with the indicated siRNAs. The percentage of cells with more than five γ -H2AX-colocalizing BRCA2 foci is indicated (mean ± s.d., N = 3). Scale bars, 5 μ m. a, c, d, f, Numbers to left or right indicate kDa.
[00061] Figure 4. Reactivation of HR in Gl phase, a, Quantitation of wild-type (WT) and 53BP1A U20S cells co-transfected with non-targeting (CTRL) or KEAP1 siRNAs and vectors expressing wild-type CtIP or the T847E (TE) mutant that were synchronized in Gl , irradiated (2 Gy) and processed for γ -H2AX and RAD51 immunofluorescence (mean ± s.d., N = 3). b, Representative micrographs from a. IR, ionizing radiation, c, Schematic of the gene-targeting assay, d, Gene-targeting efficiency at the LMNA locus in asynchronously dividing (ASN) and Gl -arrested U20S cells (mean ± s.d., N= 3). HR, homologous recombination; sgRNA, single guide RNA. e, Gene targeting at the LMNA locus in Gl -arrested cells transfected with the indicated siRNA or a PALB2-KR expression vector (mean ± s.d., N = 3). f, Model of the cell- cycle regulation of homologous recombination.
[00062] Figure 5. Suppression of PALB2-BRCA2 accumulation at DSB sites in Gl 53ΒΡ1Δ cells, a, Schematic representation of human 53BP1 gene organization and targeting sites of sgRNAs used. Boxes indicate exons (E: yellow, coding sequence; brown, untranslated regions (UTRs)). The indels introduced by CRISPR/Cas9 and their respective frequencies are indicated, b, Wild-type (WT) and 53BP1A and U20S cells were mock- or X-irradiated (10 Gy) before being processed for 53BP1 fluorescence microscopy. DAPI was used to stain DNA and trace the outline of the nucleus, c, Wild- type (WT) and 53BP1A U20S cells were processed for 53BP1 immunoblotting. Tubulin was used as a loading control, d, Wild-type (WT) and 53BP1A U20S cells either synchronized in G l following a double-thymidine block and release or asynchronously dividing (ASN), were irradiated (2 Gy) and processed for γ-Η2ΑΧ, PALB2, BRCA2 and BRCA1 , immunofluorescence. The micrographs relating to BRCA1 and BRCA2 staining in Gl are found in Figure l a. e, Wild-type (WT) and 3BP1A U20S cells synchronized in Gl after release from a double-thymidine block were irradiated (20 Gy) and processed for γ-Η2ΑΧ, BRCA1 and BRCA2 immunofluorescence. On the left are representative micrographs for the Gl -arrested cells and the quantitation of the full experiment is shown on the right (mean ± s.d., N=3). [00063] Figure 6. The BRCA1-PALB2 interaction is cell cycle regulated, a, Schematic of the LacO/LacR chromatin-targeting system, b, U20S 256 cells were transfected with the indicated mCherry-LacR and GFP-fusions. GFP fluorescence was measured at the site of the lacO array-localized mCherry focus. Each circle represents one cell analyzed and the bar is at the median. Cells were also stained with a cyclin A antibody to determine cell cycle position (N=3). IR, Ionizing radiation, c, Representative micrographs of U20S 256 cells transfected with the indicated mCherry- LacR and GFP-fusions; data is quantified in d. d, Quantification of U20S 256 cells transfected with the indicated mCherry-LacR and GFP-fusions to tether either BRCA1 or PALB2 to the lacO array (N=3). e, Schematic representation of PALB2 architecture and its major interacting proteins, f, Quantification of U20S 256 cells transfected with the indicated GFP-PALB2 mutants and mCherry-LacR-BRCAl-CC. Cells were also stained with a cyclin A antibody to determine cell cycle position (N=3).
[00064] Figure 7. Inhibition of the BRCA1-PALB2 interaction in Gl depends on CRL3-KEAP1. a, Representative micrographs of the experiment shown in Figure I d. b, Schematic representation of human KEAPl gene organization and targeting sites of sgRNAs used as described in Figure 5a. The indels introduced by CRISPR/Cas9 and their respective frequencies are indicated, c, Immunoprecipitation (IP) of PALB2 from extracts prepared from irradiated 293T cells. IP with normal IgG was performed as a control, d, 293T cells with the indicated genotypes were transfected with the indicated HA-KEAP1 constructs, synchronized in Gl or S phases and irradiated. Cells were processed for PALB2 immunoprecipitation (IP). EV, empty vector; WT, wild type, e, Quantification of U20S 256 cells transfected with the indicated GFP-PALB2 mutants and mCherry-LacR-BRCAl . Cells were also stained with a cyclin A antibody to determine cell cycle position (N=3). f, Quantification of U20S 256 cells transfected with GFP-PALB2 and mCherry-LacR-BRCAl -CC (wild type or K1406R mutant). Cells were also stained with a cyclin A antibody to determine cell cycle position. This panel shows that the sole lysine in the PALB2-interaction motif of BRCA1 is not involved in the cell cycle regulation of the PALB2-BRCA1 interaction, e, f, Each circle represents a cell analyzed and the bar is at the median (N = 3).
[00065] Figure 8. PALB2 is ubiquitylated by CRL3-KEAP1. a, HEK293 Flp-In T-REX cells expressing doxycycline (DOX)-inducible His6-Ub were transfected with the indicated siRNAs. Cells were processed for Ni-NTA pull-down (IP), b, 293T cells transfected with an siRNA targeting USP1 1 and a Flag-PALB2 expression vector were processed for Flag immunoprecipitation followed by mass spectrometry (MS). Representative MS/MS spectra of tryptic diglycine (diG)-PALB2 peptides identified are shown (K16, top; K43, bottom), c, Schematic of the /acO/LacR chromatin-targeting system and the in vivo quantification of ubiquitylated PALB2. d, Representative micrographs of U20S 256 cells transfected with the
indicated mCherry-LacR-PALB2 vectors. Cells were processed for FK2 immunofluorescence. EV, empty vector. Scale bar, 5 μιη. e, Quantification of U20S 256 cells transfected with the indicated mCherry-LacR-PALB2 vectors. Cells were processed for quantification of FK2 fluorescence at the LacO focus. Each circle represents a cell analyzed and the bar is at the median (N = 3). Cells were also stained with a cyclin A antibody to determine cell cycle position. Statistical significance was determined by a Kruskall-Wallis test (***P < 0.001 ; **P < 0.01).
[00066] Figure 9. Analysis of KEAP1- and USP1 1 -dependent modulation of PALB2 and homologous recombination, a, Site-specific chemical ubiquitylation of HA-PALB2 (1-103) at residue 20 (PALB2-KC20-Ub) and 45 (PALB2-KC45-Ub) was carried out by dichloroacetone linking. The resulting ubiquitylated PALB2 polypeptides along with their unmodified counterparts were subjected to pulldown with a fusion of MBP with the coiled-coil domain of BRCA1 (MBP-BRCA1 -CC). I, input; PD, pulldown. Asterisk indicates a non-specific band, b, Wild-type and ΚΕΑΡΙΔ 293T cells were treated with cycloheximide (CHX) for the indicated time and then processed for NRF2 and KEAP1 immunoblotting. Actin levels were also determined as a loading control, c, Immunoprecipitation (IP) of USP1 1 from extracts prepared from 293T cells that were or were not treated with camptothecin (CPT; 200 nM). Immunoprecipitation with normal IgG was performed as a control, d, U20S DR-GFP cells were transfected with the indicated siRNAs. Twenty-four hours post-transfection, cells were further transfected with the indicated siRNA-resistant USPl l expression vectors (WT, wild type; CS, C318S and CA, C318A catalytically dead mutants) or an empty vector (EV), with or without an I-Scel expression vector. The percentage of GFP-positive cells was determined 48h post-plasmid transfection for each condition and was normalized to the I-Scel plus non-targeting (siCTRL) condition (mean ± s.d., N = 3). e, Schematic representation of human USPll (top) and KEAP1 (bottom) gene organization and targeting sites of sgRNAs (as described in Figure 5a) used to generate the USPll A and USPl l A IKE API A 293T cells. The indels introduced by the CRISPR-Cas9 and their respective frequencies are indicated. The USPl l knockout was created first and subsequently used to make the USPll Δ ΙΚΕΑΡΙΔ double mutant, f, Immunoprecipitation of PALB2 from extracts prepared from 293T cells transfected with the indicated siRNA and with or without CPT (200 nM) treatment. Immunoprecipitation with normal IgG was performed as a control.
[00067] Figure 10. USPl l antagonizes KEAP1 action on PALB2. a, U20S DR-
GFP cells were transfected with the indicated siRNAs or left untransfected ( - ). Twenty-four hours post-transfection, cells were transfected with an I-Scel expression vector (circle). The percentage of GFP-positive cells was determined 48 h post-plasmid transfection for each condition and was normalized to the I-Scel plus non-targeting (CTRL) condition (mean ± range, N = 3). b, Parental 293T cells (wild type (WT)) or a USPll A derivative were transfected with the indicated GFP-PALB2 constructs, treated with CPT and processed for GFP immunoprecipitation (IP), c, Parental 293T cells (wild type) or a USPll A derivative were transfected with an empty vector (EV) or the indicated PALB2 expression vectors. Sensitivity of the cells to the PARP inhibitor olaparib was then determined by a clonogenic survival assay (mean ± s.d., N=3).
[00068] Figure 11. Characterization of USPl l protein stability a, U20S cells synchronized in Gl or S/G2 were treated with cyclohexamide (CHX) and processed at the indicated time points to monitor USPl l stability, b, Immunoprecipitation (IP) of PALB2 from extracts prepared from 293T cells that were synchronized in Gl or S phase and treated or not with IR (20 Gy). c, U20S cells were irradiated with a dose of 2 or 20 Gy and processed for USPl l immunoblotting at the indicated times post-IR. Actin was used as a loading control, d, U20S cells, mock-treated or incubated with the ATM inhibitor KU55933 (ATMi), ATR inhibitor VE-821 (ATRi) or DNA-PKcs inhibitor NU7441 (DNAPKi), were irradiated (20 Gy) and processed for USPl l and actin (loading control) immunoblotting. e, Similar experiment to d except that cells were exposed to ultraviolet (UV) radiation (50 mJ/cm" ). f, U20S cells, mock-treated or incubated with the proteasome inhibitor MG132, were irradiated (20 Gy) and processed for USPl 1 and actin (loading control) immunoblotting. g, U20S cells, mock-treated or incubated with the Cullin inhibitor MLN4924, were irradiated (20 Gy) and processed for USPl 1 and actin (loading control) immunoblotting.
[00069] Figure 12. Reactivation of RAD51 loading and unscheduled DNA synthesis in Gl . a, 53BP1A U20S cells were transfected with the indicated siRNA, synchronized in Gl or S/G2 by release from a double-thymidine block and irradiated (20 Gy) before being processed for fluorescence microscopy. DAPI was used to trace the nuclear boundary and cyclin A staining was used to determine cell cycle position. The percentage of cells with more than five γ -H2AX-colocalizing PALB2 foci is indicated as the mean ± s.d., N = 3. Scale bar, 5 μηι. b, Representative micrographs of irradiated Gl -synchronized wild-type (WT) and 53BP1A U20S cells transfected with the indicated siRNA and expressing wild-type CtlP. c, Representative micrographs of irradiated Gl -synchronized wild-type U20S cells transfected with the indicated siRNA and expressing CtIP(T847E). d, U20S 53BP1A cells were synchronized in G l , supplemented with BrdU, irradiated (2 Gy) and processed for γ -H2AX and BrdU immunofluorescence. The percentage of cells with more than five y-H2AX-colocalizing BrdU foci is indicated (mean ± s.d., N = 3). e, Micrograph of a U20S cell targeted with the CRISPR-mClover system showing the typical perinuclear expression pattern of lamin A. f, Micrograph of a U20S cell targeted with the mClover system showing an expression pattern characteristic of subnuclear PML foci, g, Timeline of the gene- targeting (LMNA) experiment presented in Figure 4d. h, Timeline of the gene targeting (LMNA or PML) experiment presented in Figure 4e and Figure 13.
[00070] Figure 13. Analysis of homologous recombination in Gl . a, Quantitation of gene targeting efficiency at the LMNA locus in asynchronously dividing U20S cells transfected with increasing amount of donor template and with (grey) or without (white) sgRNAs. Gene-targeting events were detected by flow cytometry (mean ± s.d., N > 3). b, Quantitation of gene-targeting efficiency at the LMNA locus in asynchronously dividing cells transfected with the indicated siRNA. Gene-targeting events were detected by flow cytometry (mean ± s.d., N = 3). c, Gene-targeting efficiency at the PML locus measured by flow cytometry in Gl -arrested 53BP1A U20S cells expressing the CtIP(T847E) mutant and co-transfected with the indicated siRNA or a PALB2-KR expression construct (mean ± s.d., N = 3). d, Gene-targeting efficiency at the LMNA locus measured by flow cytometry in Gl -arrested parental (wild-type (WT)) and 53 BP 1 A U20S cells transfected with KEAPl siRNA and expressing the CtIP(T847E) mutant (mean ± s.d., N = 3). e, Gene-targeting efficiency at the LMNA locus measured by flow cytometry in Gl -arrested parental (wild-type) and 53BP1A U20S cells transfected with the indicated siRNA and expressing either wild type or the CtIP(T847E) mutant (mean ± s.d., N= 3).
[00071] Figure 14 Identification of DCAF10 as a regulator of USP1 1 stability in response to DNA damage, a. siRNA screen where U20S cells were transfected with siRNAs targeting known and predicted DCAFs along with other CUL4-interacting proteins. Cells were either irradiated with IR (20 Gy) or UV (50 J/m-2), let to recover for 3 h and then processed for USP1 1 immunofluorescence. Each point plotted corresponds to the percentage of USP1 1 left after irradiation. The red dots correspond to the siRNA non-targeting controls (CTRL) and targeting USP11, whereas the red dots correspond to core CRL4 factors, that include CUL4 itself. D.U20S cells were transfected with the indicated siRNAs and then irradiated with a dose of 20 Gy and processed for USP1 1 immunoblotting at the indicated times post-ionizing radiation. Actin was used as a loading control.
[00072] Figure 15. Validation of DCAF10 as a regulator of USP1 1. a. DCAF10 interacts with USP1 1. Immunoprecipitation (IP) of Flag-USPl l from extracts prepared from 293 Flp-I /T-Rex cells. Cells were probed with DCAF10 and DCAF15 antibodies, b. Whole cell extracts of mouse embryo fibroblasts (MEFs) of the indicated genotypes were processed for USP1 1 immunoblotting. Tubulin was used as a loading control, c. U20S DR-GFP cells were transfected with the indicated siRNAs or expression vectors. Twenty-four hours post-transfection, cells were transfected with an I-Scel expression vector. The percentage of GFP -positive cells was determined 48 h post-plasmid transfection for each condition and was normalized to the I-Scel plus non- targeting (CTRL) + empty vector (EV) condition.
[00073] Figure 16. KEAP1 inhibition can activate HR in Gl cells. Gene targeting at the LMNA locus in Gl -arrested cells transfected with the indicated siRNA and vectors expressing either the Rl KEAP1 inhibitor or its FN3 scaffold control (mean ± s.d., N = 3).
DETAILED DESCRIPTION OF THE INVENTION
[00074] The preparation and use of the agents disclosed as well as the practice of the methods herein employed, unless otherwise indicated, utilize conventional techniques in molecular biology, biochemistry, chromatin structure and analysis, computational chemistry, cell culture, recombinant DNA and related fields as are within the skill of the art. The techniques are fully disclosed in the literature. [See, for example, Sambrook et al. Molecular Cloning: A Laboratory Manual, Second edition, Cold Spring Harbor Laboratory Press, 1989 and Third edition, 2001 ; Ausubel et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1987 and periodic updates; the series Methods in Enzymology, Academic Press, San Diego; Wolffe, Chromatin Structure and Function, Third edition, Academic Press, San Diego, 1998; Methods in Enzymology, Vol. 304, "Chromatin" (P.M. Wassarman and A. P. Wolffe, eds.), Academic Press, San Diego, 1999; and Methods in Molecular Biology, Vol. 1 19, "Chromatin Protocols" (P. B. Becker, ed.) Humana Press, Totowa, 1999].
[00075] Unless defined otherwise, 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. The following definitions supplement those in the art and are directed to the present application and are not to be imputed to any related or unrelated case. Although any methods and materials similar or equivalent to those described herein can be used in the practice of the invention, particular materials and methods are described herein.
[00076] As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. As used herein, the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
[00077] A "gene editing system" is a system for targeting and editing genomes, including without limitation, a TALEN (Transcription Activator-Like Effector Nucleases) system, a CRISPR (Clustered Regulatory Interspaced Short Palindromic Repeats) system and a Zinc-Finger Nucleases (ZFN) system. (See Nemudryi A. A. et al, Acta Naturae. 2014 Jul-Sep; 6(3): 19-40 for a review of TALEN and CRISPR systems; Gaj T. et al, Trends Biotechnol. 2013 Jul; 31 (7): 397-405 for a review of TALEN, CRISPR and ZFN systems; US Published Patent Application No. 201 10145940 describing a TALEN system; and Bibikova M., et al, Genetics. 2002;161(3): 1 169— 1175; Townsend J.A., et al, Nature 2009;459(7245):442-445; Zhang F., et al, Proc. Natl. Acad. Sci. USA. 2010; 107(26): 12028-12033; Torikai FL, et al; Blood. 2012; 119(24):5697-5705; Provasi E., et al, I. Nat. Med. 2012; 18(5): 807-8151, and Lombardo A., et al, Nat. Methods. 201 1 ;8(10):861— 869 describing ZFN systems).
[00078] A "CRISPR system" generally refers to transcripts and other elements involved in the expression of, or directing the activity of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated ("Cas") genes. A CRISPR system may include without limitation, sequences encoding a Cas gene, a tracr (trans- activating CRISPR) sequence (e.g. tracrRNA or an active partial tracrRNA), a tracr- mate sequence, a guide sequence, or other sequences and transcripts from a CRISPR locus. One or more elements of a CRISPR system may be derived from a type I, type II, or type III CRISPR system. A CRISPR system promotes the formation of a CRISPR complex (comprising a guide sequence hybridized to a target sequence and complexed with one or more Cas proteins) at the site of a target sequence. A "target sequence" or "target polynucleotide" refers to a sequence which is sufficiently complementary to a designed guide sequence that the target sequence hybridizes to the guide sequence promoting the formation of a CRISPR complex. A target sequence may comprise any polynucleotide, such as DNA or RNA polynucleotides, and it may be located in the nucleus, cytoplasm, or an organelle, for example, mitochondria or chloroplast. In the context of an endogenous CRISPR system, formation of a CRISPR complex in an endogenous CRISPR system results in cleavage of one or both strands in or near (e.g. within 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, or more base pairs from) the target sequence.
[00079] CRISPR systems are described in U.S. Pat. Nos. 8,697,359, 8,771 ,945, 8,795,965, 8,865,406, 8,871,445, 8,889,356, 8,889,418 and 8,895,308; US Patent Publications US 2014-0310830, US 2014-0287938, US 2014-0273234, US2014- 0273232, US 2014-0273231 , US 2014-0256046, US 2014-0248702), US 2014- 0242700, US 2014-0242699, US 2014-0242664, US 2014-0234972, US 2014- 0227787, US 2014-0189896, US 2014-0186958, US 2014-0186919, US 2014-0186843, US 2014-0179770 and US 2014-0179006, US 2014-0170753, and US 20150232883; European Patent Applications EP 2771468 (EP13818570.7), EP 2764103 (EP13824232.6), and EP 2784162 (EP14170383.5); and PCT Patent Publications WO2014/093661 (PCT/US2013/074743), WO2014/093694 (PCT/US2013/074790), WO2014/093595 (PCT/US2013/07461 1), WO2014/093718 (PCT/US2013/074825), WO2014/093709 (PCT/US2013/074812), WO2014/093622 (PCT/US2013/074667), WO2014/093635 (PCT/US2013/074691), WO2014/093655 (PCT/US2013/074736), WO2014/093712 (PCT/US2013/074819), WO2014/093701 (PCT/US2013/074800), WO2014/018423 (PCT/US2013/051418) and WO2014/093622 (PCT/US2013/074667). General information on CRISPR-Cas Systems is also described in the following publications: Cong, L., et al, Science, February 15; 339(6121):819-23 (2013); Jiang W., et al., Nat Biotechnol March; 31(3):233-9 (2013); Wang H., et al, Cell May 9; 153(4):910-8 (2013); Konermann S, et al, Nature. 2013 Aug. 22; 500(7463):472-6. doi: 10.1038/Nature 12466. Epub 2013 Aug. 23; Ran, F A., et al, Cell August 28. pii: S0092- 8674(13)01015-5. (2013); Hsu, P., et al, Nat Biotechnol doi: 10.1038/nbt.2647 (2013); Ran, F A., et al, Nature Protocols November; 8(1 1 ):2281 -308. (2013); Shalem, O., et al, Science December 12. (2013). [Epub ahead of print]; Nishimasu, H., et al, Cell Feb. 27. (2014). 156(5):935-49; Wu X., et al, Nat Biotechnol. (2014) Apr. 20. doi: 10.1038/nbt.2889; Piatt et al, Cell 159(2): 440-455 (2014) DOI: 10.1016/j.cell.2014.09.014; Hsu et al. Cell 157, 1262-1278 (Jun. 5, 2014) (2014); Wang et al, Science. 2014 Jan. 3; 343(6166): 80-84. doi: 10.1 126/science. 1246981 ; Doench et al., Nature Biotechnology published online 3 Sep. 2014; doi: 10.1038/nbt.3026; Storrs, The Scientist, Article No. 39239, March 1 , 2014; and Swiech et al, Nature Biotechnology; published online 19 Oct. 2014; doi: 10.1038/nbt.3055). Several programs are available to design guide sequences, for example, MIT's CRISPR Design [http://crispr.mit.edu] and E-CRISP [www.e-crisp.org/E-CRISP] developed by the German Cancer Research Center. CRISPR systems also include the systems developed by or available from Editas Medicine (Cambridge, MA), Caribou Biosciences (Berkeley, CA), CRIPSR Therapeutics (Basel, Switzerland), Addgene (Cambridge, MA) and Intellia Therapeutics (Cambridge, MA).
[00080] '"DNA end resection" generally refers to nucleolytic degradation of the 5'-terminated strand of a DNA double- stranded break leading to the formation of 3'- terminated single- stranded DNA. DNA end resection in eukaryotes comprises two phases: a slow initial phase, catalyzed by the Mrel l-Rad50-Nbsl (MRN) complex in mammals, and a second and faster phase catalyzed by the exonuclease Exol or the helicase Bloom Syndrome Protein (BLM). DNA end resection is initiated by a cell cycle activation step comprising phosphorylation of the accessory protein CtIP (also known as retinoblastoma binding protein 8). Pathways involved in DNA end resection may be activated by stimulating or activating BRCA1 recruitment to DNA double- strand breaks by inhibiting TP53BP1 (53BP1) or RIF, or blocking recruitment of 53BP1 or RIF to DNA double-stranded break sites. In an aspect, DNA end resection may be activated by inhibiting 53BP1 (or RIF) expression and/or activity and expressing a mutated form of CtIP that mimics constitutive phosphorylation, for example CtIP-Thr879Glu. In an aspect, DNA end resection is reconstituted or activated using inhibitors of 53BP1 and a mutated form of CtIP that mimics constitutive phosphorylation, in particular CtIP-Thr879Glu. In an aspect, DNA end resection may be reconstituted or activated using purified human proteins: Bloom helicase (BLM); DNA2 helicase/nuclease; Exonuclease 1 (EXOl ); the complex comprising MRE11 , RAD50, and NBSl (MRN); and Replication protein A (RPA.) (See Nimonkar A.V. et al, Genes & Development 25:350-362, 201 1 ; Huertas, P, Nat Struct Mol Biol, 17(10: 1 1-16, doi: 10.1038/nsmb. l710, 2010; Jimeno S., et al, Nucl. Acids Res doe: 101093/nar/gkui384, 2015 for descriptions of DNA end resection).
[00081] "Homologous recombination" and "HR" refer to a type of genetic recombination in which DNA strands of similar or identical nucleotide sequences are exchanged. HR can be used by cells to repair DNA double-strand breaks (DSB) by the following general steps. HR is initiated when the DSB is resected by nucleases and helicases, generating 3' single-stranded DNA (ssDNA) overhangs onto which the RAD51 recombinase assembles as a nucleoprotein filament. This structure can invade homologous duplex DNA, which is used as a template for repair DNA synthesis. The resulting intermediates can be metabolized to yield non-crossover products thereby restoring the damaged DNA molecule as it existed before the double-strand break (San Filippo et al., Annu. Rev. Biochem. 2008. 77:229-57). The terms also include recombination using single-stranded donor oligonucleotides (ssODNs), in particular recombination using single-stranded donor oligonucleotides (ssODNs) requiring resection and which may be activated by 53BP1 inhibitors.
[00082] "HR Disease" refers to any disorder, disease, condition, syndrome or combination of manifestations or symptoms recognized or diagnosed as a disorder which may be associated with or characterized by a HR defect. Exemplary diseases include, for example, cancer, cardiovascular diseases including heart failure, hypertension and atherosclerosis, respiratory diseases, renal diseases, gastrointestinal diseases including inflammatory bowel diseases such as Crohn's disease and ulcerative colitis, hepatic, gallbladder and bile duct diseases, including hepatitis and cirrhosis, hematologic diseases, metabolic diseases, endocrine and reproductive diseases, including diabetes, bone and bone mineral metabolism diseases, immune system diseases including autoimmune diseases such as rheumatoid arthritis, lupus erythematosus, and other autoimmune diseases, musculoskeletal and connective tissue diseases, including arthritis, achondroplasia infectious diseases and neurological diseases such as Alzheimer's disease, Huntington's disease and Parkinson's disease.
[00083] Methods of the invention may be used to monitor or treat a disease caused by a defect in a gene that mediates homologous recombination, for example, BRCA1 , BRCA2, PALB2, PARP-1 , USP11 , RAD51 , and/or DCAF10.
[00084] Embodiments of the invention provide for monitoring or treatment of various cancers including but not limited to carcinomas, melanomas, lymphomas, sarcomas, blastomas, leukemias, myelomas, osteosarcomas, neural tumors, and cancer of organs such as the breast, ovary, and prostate.
[00085] In embodiments, the invention provides for monitoring or treatment of cancer with BRCA-1 defects, BRCA-2 defects, dual BRCA-l/BRCA-2 defects, and Fanconi anemia. In embodiments of the invention, the cancer is breast cancer, in particular invasive ductal carcinoma and invasive lobular carcinoma. In embodiments of the invention, the cancer is ovarian cancer, in particular epithelial ovarian tumors, germ cell ovarian tumors, and sex cord stromal tumors.
[00086] Methods of the invention for activating or modulating homologous recombination may be used to genetically modify polynucleotides associated with a genetic disorder. In some embodiments, the genetic disorder is a monogenetic disorder. In some embodiments, the genetic disorder is a multigenetic disorder. In some embodiments, the genetic disorder is associated with one or more SNPs. In particular embodiments of the invention, the genomic modification corrects a point mutation.
[00087] Examples of genetic disorders and polynucleotide sequences associated with the genetic disorders may be found on the World Wide Web (see for example, the National Center for Biotechnology Information, National Library of Medicine (Bethesda, MA) or the McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University (Baltimore, Md)), listed in published patents and applications (see, for example, US Published Application No. 2015/0247150), and in publications (see for example, Turitz Cox D.B. et al, Nature Medicine 21 , 121 -131 , 2015; and O'Connor T.P. and R.G. Crystal, Nature Reviews/Genetics Volume 7, April 2006, pages 261 -276 including Supplementary Information, and publications cited therein).
[00088] In an aspect, the genetic disorder is a genetic disorder of muscle. In an aspect, the genetic disorder is myotonic dystrophy type 1. In an aspect, the genetic disorder is myotonic dystrophy type 2. In an aspect, the genetic disorder is Duchenne muscular dystrophy (DMD). In an aspect, the genetic disorder is Becker muscular dystrophy.
[00089] In an aspect, the genetic disorder is a genetic disorder of the liver, for example, alpha- 1 antitrypsin deficiency, Wilson Disease, hereditary hemochromatosis, Type I tyrosinemia, glycogen storage disease Type IV, argininosuccinate lyase deficiency, citrin deficiency, cholesterol ester storage disease and hereditary fructose intolerance. [00090] In an aspect, the genetic disorder is alpha- 1 antitrypsin deficiency which is an autosomal recessive (codominant) disease due to mutations in the SERPINA1 gene that encodes the serine protease inhibitor AAT.
[00091] In an aspect, the genetic disorder is Wilson disease which depends on mutations in the gene encoding the ATP7B Cu translocase, a protein mainly expressed by the hepatocyte that regulates the levels of copper in the liver.
[00092] In an aspect, the genetic disorder is a genetic disorder of the lungs.
[00093] In an aspect, the genetic disorder is cystic fibrosis, an autosomal recessive disease caused by mutations of the Cystic Fibrosis Transmembrane Regulator (CFTR) protein, a member of the ATP -binding cassette superfamily of transmembrane proteins.
[00094] In other aspects of the invention the genetic disorder may be heamophilia, a 1 -antitrypsin deficiency, Canavan disease, Adenosine deaminase deficiency, X-linked severe combined immunodeficiency, familial amyloidotic polyneuropathy, thalassemia, Tay-Sachs disease, late infantile ceroid lipofuscinosis, mucopolysaccharidosis, Niemann-Pick disease, achondroplasia, Huntington disease, spino-cerebellar ataxia, Fredriech ataxia, Amyotrophic Lateral Sclerosis, monogenic hypercholesterolemia and other monogenic disorders.
[00095] In aspects of the invention the genetic disorder is sickle cell anemia and a method of the invention comprises correcting the mutated HBB hemoglobin gene by gene conversion with its paralog HBD.
[00096] An "effective amount" refers to an amount of a compound or composition, as described herein effective to achieve a particular biological result. Such results include, without limitation, the treatment of a disease or condition disclosed herein as determined by any means suitable in the art.
[00097] "PARP Inhibitor" refers to an inhibitor of the nuclear enzyme poly(adenosine 5'-diphospho-ribose) polymerase ["poly(ADP-ribose) polymerase" or "PARP", which is also referred to as ADPRT (NAD:protein (ADP-ribosyl transferase (polymerising)) and PARS (poly(ADP-ribose) synthetase). PARP inhibitors have been described in Banasik et al., "Specific Inhibitors of Poly(ADP-Ribose) Synthetase and Mono(ADP-Ribosyl)-Transferase", J. Biol. Chem., 267:3, 1569-75 (1992), and in Banasik et al, "Inhibitors and Activators of ADP-Ribosylation Reactions", Molec. Cell. Biochem., 138, 185-97 (1994). PARP inhibitors have been disclosed and described in the following patents and patent applications: WO 00/42040; WO 00/39070; WO 00/39104; WO 99/1 1623; WO 99/1 1628; WO 99/11622; WO 99/59975; WO 99/1 1644; WO 99/1 1945; WO 99/1 1649; and WO 99/59973; US Patent No. 8,894,989, US Patent No. 8,946,221 ; 8,778,966; 8,669,249; 8,623,884; 8,592,416; 8,546,368; 8,541 ,417;
8,541,403 8,420,650 8,362,030; 8,236,802 8,217,070; 8,188,103 8,188,084; 8,183,250 8,173,682 8,129,382; 8,088,760 8,080,557; 8,071 ,623 8,058,275;
8,012,976 8,008,491 7,999, 117; 7,956,064 7,875,621 ; 7,820,668 7,750,008;
7,732,491 7,728,026 7,652,014; 7,601 ,719 7,462,724; 7,087,637 7,041 ,675;
6,977,298 6,924,284 6,737,421 ; 6,635,642 6,495,541 ; 6,444,676 6,395,749;
6,380,211 6,380,193; 6,346,536; 6,197,785; 5,756,510; and Re. 36,397.
[00098] In aspects of the invention, the PARP inhibitor is Olaparib
(AstraZeneca). In aspects of the invention, the PARP inhibitor is Veliparib (AbbVie Inc, Chicago, IL). In aspects of the invention, the PARP inhibitor is Rucaparib (Clovis Oncology, Inc., Boulder, CO). In aspects of the invention, the PARP inhibitor is ΓΝΟ- 1001 (Inotek Pharmaceuticals Corp, Lexington, MA). In aspects of the invention, the PARP inhibitor is MK-4827 (niraparib) (Tesaro, Waltham, MA, also see Montoni et al, Frontiers in Pharmacology, [4], Article 18, pages 1 -7). In aspects of the invention, the PARP inhibitor is talazoparib (Medivation, Inc, San Francisco CA).
[00099] A "sample" is a sample derived from any biological source, such as tissues, extracts, or cell cultures, including cells (e.g. tumor cells), cell lines, cell lysates, and physiological fluids, such as, for example, blood or subpopulations thereof (e.g. white blood cells, erythrocytes), plasma, serum, saliva, ocular lens fluid, cerebrospinal fluid, sweat, urine, fecal matter, tears, bronchial lavage, swabbings, milk, ascites fluid, nipple aspirate, needle aspirate, synovial fluid, peritoneal fluid, lavage fluid, and the like. The sample can be obtained from animals, preferably mammals, most preferably humans. Samples can be from a single individual or pooled prior to analysis. The sample can be treated prior to use, such as preparing plasma from blood, diluting viscous fluids, and the like. Methods of treating samples can involve filtration, distillation, extraction, centrifugation, concentration, inactivation of interfering components, the addition of reagents, and the like.
[000100] In embodiments of methods of the invention, the sample is a mammalian tissue sample. In another embodiment the sample is a cell lysate. In another embodiment the sample is a cell. In another embodiment the sample is a human physiological fluid. In a particular embodiment, the sample is human serum. In a further embodiment, the sample is white blood cells or erythrocytes. [000101] The terms "subject", "individual" or "patient" refer, interchangeably, to a warm-blooded animal such as a mammal. In particular, the terms refer to a human. A subject, individual or patient may be afflicted with or suspected of having or being predisposed to a disease as described herein. The term also includes animals bred for food, as pets, or for study including horses, cows, sheep, poultry, fish, pigs, cats, dogs, and zoo animals goats, apes (e.g. gorilla or chimpanzee), and rodents such as rats and mice.
[000102] NCBI Accession Numbers for USPl l , PALB2, BRCA1 , BRCA2, KEAP1 , 53BP1 , DCAFIO, RBX1 , CUL3 and CtIP are in Table 1 and the human sequences for same are in the Sequence Listing.
Screening and Monitoring Assays
[000103] The invention provides a method for monitoring activity or expression of USPl l by assaying the interaction of BRCA1 and PALB2, the interaction of BRCA1 , PALB2 and BRCA2, the interaction of USPl l and DCAFIO, and/or the interaction of USP11 and PALB2. Routine methods known to persons skilled in the art can be used to assay protein interactions in a sample. For example, BRCA1-PALB2, BRCA1-PALB2- BRCA2, USPl l-DCAFlO, or USP11 -PALB2 complexes may be isolated using affinity techniques such as for example immunologically-based purification (e.g. immunoaffmity chromatography), peptides may be prepared from the isolated complexes using conventional methods (e.g. gel electrophoresis, liquid chromatography, capillary electrophoresis, nano-reversed phase liquid chromatography, high performance liquid chromatography, or reverse phase high perfonnance liquid chromatography), and the peptides or peptide fragments may be subjected to mass spectrometry (e.g., quantitative mass spectrometry such as selected reaction monitoring mass spectrometry (sMRM), high resolution data independent analyses (SWATH), high resolution multiple reaction monitoring (MRMHR) or MSI based quantitation).
[000104] The invention also provides a method for monitoring activity of USPl l by assaying ubiquitylation of the N-terminus of PALB2. Routine methods known to persons skilled in the art can be used to assay ubiquitination in a sample. For example, ubiquitination or PALB2 may be assayed by measuring changes in PALB2 (e.g., weight; see US Patent No. 6,413,725), the amount of poly-ubiquitin bound to CRL3- KEAP1 E3 ligase (see for example, EP 1268847), and/or the activity of CRL3-KEAP1 E3 ligase (see for example, US Publication No. 2013/01 16152). Mass spectrometry techniques such as selected reaction monitoring mass spectrometry (sMRM), high resolution data independent analyses (SWATH), high resolution multiple reaction monitoring (MRM ) or MSI based quantitation) can also be used to monitor ubiquitin remnants on peptides from the PALB2 N-terminus following protease digestion. In a more specific example, preparation of isotopically labeled synthetic peptides corresponding to tryptic digests of ubiquitylated PALB2, especially those that correspond to ubiquitylation on Lysl4, Lyl 6, Lys20, Lys25, Lys30, Lys43 or Lys45 can be used as internal standards to quantitate the extent of PALB2 ubiquitylation.
[000105] In an aspect, the invention provides a method for assaying ubiquitylation of PALB2 polypeptides in a sample, the method comprising digesting ubiquitinated PALB2 polypeptides in the sample with a protease, thereby generating a plurality of test peptides; determining the presence of at least one isopeptide bond between ubiquitin and a lysine residue of the test peptides by mass spectrometry to determine the numbers of ubiquitination sites and thereby the amount of ubiquitination of PALB2 polypeptides in the sample. In an embodiment, the test peptides are from the PALB2 N- terminus. In an embodiment, the lysine residue corresponds to Lysl4, Lysl6, Lys20, Lys25, Lys30, Lys43 or Lys45. The method may utilize peptide internal standards corresponding to different peptide subsequences of PALB2 to provide for controls in a quantitative assay. In one aspect, different synthetic peptide internal standards corresponding to PALB2 are generated and differentially labeled.
[000106] Proximity ligation assays (PLA) may also be used to assay activity of USP1 1 by assaying the interaction between BRCA1 and PALB2 and/or PALB2- interacting proteins such as BRCA2, using DNA-based detection. For example, primary antibodies against binding partners of an interaction (e.g., PALB2 and BRCA1) are added to a cell lysate. A second set of antibodies, termed PLA probes or proximity probes, recognize the first set of primary antibodies. The PLA probes contain DNA strands that assemble into an assay-specific DNA molecule when in close proximity. This DNA molecule can then be amplified and detected using, for example, fluorescent probes. [See, for example, Soderberg O. et al., Nat. Methods., 2006 December; 3(12):995-1000; Jarvius M. et al., Mol. Cell. Proteomics, 2007 September; 6(9): 1500- 9)]·
[000107] In an aspect, the invention provides a method for assaying BRCA1- PALB2 or BRCA1-PALB2-BRCA2 interactions in a sample comprising: contacting the sample with primary antibodies to each binding partner in the interaction; contacting the sample with proximity probes comprising a secondary antibody that binds to a corresponding primary antibody, wherein each proximity probe has an oligonucleotide conjugated thereto; wherein when the oligonucleotides of the proximity probes are in sufficient proximity to each other, the oligonucleotides of the proximity probes interact to form circular products that are amplified by rolling circle replication producing amplification products; and, measuring the amplification products to thereby assay or measure the interactions.
[000108] Assays that monitor PALB2 (or associated proteins such as BRCA2) in situ co-localization with BRCA1 can also provide a method for monitoring USP1 1 activity (see Example herein). For example, PALB2 localization at sites of DNA damage (marked by BRCA1 or other markers such as γ-Η2ΑΧ) is dependent on USP1 1 activity. In such assays, cells are fixed, permeabilized and then incubated with antibodies that detect PALB2, BRCA2 or their associated proteins (e.g., BRCA1). Addition of labeled secondary antibodies enable the in situ visualization of protein accumulation at DNA damage sites in subnuclear structures termed foci. Addition of a genotoxic insult (such as ionizing radiation or other clastogenic treatments) increases the number of "foci" and can be included to augment the dynamic range of the assay.
[000109] It will be appreciated that proximity ligation assays and in situ co- localization assays may be used to assay any of the interactions disclosed herein.
[000110] The methods of the invention may be performed in the presence or absence of a test compound or agent and detection of an increase or decrease in activity or expression of one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAFlO complex, CRL3-KEAP1 complex, CRL3-KEAP1-PALB2 complex, and KEAP1- PALB2 complex, as compared to a control in the absence of the test compound or agent indicates that the test compound or agent may be useful as a therapeutic agent, or for modulating homologous recombination.
[000111] In an aspect, the invention provides a method for identifying or evaluating an agent for its ability to sensitize or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on USP11 activity using a method of the invention. In an aspect, a negative effect on USP1 1 indicates that the agent is a sensitizer of cells to PARP inhibitors or can reverse or delay emergence of resistance to PARP inhibitors. In an aspect, a positive effect on USP1 1 indicates that the agent is a poor sensitizer of cells to PARP inhibitors. In an aspect, the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by decreased levels of USP1 1 activity when compared to such levels obtained from a control. In an aspect, the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by increased levels of USP1 1 activity when compared to such levels obtained from a control.
[000112] The invention also relates to a method of identifying or evaluating an agent for its ability to sensitize cells or reverse or delay emergence of resistance to PARP inhibitors by determining the effect of the agent on KEAP1 , CRL3-KEAP1 , or KEAP1-PALB2. In an aspect, a negative effect on KEAP1 (loss of KEAP1 or CRL3- KEAP1 activity) indicates that the agent is a poor sensitizer of cells to PARP inhibitors. In an aspect, a positive effect on KEAP1 or CRL3-KEAP1 activity indicates that the agent is a sensitizer of cells to PARP inhibitors or can reverse or delay emergence of resistance to PARP inhibitors. In an aspect, the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by increased levels of KEAP1 , CRL3-KEAP1 , KEAP1-PALB2 or CRL3-KEAP1 activity or expression when compared to such levels obtained from a control. In an aspect, the ability of an agent to sensitize or reverse or delay emergence of resistance to PARP inhibitors is determined by decreased levels of KEAP1 , CRL3-KEAP1 , KEAP1-PALB2 or CRL3- KEAP1 activity or expression when compared to such levels obtained from a control.
[000113] Still further the present invention contemplates methods of detecting an anti-cancer agent comprising performing a test assay comprising contacting an immortalized cell with a test compound and measuring USP11 activity or CRL3- KEAP1 using a method of the invention and comparing to a control test assay in the absence of the test compound. In an aspect, USP1 1 activity is assayed by measuring USP1 1 , PALB2, DCAF10, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP11-DCAF10 complex, CRL3-KEAP1 and/or CRL3-KEAP1 -PALB2 complex in the cell. In an aspect, detecting a negative effect of the agent on USP11 activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer. In an aspect, detecting a negative effect of the agent on BRCA1-PALB2-BRCA2 complex and/or PALB2- USP1 1 complex activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer. In an aspect, detecting a positive effect of the agent on KEAP1 , CRL3-KEAP1 and/or CRL3 -KEAP 1 -P ALB2 complex activity or expression as compared with a control indicates a potential anti-cancer agent or PARP inhibitor sensitizer. In an aspect, decreased levels of USP11 activity when compared to such levels obtained from a control are indicative that agent has anticancer activity or is a PARP inhibitor sensitizer.
[000114] The invention provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination comprising determining the effect of a test compound or agent on one or more of USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 -PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 complex and CRL3-KEAP1-PALB2 complex. In an aspect, the invention provides a method for identifying or evaluating an agent for its ability to modulate homologous recombination in a cell comprising (i) assaying in a sample USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1 - PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP1 1 -DCAF10 complex, CRL3-KEAP1 and/or CRL3-KEAP1 -PALB2 complex in the cell in the presence or absence of the agent, and (ii) detecting an increase or decrease in USP1 1 , DCAF10, PALB2, PALB2 ubiquitylation, BRCA1-PALB2-BRCA2 complex, PALB2- USP1 1 complex, KEAP1, USP11 -DCAF10 complex, CRL3-KEAP1 complex and/or CRL3-KEAP1 -PALB2 complex in the sample compared to a control as an indication of the ability of the agent to modulate homologous recombination.
[000115] The invention provides a method of screening for a therapeutic agent for treatment of a disease associated with defects in HR (i.e., HR Disease), comprising identifying an agent that disrupts or modulates one or more of USP1 1 , PALB2, PALB2 ubiquitylation, DCAF10, BRCA1 -PALB2-BRCA2 complex, PALB2-USP1 1 complex, KEAP1 , USP11 -DCAF10 complex, CRL3-KEAP1 or CRL3 -KEAP 1 -P ALB2 complex. In an aspect, detecting a positive effect of the agent on USP1 1 , BRCA1-PALB2- BRCA2 complex and/or, PALB2-USP1 1 complex activity or expression as compared with a control indicates a potential therapeutic agent for treatment of a HR Disease. In an aspect, detecting a negative effect of the agent on, DCFA10, KEAP1 , CRL3-KEAP1 or CRL3-KEAP1-PALB2 complex activity or expression as compared with a control indicates a potential therapeutic agent for treatment of a HR Disease.
[000116] Test compounds used in the methods of the invention can be any product in isolated form or in a mixture. The test compound may be defined by structure or function or it may be undefined. Examples of undefined test compounds include without limitation tissue samples, biological fluids, cell supernatants, vegetal preparations; etc. Test compounds may be peptides such as soluble peptides including Ig-tailed fusion peptides, members of random peptide libraries and combinatorial chemistry-derived molecular libraries made of D- and/or L-configuration amino acids, carbohydrates, nucleic acids, antisense molecules, phosphopeptides (including members of random or partially degenerate, directed phosphopeptide libraries), antibodies [e.g. polyclonal, monoclonal, humanized, anti-idiotypic, chimeric, single chain antibodies, fragments, (e.g. Fab, F(ab)2, and Fab expression library fragments, and epitope-binding fragments thereof)], small organic or inorganic molecules, or libraries of compounds. A test compound may be an endogenous physiological compound or natural or synthetic compounds.
[000117] In embodiments, the methods of the invention for identifying agents, in particular anti-cancer agents, comprise contacting more than one test compound, in parallel. In some embodiments, the methods comprises contacting 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50, 100, 1000, at least 2, at least 5, at least 10, at least 50, at least 100, or at least 1000 test compounds in parallel. In some embodiments, high throughput screening of compounds and complete combinatorial libraries are assayed. Methods for performing high throughput screens are well known in the art. The methods can also be automated such that a robot can perform the experiments.
[000118] In embodiments, the methods of the present invention for identifying agents, in particular anti-cancer agents, comprises the step of contacting a cell in the presence of a test compound. The cells can then be observed to determine if the test compound(s) effects USPl l activity, DCAFI O, PALB2, ubiquitination of PALB2, KEAPl , CRL-KEAPl activity, the interaction of BRCAl and PALB2, the interaction of USP1 1 and PALB2, the interaction of USP1 1 and DCAFI O, and/or the interaction of BRCAl , PALB2 and BRCA2. Positive and negative controls may be performed in which known amounts of test compound and no compound, respectively, are added to the assay. One skilled in the art can select and perform the appropriate controls.
[000119] The activity of a test compound(s) may be unknown, and the methods of the invention may be used to identify compounds exhibiting the selected property (e.g., PARP inhibitor sensitizer). In some embodiments, the activity or type of activity of the test compound(s) is known or expected, and the methods of the invention can be used to further characterize or optimize the activity (e.g., specificity, efficacy, etc).
[000120] A method of the invention may also comprise assaying PARP activity in the presence of the test compound. PARP activity may be assayed by measuring a change of poly(ADP-ribose) polymers (PAR), and measuring NAD levels and/or ATP levels using methods routine to one of ordinary skill in the art. In some embodiments, the levels of NAD are depleted in the presence of the test compound. In some embodiments the levels of ATP are depleted in the presence of the test compound. In some embodiments, the levels of NAD are increased in the presence of the test compound. In some embodiments the levels of ATP are increased in the presence of the test compound.
[000121] A method of the invention may comprise the step of determining if a cell has undergone necrosis following administration of a test compound. The physical characteristics of the cell can be analyzed using routine methods known to those skilled in the art to determine if a cell has undergone necrosis. For example, necrosis may be determined by measuring organelle swelling, plasma membrane disintegration, intracellular vacuole formation, and nuclear degradation without condensation.
[000122] The screening methods of the invention may further comprise conducting therapeutic profiling of the identified agents or further analogs thereof, for efficacy and toxicity in animals; optionally formulating a pharmaceutical composition including one or more agents identified as having an acceptable therapeutic profile; and optionally administering the agent to a subject or individual.
[000123] In an aspect, the therapeutic activity of agents and compositions identified using a method of the invention may be evaluated in vivo using a suitable animal model. Thus, the screening methods of the invention may further comprise conducting in vivo studies comprising administering the agent to a suitable animal model.
[000124] The invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising assaying one or more of USP1 1 , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3-KEAP1 , BRCAl -PALB2-BRCA2 complex, PALB2-USP11 complex and C RL3 -KEAP l - PALB2 complex or PALB2 ubiquitylation in a sample from the subject using a method of the invention. Significantly different levels of one or more of USP1 1 , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3-KEAP1 , BRCAl -PALB2-BRCA2 complex, PALB2-USP1 1 complex and CRL3 -KEAP 1 -PALB2 complex or PALB2 ubiquitylation compared to a control indicate responsiveness (e.g., sensitivity) to the PARP inhibitor.
[000125] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USP1 1 , BRCAl , BRCA2, PALB2 and KEAPl in a sample from the subject using a method of the invention. In an embodiment, significantly different levels of USPl l , BRCAl , BRCA2, PALB2 and KEAPl compared to a control indicate responsiveness (e.g., sensitivity) to the PARP inhibitor.
[000126] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USPl l , DCAF10, BRCAl, BRCA2, PALB, KEAPl and CRL3 in a sample from the subject using a method of the invention. In an embodiment, significantly different levels of USP1 1 , DCAF10, BRCAl , BRCA2, PALB, KEAPl and CRL3 compared to a control indicate responsiveness (e.g., sensitivity) to the PARP inhibitor.
[000127] In an aspect, a subject is categorized as responsive to a PARP inhibitor if there is a decrease in one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl, CRL3, CRL3 -KEAPl, BRCAl -PALB 2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control. In an aspect, a subject is categorized as responsive to a PARP inhibitor if there is an increase in one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3 -KEAPl , BRCA1- PALB2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control. In an embodiment, significantly different levels (e.g., lower levels) of USPl l activity compared to a control indicate sensitivity to the PARP inhibitor.
[000128] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting one or more of USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl , CRL3 and CRL3- KEAPl activity or expression or PALB2 ubiquitylation in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
[000129] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USP1 1 , BRCAl , BRCA2, PALB2 and KEAPl activity or expression in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
[000130] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in a subject comprising detecting USPl l , DCAF10, BRCAl , BRCA2, PALB2, KEAPl and CRL3 activity in a sample from the subject and comparing to a control to determine if the subject will be responsive (e.g., sensitive) to the PARP inhibitor.
[000131] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising assaying USP1 1 activity or expression in a sample from the individual using a method of the invention. In an embodiment, significantly different levels (e.g., lower levels) of USP1 1 activity or expression compared to a control indicate sensitivity to the PARP inhibitor.
[000132] In an aspect, the invention provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting KEAP1 in a sample from the individual and comparing to a control to determine if the individual will be sensitive to a PARP inhibitor. In an embodiment, significantly different levels (e.g., higher levels) of KEAP1 compared to a control indicate sensitivity to the PARP inhibitor.
[000133] The invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting CRL3-KEAP1 activity in a sample from the individual and comparing to a control to determine if the individual will be sensitive to a PARP inhibitor. In an embodiment, significantly different levels (e.g., higher levels) of CRL3-KEAP1 compared to a control indicate sensitivity to the PARP inhibitor.
[000134] The invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting PALB2 ubiquitylation in a sample from the individual and comparing to a control to determine if the individual will be sensitive to a PARP inhibitor. In an embodiment, significantly different levels of PALB2 ubiquitylation compared to a control indicate sensitivity to the PARP inhibitor.
[000135] The invention also provides a method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising detecting complexes of BRCA1 , PALB2 and BRCA2 in a sample from the subject and comparing to a control to determine if the individual will be responsive (e.g., sensitive) to a PARP inhibitor. In an embodiment, significantly different levels (e.g., absence or low levels) of complexes of BRCA1 , PALB2 and BRCA2 indicate sensitivity to the PARP inhibitor.
[000136] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USPl l , DCAFIO, PALB2, PALB2 ubiquitylation, BRCAl -PALB2-BRCA2 complex, PALB2-USP1 1 complex, USP1 1 -DCAF10 complex, KEAPl , CRL3-KEAP1 or CRL3-KEAP1 -PALB2 complex in a sample from the individual using a method of the invention.
[000137] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying USPl l activity in a sample from the individual using a method of the invention.
[000138] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising assaying CRL3-KEAP activity in a sample from the individual.
[000139] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating USPl l , BRCAl , BRCA2, PALB2 and KEAPl in a sample from the individual.
[000140] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating USPl l , DCAFIO, BRCAl , BRCA2, PALB2 and KEAPl in a sample from the individual.
[000141] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating BRCAl -PALB2-BRCA2 complex in a sample from the individual.
[000142] The invention further provides a method for assigning an individual to one of a plurality of categories in a clinical trial for a PARP inhibitor comprising detecting or quantitating PALB2 ubiquitylation in a sample from the individual.
[000143] In an aspect, an individual is assigned to a category for a clinical trial for a PARP inhibitor based on a decrease in one or more of USPl l , DCAFIO, BRCAl , BRCA2, PALB2, KEAPl , CRL3, CRL3 -KEAPl , BRCAl -PALB2 and BRCAl - PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control. In an aspect, an individual is assigned to a category for a clinical trial for a PARP inhibitor based on an increase in one or more of USPl l , DCAFIO, BRCAl , BRCA2, PALB2, KEAPl, CRL3, CRL3-KEAP1 , BRCA1-PALB2 and BRCAl -PALB2-BRCA2 activity or expression or PALB2 ubiquitylation compared to a control. [000144] A variety of routine methods known to a person skilled in the art can be employed for detecting or assaying the biomarkers USP1 1 , DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 , CRL3 and/or complexes thereof in a sample. Biomarker levels present in a sample may be determined by any suitable assay, which may comprise the use of any of the group comprising or consisting of immunoassays, spectrometry, mass spectrometry, Matrix Assisted Laser Desorption/Ionization Time-of-Flight (MALDI- TOF) Mass Spectrometry, microscopy, northern blot, isoelectric focussing, SDS- PAGE, PCR, quantitative RT-PCR, gel electrophoresis, DNA microarray, and antibody microarray, or combinations thereof.
[000145] The invention also provides a system comprising: an assay for determining the level of USP11 activity, complexes or biomarker levels in a sample obtained from the subject; a processor for processing the results; computer coded instructions for comparing the results with a database; and a user display for providing the results of the comparison. The database may comprise reference values for USP1 1 activity or biomarker levels.
Treatment Methods
[000146] A method of the invention for predicting or characterizing responsiveness to a PARP inhibitor may further comprise administering the PARP inhibitor to the individual or subject. In an aspect, the invention relates to a method of treating a subject with a PARP inhibitor comprising:
a) assaying a sample from the subject for responsiveness or sensitivity to one or more PARP inhibitors using a method of the invention;
b) identifying a PARP inhibitor that the subject effectively responds or is sensitive to; and
c) administering the PARP inhibitor to the subject.
[000147] In an aspect, the invention provides a method for treating a patient in need of treatment with a PARP inhibitor comprising (a) requesting a test providing the results of an analysis to determine if the patient is sensitive to the PARP inhibitor by detecting USP1 1 , DCAF10, BRCA1 , BRCA2, PALB2, KEAP1 and/or CRL3 in a sample from the subject and comparing to a control to determine if the patient is sensitive to the PARP inhibitor; and (b) administering the PARP inhibitor to the patient if the patient is sensitive to the PARP inhibitor. In an aspect of this method of the invention, the patient has breast cancer. In an aspect of this method of the invention, the patient has ovarian cancer. In an aspect of the invention, the test detects USP11 expression or activity using a method disclosed herein.
[000148] The invention further provides a method for treating cancer in a subject the method comprising: (i) selecting a subject who is responsive to a PARP inhibitor using a method of the invention, and (ii) administering to said subject the PARP inhibitor in an effective amount to treat the cancer. In an embodiment, the cancer is breast cancer. In an embodiment, the cancer is ovarian cancer.
[000149] Agents identified using the methods of the present invention have numerous therapeutic applications related to, for example, cancer, ischemia reperfusion injury, inflammatory diseases, degenerative diseases, protection from adverse effects of cytotoxic compounds, and potentiation of cytotoxic cancer therapy. Agents identified using the methods of the invention may be used to potentiate radiation and chemotherapy by increasing apoptosis of cancer cells, limiting tumor growth, decreasing metastasis, and prolonging the survival of tumor-bearing subjects. In aspects of the invention, the agents can be used to treat leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer and cervical carcinomas.
[000150] In other aspects of the invention, the agents can be used to treat, without limitation, retroviral infection, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, uveitis, diabetes, Parkinson's disease, myocardial infarction, stroke, other neural trauma, organ transplantation, reperfusion of the eye, reperfusion of the kidney, reperfusion of the gut, reperfusion of skeletal muscle, liver toxicity following acetaminophen overdose, cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents, and skin damage secondary to sulfur mustards.
[000151] In some embodiments, the invention provides a method for sensitizing an individual to treatment with PARP inhibitors comprising identifying an agent that sensitizes cells to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
[000152] In some embodiments, the invention provides a method for treating an individual being treated with a PARP inhibitor comprising administering to the individual an agent that sensitizes cells to the PARP inhibitor identified using a method of the invention. [000153] In some embodiments, the invention provides a method for reversing or delaying emergence of resistance to PARP inhibitors in an individual comprising identifying an agent that reverses or delays emergence of resistance to PARP inhibitors in accordance with a method of the invention and administering the agent to the individual.
[000154] In some embodiments, the present invention provides methods of treating cancer in an individual comprising identifying an anti-cancer agent in accordance with a method of the invention and administering the agent to the individual.
[000155] In another embodiment, the present invention provides a method of treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer or cervical carcinomas in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of an agent identified using a method of the invention or a therapeutically acceptable salt thereof.
[000156] In another embodiment, the present invention provides a method of potentiation of cytotoxic cancer therapy in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of an agent identified using a method of the invention that potentiates cytotoxic cancer therapy or a therapeutically acceptable salt thereof.
[000157] In an aspect, the invention provides methods of treating a disease associated with defects in HR (i.e., HR Disease) in an individual comprising identifying an agent that modulates HR in accordance with a method of the invention and administering the agent to the individual.
[000158] In another embodiment, the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for treating a HR Disease in a mammal in need of such treatment. In another embodiment, the present invention provides a use of an agent identified using a method of the invention, to prepare a medicament for inhibiting tumor growth in a mammal in need of such treatment. In another embodiment, the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for treating cancer in a mammal in need of such treatment. In another embodiment, the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, ovarian cancer or cervical carcinomas in a mammal in need of such treatment. In another embodiment, the present invention provides a use of an agent identified using a method of the invention to prepare a medicament for potentiation of cytotoxic cancer therapy in a mammal in need of such treatment comprising administering to the mammal a therapeutically acceptable amount of the agent.
[000159] In an embodiment, the present invention provides a pharmaceutical composition comprising an agent identified using a method of the invention, or a therapeutically acceptable salt thereof, in combination with a therapeutically acceptable carrier. Pharmaceutical compositions can be prepared by per se known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle. Suitable carriers are described, for example, in Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., USA 1985). On this basis, the compositions include, albeit not exclusively, solutions of the active agents in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
[000160] The pharmaceutical compositions are indicated as therapeutic agents either alone or in conjunction with other therapeutic agents or other forms of treatment. The compositions of the invention may be administered concurrently, separately, or sequentially with other therapeutic agents or therapies, for example PARP inhibitors. Homologous Recombination Methods
[000161] The invention provides a method for activating or modulating homologous recombination in a cell comprising:
(a) promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA 1 -PALB2-BRCA2 complexes in the cell;
(b) promoting or stimulating BRCA1 recruitment to DNA double-strand break (DSB) sites;
(c) contacting the cell with BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes;
(d) inhibiting KEAP1 or CRL3-KEAP1 ;
(e) inhibiting the degradation of USP11 or promoting USP1 1 activity; and/or (f) inhibiting DC AF 10.
[000162] In an aspect the cell is in the Gl phase of the cell cycle (Gl). In an aspect the cell is a non-dividing cell or a donnant cell in Gl . In an aspect, the cell is in the GO phase of the cell cycle (GO). In an aspect, the methods of the invention are used in vitro to activate or modulate homologous recombination in cells.
[000163] In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in the Gl phase of the cell cycle (Gl ) or GO phase of the cell cycle (GO), comprising (a) promoting or stimulating the assembly or occurrence of BRCA1-PALB2 or BRCA1-PALB2-BRCA2 complexes in the cell; and/or (b) contacting the cell with BRCA1 -PALB2 or BRCA1 - PALB2-BRCA2 complexes.
[000164] In embodiments, the assembly of BRCA1-PALB2 or BRCA1 -PALB2- BRCA2 complexes is promoted or stimulated by administering an agent that promotes or stimulates BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes identified using a method of the invention.
[000165] In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting KEAP1 or CRL3-KEAP1. In an aspect, the invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of blocking the degradation of USP1 1 or promoting USP1 1 activity. In an embodiment, the method comprises administering USP1 1 or an agonist thereof. In an aspect, the invention provides a method for activating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting KEAP1 and blocking the degradation of USP1 1. In an aspect, the invention provides a method for activating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting CRL3-KEAP1 and blocking the degradation of USP1 1. In an aspect, the invention provides a method for activating homologous recombination in a cell, in particular a cell in Gl or GO, comprising the step of inhibiting CRL3 and blocking the degradation of USP1 1.
[000166] Methods of the invention may be performed in a cell, in particular a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA.
[000167] The invention provides a method for activating or modulating homologous recombination in a cell, in particular a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell. In an embodiment, the assembly of the complexes is promoted or stimulated by administering an agent that promotes or stimulates assembly or occurrence of BRCA1- PALB2 or BRCA1-PALB2-BRCA2 complexes identified using a method of the invention.
[000168] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO), comprising promoting or stimulating the assembly or occurrence of BRCA1- PALB2 or BRC A 1 -PALB2-BRC A2 complexes in the cell.
[000169] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell. In an embodiment, the assembly of the complexes is promoted or stimulated by administering an agent that promotes or stimulates the assembly or occurrence of BRCA1-PALB2 or BRCA1-PALB2-BRCA2 complexes identified using a method of the invention.
[000170] The invention also provides a method for repairing DNA double-strand breaks in a cell in the Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO) in which DNA end resection is or has been activated generating single-stranded DNA, comprising contacting the cell with BRCA1 -PALB2 or BRC A 1 -P ALB2-BRC A2 complexes.
[000171] The invention also provides a method for repairing DNA double-strand breaks in a cell in Gl or GO in which DNA end resection is or has been activated generating single-stranded DNA, the method comprising (a) inhibiting KEAP1 and/or CRL3-KEAP1 ; (b) blocking the degradation of USPl l or promoting or stimulating USPl l activity; (c) administering USPl l or an agonist thereof; and/or (d) inhibiting CRL3-KEAP1 and blocking the degradation of USP1 1.
[000172] A method for activating homologous recombination in a cell may further comprise activating or promoting single strand DNA (ssDNA) generation pathways. In an aspect, ssDNA generation pathways are activated by DNA end resection. In an embodiment, a method for activating homologous recombination in a cell further comprises activating DNA end resection.
[000173] In an embodiment, DNA end resection is activated or promoted by inhibiting 53BP1 (or RIF1) expression or activity (e.g., recruitment of 53BP1 to DSB sites) and/or upregulating or expressing CtlP. In an embodiment, DNA end resection is activated or promoted by inhibiting 53BP1 (or RIF1 ) expression or activity (e.g., recruitment of 53BP1 to DSB sites) and upregulating or expressing CtlP. In an embodiment, the method involves inhibiting 53BP1 using antagonists, including without limitation short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs) or an inhibitor of the histone deacetylase (HDAC) family of enzymes (for example, trichostatin A), and using an analog of CtlP that mimics constitutive phosphorylation, for example CtIP-Thr879Glu.
[000174] A method for activating homologous recombination in a cell may comprise activating BRCA1 recruitment to DNA double-strand break (DSB) sites. In an embodiment, BRCA1 recruitment is activated by inhibiting expression of 53BP1 (TP53BP1 ) or RIF1 , or inhibiting the recruitment of 53BP1 or RIF1 to DSB sites. 53BP1 or RIF1 may be inhibited using antagonists, including without limitation short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs). In an embodiment, 53BP1 is inhibited with an inhibitor of the histone deacetylase (HDAC) family of enzymes, in particular a histone deacetylase inhibitor (HDACi), preferably trichostatin (Fukuda T. et al, Cancer Sci. 2015 Aug; 106(8): 1050-6. doi: 10.111 1/cas. l2717. Epub 2015 Jul 14).
[000175] In an aspect, a method for activating or stimulating homologous recombination in a cell further comprises a gene editing system. In an aspect the gene editing system comprises contacting the cell with a nuclease. Examples of nucleases include without limitation, zinc finger nucleases (ZFNs), engineered meganucleases, transcription activator like effector nucleases (TALENS), mega or homing endonucleases, clustered regularly interspaced short palindromic repeats (CRISPR)- associated (Cas) protein, Ttago nucleases, and fusions between nucleases, such as mega-TALs and compact TALENs.
[000176] In an aspect, the gene editing steps comprise a TALEN system.
[000177] In an aspect, the gene editing steps comprise a ZFN system.
[000178] In an aspect, the gene editing steps comprise a CRISPR/Cas9 system. [000179] In aspects of the invention the gene editing system may correct a genomic modification. A genetic modification may comprise at least one mutation in a polynucleotide sequence having a locus associated with a genetic disorder. In an aspect, the genomic modification is selected from the group consisting of insertions, deletions and combinations thereof. In some embodiments, the genetic disorder is a monogenetic disorder. In some embodiments, the disorder is a multigenetic disorder. In some embodiments, the disorder is associated with one or more SNPs. In particular embodiments of the invention, the genomic modification corrects a point mutation.
[000180] In an aspect of a method of the invention to correct a genomic modification, the gene editing system comprises contacting the cell with a clustered regularly interspaced short palindromic repeats (CRISPR)-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of a target polynucleotide sequence associated with a genetic disorder, wherein the target polynucleotide sequence is cleaved.
[000181] In an aspect, the invention provides a method for altering a genetic disorder associated with a target polynucleotide sequence in a cell comprising: (1) contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 or agents that maintain the BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions throughout the cell cycle; and (2) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved. The method may reduce expression of the target polynucleotide sequence, knock out the target polynucleotide sequence, or correct the target polynucleotide sequence from an undesired sequence to a desired sequence.
[000182] In an aspect, the invention provides a method for altering a genetic disorder associated with a target polynucleotide sequence in a cell comprising: (1) contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises a kit, vector(s) or composition of the invention, in particular the system comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation, preferably the system comprises a KEAP1 inhibitor, an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP- Thr879Glu; and (2) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved. The method may reduce expression of the target polynucleotide sequence, knock out the target polynucleotide sequence, or correct the target polynucleotide sequence from an undesired sequence to a desired sequence.
[000183] The invention contemplates a method for treating or preventing a genetic disorder in a subject, the method comprising altering a target polynucleotide sequence associated with the genetic disorder in a cell by contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises BRCA1 - PALB2 or BRCA1-PALB2-BRCA2 or agents that maintain the BRCA1-PALB2 or BRCA1-PALB2-BRCA2 interactions throughout the cell cycle; and contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved, thereby treating or preventing the genetic disorder.
[000184] In an aspect, a method is provided for treating or preventing a genetic disorder in a subject, the method comprising (a) altering a target polynucleotide sequence associated with the genetic disorder in a cell by contacting the cell with a system which activates homologous recombination in the cell wherein the system comprises a kit, vector(s) or composition of the invention, in particular the system comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation, preferably the system comprises a KEAP1 inhibitor, an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu; and (b) contacting the target polynucleotide sequence with a clustered regularly interspaced short palindromic repeats-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved, thereby treating or preventing the genetic disorder.
[000185] A method for treating or preventing a genetic disorder may comprise introducing the cell into the subject, thereby treating or preventing the genetic disorder associated with the target polynucleotide sequence. The method may comprise repairing the cleaved target polynucleotide sequence by inserting an exogenous template polynucleotide, wherein said repair results in a mutation comprising an insertion, deletion, or substitution of one or more nucleotides of said target polynucleotide sequence.
[000186] In an aspect, the target polynucleotide sequence is associated with a genetic disorder of the lung. In an embodiment the target polynucleotide sequence is associated with cystic fibrosis, in particular the polynucleotide sequence is the cystic fibrosis transmembrane conductor receptor (CFTR) locus. Mutations in the CFTR (e.g., deletion of phenylalanine at position 508 in exon 1 1 ) cause cystic fibrosis.
[000187) In an aspect, the target polynucleotide sequence is associated with a genetic disorder of muscle. In an aspect, the target polynucleotide sequence is associated with muscular dystrophies. In an aspect, the target polynucleotide sequence is associated with Duchenne muscular dystrophy (DMD) (mutations in the dystrophin gene). In an aspect, the target polynucleotide sequence is associated with Becker muscular dystrophy (mutations in the dystrophin gene). In an aspect the target polynucleotide is associated with myotonic dystrophy type 1 (mutations in the DMPK gene) or myotonic dystrophy type 2 (mutations in the CNBP gene). In an aspect, the target polynucleotide sequence is associated with sickle cell anemia (mutated HBB hemoglobin).
[000188] In aspects of the invention, the targeted polynucleotide sequence is associated with a genetic disorder of the liver. In an aspect, the target polynucleotide sequence is associated with alpha- 1 antitrypsin deficiency (mutations in the SERPINA1 gene). In an aspect, the targeted polynucleotide sequence is associated with Wilson disease (mutations in the gene encoding the ATP7B Cu translocase).
[000189] In an aspect, the methods of the invention further comprise providing a functional protein with enhanced characteristics as compared to its naturally occurring counterpart, in particular a functional protein lacking or deficient in a subject, for example for treating genetic disorders. In embodiments of the invention, the methods comprise integrating a sequence encoding a functional protein in a cell in a subject in need thereof by sequential administration of a gene editing system and one or more transgene encoding a non-naturally occurring protein with enhanced properties as compared to its naturally occurring counterpart. In other embodiments, the methods comprise administering to the subject a genetically modified cell expressing a functional version of one or more proteins aberrantly expressed in a subject. Thus, an isolated cell may be introduced into the subject (ex vivo cell therapy) or a cell may be modified when it is part of the subject (in vivo). In certain embodiments, transgene(s) are delivered using a viral vector, a non-viral vector and/or combinations thereof.
[000190] The invention also provides a method for suppressing homologous recombination in a cell, in particular a cell in Gl , comprising suppressing the assembly of BRCA1-PALB2-BRCA2 complexes in the cell. In an embodiment, the interaction is suppressed by administering KEAP1 or CRL3-KEAP1 or an agonist thereof. In an embodiment, the interaction is suppressed by administering a USP1 1 antagonist/inhibitor (e.g., mitoxantrone). In an embodiment, the interaction is suppressed by administering an agent that suppresses homologous recombination identified using a method of the invention.
[000191] Components of the methods of the invention may be delivered by delivery systems known in the art, including without limitation viral based systems or non-viral based systems. Conventional viral based systems may comprise, for example, retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. In an aspect, the expression vector is selected from the group consisting of a plasmid vector, a lentiviral vector, an adenoviral vector, and an adeno-associated virus vector. In an embodiment, the viral based system, is an adenoviral vector or adeno-associated viral vector. Examples, of non-viral based systems include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virons and agent-enhanced uptake of DNA.
[000192] In an aspect, the invention provides vectors comprising activators or modulators of homologous recombination, and optionally activators or modulators of DNA end resection. In an aspect, the invention provides a vector (e.g. viral vector) comprising one or more of the following components encoded in the vector: 1) an activator of DNA end resection, for example, an inhibitor of 53BP1 (or RTF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) a factor that activates homologous recombination, for example, a factor that maintains BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, a TALEN system or a zinc finger nuclease system. In an embodiment, the components of the gene editing system are encoded in one or more separate expression vectors.
[000193] In another aspect, the invention provides a composition comprising activators or modulators of homologous recombination, and optionally activators or modulators of DNA end resection. In an aspect, the invention provides a composition comprising one or more of the following components: 1) an activator of DNA end resection, for example, an inhibitor of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) a factor that activates homologous recombination, for example, a factor that maintains BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system. In an embodiment, the components of the gene editing system are in one or more separate compositions.
[000194] Examples of activators of DNA end-resection include without limitation, the coding sequence of CtIP-Thr847Glu, a shRNA against the TP53BP1 mRNA, and a shRNA against KEAP1. The shRNA against TP53BP1 may be substituted with a shRNA against RIF1 or agents that block 53BP1 recruitment to DSB sites including a dominant-negative 53BP1 protein. The shRNA against KEAP1 may be substituted with a the coding sequence of a PALB2 mutant that contains mutations of its Lys20, Lys25 and Lys30 residues or that contains a mutation that disrupts its interaction with KEAP1.
[000195] Examples of factors that maintain BRCA1 -PALB2 or BRCA1-PALB2- BRCA2 interactions during the cell cycle include without limitation, inhibitors of KEAP1 , inhibitors of DCAF10, RNA interference agents that maintain USP1 1 expression in GO and Gl cells or a mutated form of PALB2 that is insensitive to ubiquitylation by KEAP1 -CUL3-RBX1 which involves the mutation of one or more of the Lys20, Lys25 or Lys30 residues. An example of a KEAP1 inhibitor is the monobody that is a potent competitive inhibitor of the KEAP1-NRF2 interaction disclosed in Guntas, G. et al, [44]. KEAP1 inhibitors are also described, for example in Canning P. et al, Acta Pharm Sin B., 2015 (4):285-99 and Wells, G., Biochem Soc Trans. 2015,43(4): 674-9.
[000196] In an embodiment, a vector of the invention comprises sequences encoding an inhibitor of 53BP1, a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation. In a particular embodiment, a vector of the invention comprises sequences encoding a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu. In a particular embodiment, a vector of the invention comprises sequences encoding a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
[000197] In an embodiment, a composition of the invention comprises an inhibitor of 53BP1 , a KEAP1 inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation. In a particular embodiment, a composition of the invention comprises a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu. In a particular embodiment, a composition of the invention comprises a KEAP1 inhibitor (e.g., Rl KEAP1 inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
Kits
[000198] The invention further provides a kit for performing an assay or method disclosed herein or comprising compositions or vectors disclosed herein. In an embodiment, a kit of the invention comprises at least one reagent for determining USP1 1 activity in a sample. In another embodiment, a kit of the invention comprises at least one reagent for determining BRCA1 -PALB2-BRCA2, PALB2-KEAP1 , BRCA1- PALB2, USP11-DCAF10, or USP1 1 -PALB2 complexes in a sample. In another embodiment, a kit of the invention comprises at least one reagent for determining BRCA1-PALB2-BRCA2, PALB2-KEAP1 or USP11-PALB2 complexes in a sample. In another embodiment, a kit of the invention comprises reagents for determining the levels of BRCA1 , BRCA2, PALB2, USP1 1 , DCAF10 and KEAP1 in a sample. In another embodiment, a kit of the invention comprises at least one reagent for determining ubiquitylation of PALB2, in particular ubiquitylation of the N-terminus of PALB2, in a sample. In some embodiments the reagent is an antibody or a nucleic acid or primers for use in a PCR reaction.
[000199] A kit may also comprise instructions for suitable operational parameters in the form of an insert. The instructions may inform a consumer about how to collect the sample. The kit may comprise samples, to be used as standard(s) for calibration and comparison. The kit may also comprise instructions to compare the level of activity or biomarkers detected in a sample with a calibration sample or chart. The kit may also include instructions indicating what level of activity or biomarkers is diagnostic of a disease disclosed herein.
[000200] In an aspect, the invention provides a kit comprising one or more of the components of a method of the invention for activating homologous recombination and optionally components of a gene editing system. A kit of the invention may also include or be used in combination with a CRISPR system, a TALEN system or zinc finger nuclease system. In an embodiment, a kit of the invention includes or is used in combination with a CRISPR system. In an embodiment, a kit of the invention includes or is used in combination with a TALEN system. In an embodiment, a kit of the invention includes or is used in combination with a zinc finger nuclease system.
[000201] In some embodiments, a kit of the invention comprises vector systems and instructions for using the kit. In an aspect, the kit comprises a vector comprising activators of DNA end resection and activators of homologous recombination discussed herein. In an aspect, the kit comprises one or more vectors (e.g. viral vectors) comprising one or more of the following components: 1 ) activators of DNA end resection, for example, inhibitors of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) factors that activate homologous recombination, for example, factors that maintain BRCA1-PALB2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system. Examples of factors that maintain BRCA1-PALB2 interactions during the cell cycle are described herein and include without limitation, inhibitors of KEAP1 , for example, RNA interference agents that maintain USP1 1 expression in GO and G l cells or a mutated form of PALB2 that is insensitive to ubiquitylation by KEAP1 - CUL3-RBX1 which involves the mutation of one or more of the Lys20, Lys25 or Lys30 residues. Examples of activators of DNA end resection include without limitation, the coding sequence of CtIP-Thr847Glu, a shRNA against the TP53BP1 inRNA, and a shRNA against KEAP1. The shRNA against TP53BP1 may be substituted with a shRNA against RIF1 or agents that block 53BP1 recruitment to DSB sites including a dominant-negative 53BP1 protein. The shRNA against KEAP1 may be substituted with a coding sequence of a PALB2 mutant that contains mutations of its Lys20, Lys25 and Lys30 residues or that contains a mutation that disrupts its interaction with KEAP1. [000202] In an embodiment, a kit of the invention comprises one or more vectors comprising sequences encoding an inhibitor of 53BP1 , a KEAPl inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation. In a particular embodiment, a kit of the invention comprises one or more vectors comprising sequences encoding a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu. In a particular embodiment, a kit of the invention comprises one or more vectors comprising sequences encoding a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
[000203] In some embodiments, a kit of the invention comprises a composition of the invention and instructions for using the kit. In an aspect the kit comprises a composition comprising activators or modulators of DNA end- resection and activators or modulators of homologous recombination discussed herein. In an aspect, the kit comprises a composition comprising one or more of the following components: 1 ) an activator of DNA end resection, for example, an inhibitor of 53BP1 (or RIF) expression or activity and/or a CtIP compound that mimics constitutive phosphorylation; 2) a factor that activates homologous recombination, for example, a factor that maintains BRCA1-PALB2 or BRCA1-PALB2-BRCA2 interactions during the cell cycle; and, optionally, 3) components of a gene editing system, in particular components of a CRISPR system, TALEN system or zinc finger nuclease system. In an embodiment, the components of the gene editing system are in separate kit(s).
[000204] In an embodiment, a kit of the invention comprises a composition comprising an inhibitor of 53BP1 , a KEAPl inhibitor or DCAF10 inhibitor, and an analog of CtIP that mimics constitutive phosphorylation. In a particular embodiment, a kit of the invention comprises a composition comprising a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 and CtIP-Thr879Glu. In a particular embodiment, a kit of the invention comprises a composition comprising a KEAPl inhibitor (e.g., Rl KEAPl inhibitor; see Example 3), an inhibitor of 53BP1 chosen from short interfering (si) RNA, short hairpin (sh) RNA and microRNAs (miRNAs), and CtIP-Thr879Glu.
[000205] In some aspects, a kit of the invention is used in combination with a gene editing kit, in particular a kit for a CRISPR system, a TALEN system or zinc finger nuclease system. Gene editing kits are commercially available, for example from Addgene (Cambridge, MA), ThermoFisher Scientific, System Biosciences Inc., and OriGene Technologies (MD), Clontech.
[000206] The following non-limiting examples are illustrative of the present invention:
EXAMPLE 1
[000207] The following materials and methods were used in the study described in the Example.
Plasmids
[000208] The cDNA of PALB2 was obtained from the Mammalian Gene Collection (MGC). Full length PALB2 and BRCA1 were amplified by PCR, subcloned into pDONR221 and delivered into the pDEST-GFP, pDEST-Flag and the mCherry- LacR vectors using Gateway cloning technology (Invitrogen). Similarly, the coiled-coil domain of BRCA1 (residues 1363-1437) was amplified by PCR, subcloned into the pDONR221 vector and delivered into both mCherryLacR and pDEST-GFP vectors. The N-terminal domain of PALB2 was amplified by PCR and introduced into the GST expression vector pET30-2-His-GST-TEV [31 ] using the EcoRI/XhoI sites. The coiled- coil domain of BRCA1 was cloned into pMAL-c2 using the BamHI/Sall sites. Truncated forms of PALB2 were obtained by introducing stop codons or deletions through site-directed mutagenesis. Full-length CtIP was amplified by PCR, subcloned into the pDONR221 and delivered into the lentiviral construct pCW57.1 (a gift of Dr. David Root; Addgene plasmid #41393) using Gateway cloning technology (Invitrogen). The USPl 1 cDNA was a gift of David Cortez and was amplified by PCR and cloned into the pDsRed2-C l vector using the EcoRI/Sall sites. The bacterial codon-optimized coding sequence of pig USP1 1 (USP1 1 ) was subcloned into the 6 x His-GST vector pETM-30-Htb using the BamHI/EcoRI sites. siRNA-resistant versions of PALB2, BRCA1 and USPl 1 constructs were generated as previously described [14]. Full-length CUL3 and RBX1 were amplified by PCR from a human pancreas cDNA library (Invitrogen) as previously described [32] and cloned into the dual expression pFBDM vector using Nhel/Xmal and BssHII/NotI respectively. The NEDD8 cDNA was a gift of Dmitris Xirodimas and was fused to a double StrepII tag at its C-terminus in the pET17b vector (Millipore). Human DEN1 was amplified from a vector supplied by Aude Echalier and fused to a non-cleavable N-terminal StrepII2 x tag by PCR and inserted into a pET17b vector. The pCOOL-mKEAPl plasmid was a gift from Dr. Feng Shao. The pcDNA3-HA2-KEAPl and pcDNA3 -H A2-KE AP 1 ΔΒΤΒ were gifts from Dr. Yue Xiong (Addgene plasmids #21556 and 21593). gRNAs were synthesized and processed as described previously [33]. Annealed gRNAs were cloned into the Cas9- expressing vectors pSpCas9(BB)-2A-Puro (PX459) or pX330-U6- Chimeric_BB-CBh- hSpCas9, a gift from Feng Zhang (Addgene plasmids #48139 and 42230). The gRNAs targeting the LMNA or the PML locus and the mClover-tagged LMNA or PML are previously described [45] The lentiviral packaging vector psPAX2 and the envelope vector VSV-G were a gift from Didier Trono (Addgene plasmids #12260 and 12259). His6-Ubiquitin was cloned into the pcDNA5-FRT/TO backbone using the Xhol/Hindlll sites. All mutations were introduced by site-directed mutagenesis using QuikChange (Stratagene) and all plasmids were sequence-verified.
Cell culture and plasmid transfection
[000209] All culture media were supplemented with 10% fetal bovine serum (FBS). U-2-OS (U20S) cells were cultured in McCoy's medium (Gibco). 293T cells were cultured in DMEM (Gibco). Parental cells were tested for mycoplasma contamination and authenticated by STR DNA profiling. Plasmid transfections were carried out using Lipofectamine 2000 Transfection Reagent (Invitrogen) following the manufacturer's protocol. Lentiviral infection was carried out as previously described
[18]. U20S and 293T cells were purchased from ATCC. U20S 256 cells were a gift from R. Greenberg.
Antibodies
[000210] The following antibodies were employed: rabbit anti-53BPl (A300- 273A, Bethyl), rabbit anti-53BPl (sc-22760, Santa Cruz), mouse anti-53BPl (#612523, BD Biosciences), mouse anti-y-H2AX (clone JBW301 , Millipore), rabbit anti-y-H2AX (#2577, Cell Signaling Technologies), rabbit anti-KEAPl (ab66620, Abeam), rabbit anti-NRF2 (ab62352, Abeam), mouse anti-Flag (clone M2, Sigma), mouse anti-tubulin (CP06, Calbiochem), mouse anti-GFP (#1 1814460001 , Roche), mouse anti-CCNA (MONX10262, Monosan), rabbit anti-BRCA2 (ab9143, Abeam), mouse anti-BRCA2 (OP95, Calbiochem), rabbit anti-BRCAl (#07-434, Millipore), rabbit anti-USPl l (abl09232, Abeam), rabbit anti-USPl l (A301 -613A, Bethyl), rabbit anti-RAD51 (#70- 001 , Bioacademia), mouse anti-BrdU (RPN202, GE Healthcare), mouse anti-FK2 (BMLPW8810, Enzo), rabbit anti-PALB2 [34], rabbit anti-GST (sc-459, Santa Cruz), rabbit anti-CUL3 (A301 -108A, Bethyl), mouse anti-MBP (E8032, NEB), mouse anti- HA (clone 12CA5, a gift of Dr. M. Tyers), rabbit anti-Ubiquitin (Z0458, Dako) and mouse anti-actin (CPOl , Calbiochem). The following antibodies were used as secondary antibodies in immunofluorescence microscopy: Alexa Fluor 488 donkey anti-rabbit IgG, Alexa Fluor 488 donkey anti-goat IgG, Alexa Fluor 555 donkey anti- mouse IgG, Alexa Fluor 555 donkey anti-rabbit IgG, Alexa Fluor 647 donkey anti- mouse IgG, Alexa Fluor 647 donkey anti-human IgG, Alexa Fluor 647 donkey anti- goat IgG (Molecular Probes).
RNA interference
[000211] All siRNAs employed in this study were single duplex siRNAs purchased from ThermoFisher. RNA interference (RNAi) transfections were performed using Lipofectamine RNAiMax (Invitrogen) in a forward transfection mode. The individual siRNA duplexes used were: BRCA1 (D-003461 -05), PALB2 (D-012928- 04), USP11 (D-006063-01), CUL1 (M-004086-01), CUL2 (M-007277-00), CUL3 (M- 010224-02), CUL4A (M-012610-01), CUL4B (M-017965-01), CUL5 (M-019553-01), KEAP1 (D-12453-02), RAD 51 (M-003530-04), QIP/RBBP8 (M-001376-00), BRCA2 (D-003462-04), 53BP1 (D-003549-01) and non-targeting control siRNA (D-001210- 02). Except when stated otherwise, siRNAs were transfected 48 h prior to cell processing.
Inhibitors and fine chemicals
[000212] The following drugs were employed at the indicated concentrations: cycloheximide (CHX; Sigma) at 100 ng/mL"1, camptothecin (CPT; Sigma) at 0.2 μΜ, ATM inhibitor (KU55933; Selleck Chemicals) at 10 μΜ, ATR inhibitor (VE-821 ; gift of Philip Reaper) at 10 μΜ, DNA-PKcs inhibitor (NU7441 ; Genetex) at 10 μΜ, proteasome inhibitor MG132 (Sigma) at 2 μΜ, Lovastatin (S2061 ; Selleck Chemicals) at 40 μΜ, Doxycycline (#8634-1 ; Clontech), Nedd8-activating enzyme inhibitor (MLN4929; Active Biochem) at 5 μΜ and olaparib (Selleck) at the indicated concentrations.
Immunofluorescence microscopy
[000213] In most cases, cells were grown on glass coverslips, fixed with 2% (w/v) paraformaldehyde in PBS for 20 min at room temperature, permeabilized with 0.3 % (v/v) Triton X-100 for 20 min at room temperature and blocked with 5% BSA in PBS for 30 min at room temperature. Alternatively, cells were fixed with 100% cold methanol for 10 min at -20°C and subsequently washed with PBS for 5 min at room temperature before PBS-BSA blocking. Cells were then incubated with the primary antibody diluted in PBS-BSA for 2 h at room temperature. Cells were next washed with PBS and then incubated with secondary antibodies diluted in PBS-BSA supplemented with 0.8 μg/ \ of DAPI to stain DNA for 1 h at room temperature. The coverslips were mounted onto glass slides with Prolong Gold mounting agent (In vitro gen). Confocal images were taken using a Zeiss LSM780 laser-scanning microscope. For Gl vs. S/G2 analysis of the BRCA1-PALB2-BRCA2 axis, cells were first synchronized with a double-thymidine block, released to allow entry into S phase and exposed to 2 or 20 Gy of X-irradiation at 5h and 12h post-release and fixed at 1 to 5 hours post-treatment (where indicated). For the examination of DNA replication, cells were pre-incubated with 30 μΜ BrdU for 30 min before irradiation and processed as previously described. CRISPR/Cas9 genome editing of USP11/KEAP1
[000214] 293T and U20S cells were transiently transfected with 3 distinct sgRNAs targeting either 53BP1 , USP1 1 or KEAP1 and expressed from the pX459 vector containing Cas9 followed by the 2A-Puromycin cassette. The next day, cells were selected with puromycin for 2 days and subcloned to form single colonies or subpopulations. Clones were screened by immunoblot and/or immunofluorescence to verify the loss of 53BP1 , USP1 1 or EAP1 expression and subsequently characterized by PCR and sequencing. The genomic region targeted by the CRISPR/Cas9 was amplified by PCR using Turbo Pfu polymerase (Agilent) and the PCR product was cloned into the pCR2.1 TOPO vector (Invitrogen) before sequencing.
Olaparib clonogenic assay
[000215] 293T cells were incubated with the indicated doses of olaparib (Selleck Chemicals) for 24 h, washed once with PBS and counted by trypan blue staining. Five- hundred cells were then plated in duplicate for each condition. The cell survival assay was performed as previously described [35].
Recombinant protein production
[000216] GST and MBP fusions proteins were produced as previously described [36, 37]. Briefly, MBP proteins expressed in Escherichia coli were purified on amylose resin (New England Biolabs) according to the batch method described by the manufacturer and stored in IX PBS, 5% glycerol. GST proteins expressed in E. coli were purified on glutathione sepharose 4B (GE Healthcare) resin in 50 mM Tris HC1 pH 7.5, 300 mM NaCl, 2 mM dithiothreitol (DTT), 1 mM EDTA, 15 μg/mL"1 AEBSF and lx Complete protease inhibitor cocktail (Roche). Upon elution from the resin using 50 mM glutathione in 50 mM Tris HC1 pH 8, 2 mM DTT, the His6-GST tag was cleaved off using His-tagged TEV protease (provided by F. Sicheri) in 50 mM Tris HC1 pH 7.5, 150 mM NaCl, 10 mM glutathione, 10% glycerol, 2 mM sodium citrate and 2 niM β-mercaptoethanol. His6-tagged proteins were depleted using Ni-NTA-agarose beads (Qiagen) in 50 mM Tris HC1 pH 7.5, 300 mM NaCl, 20 mM imidazole, 5 mM glutathione, 10% glycerol, ImM sodium citrate and 2 mM β-mercaptoethanol followed by centrifugal concentration (Amicon centrifugal filters, Millipore). GST-mKEAPl was purified as described previously [38], with an additional anion exchange step on a HiTrap Q HP column (GE Healthcare). The GST tag was left on the protein for in vitro experiments. Purification of CUL3 and RBX1 was performed as previously described [32]. Nedd8 and Denl were expressed in E.coli BL21 grown in Terrific broth media and induced overnight with 0.5 mM isopropyl-p-D-thiogalactoside (IPTG) at 16°C. Cells were harvested and resuspended in wash buffer (400 mM NaCl, 50 mM Tris-HCl, pH 8, 5% glycerol, 2 mM DTT), supplemented with Iysozyme, universal nuclease (Pierce), benzamidine, leupeptin, pepstatin, PMSF and Complete protease inhibitor cocktail (Roche), except for DEN 1 -expressing cells where the protease inhibitors were omitted. Cells were lysed by sonication and the lysate was cleared by centrifugation at 20,000 rpm for 50 min. The soluble supernatant was bound to a 5 ml Strep30 Tactin Superflow Cartridge with a flow rate of 3 ml/min"1 using a peristaltic pump. The column was washed with 20 column volumes (CV) of washing buffer and eluted with 5 CV washing buffer, diluted 1 :2 in water to reduce the final salt concentration, and supplemented with 2.5 mM desthiobiotin. The elution fractions were pooled and concentrated to a total volume of 4 ml using a 3 kDa cut-off Amicon concentrator. DEN1 was further purified over a Superdex 75 size exclusion column, buffer exchanged into 150 mM NaCl, HEPES, pH 7.6, 2% glycerol and 1 mM DTT. The C- terminal pro-peptide and StrepII2x-tag were removed by incubation with StrepII2x- DEN1 in 1 :20 molar ratio for 1 hour at room temperature. The DEN1 cleavage reaction was buffer exchanged on a Zeba MWCO desalting column (Pierce), to remove the desthiobiotin, and passed through a Strep-Tactin Cartridge, which retains the C- terminal pro-peptide and DENl . The GST-tagged Sus scrofa (pig) USP1 1 proteins were expressed in E. coli as described [39]. Cells were lysed by Iysozyme treatment and sonication in 50mM Tris pH 7.5, 300mM NaCl, ImM EDTA, ImM AEBSF, 1 x Protease Inhibitor mix (284 ng/ml leupeptin, 1.37 μg/mΓ1 pepstatin A, 170 μg/mΓ1 PMSF and 330 g mΓ1 benzamidine) and 5% glycerol. Cleared lysate was applied to a column packed with glutathione sepharose 4B (GE Healthcare), washed extensively with lysis buffer before elution in 50mM Tris pH 7.5, 150 mM NaCl, 5% glycerol and 25 mM reduced glutathione. DUB activity was assayed on fluorogenic Ubiquitin-AMC (Enzo life sciences), measured using a Synergy Neo microplate reader (Biotek). His6- TEV-Ubiquitin-G76C was purified on chelating HiTrap resin, following the manufacturers' instructions, followed by size exclusion chromatography on a S-75 column (GE healthcare). The protein was extensively dialysed in ImM acetic acid and lyophilised.
In vitro ubiquitylation and deubiquitylation of PALB2
[000217] HA-tagged N-terminal fragments of PALB2 (1-103) (1 μΜ) were in vitro ubiquitylated using 50μΜ wild-type (Ubi WT, Boston Biochem) or a lysine-less ubiquitin (Ubi K0, Boston Biochem), 100 nM human UBA1 (El), 500 nM CDC34 (provided by F. Sicheri and D. Ceccarelli), 250 nM neddylated CUL3/RBX1 , 375 nM GST-mKEAPl and 1.5 mM ATP in a buffer containing 50 mM Tris HC1 pH 7.5, 20 mM NaCl, 10 mM MgCl2 and 0.5 mM DTT. Ubiquitylation reactions were carried out at 37°C for 1 hour, unless stated otherwise. For USP1 1 -mediated deubiquitylation assays, HA-PALB2 (1 -103) was first ubiquitylated using lysine-less ubiquitin with enzyme concentrations as described above in 50 reactions in a buffer containing 25mM HEPES pH 8, 150 mM NaCl, 10 mM MgCl2, 0.5 mM DTT and 1.5 mM ATP for 1.5 h at 37°C. Reactions were stopped by the addition of 1 unit Apyrase (New England Biolabs). Reaction products were mixed at a 1 : 1 ratio with wild-type or catalytically inactive (C270S) USP11 , or USP2 (provided by Dr. F. Sicheri and E. Zeqiraj) using final concentrations of 100 nM, 500 nM and 2500 nM (USP1 1 ) and 500 nM (USP2) and incubated for 2 h at 30°C in a buffer containing 25 mM HEPES pH 8, 150 mM NaCl, 2 mM DTT, 0.1 mg/mL BSA, 0.03% Brij-35, 5 mM MgCl2, 0.375 mM ATP.
Pulldown experiments between purified PALB2 and BRCA1
[000218] PALB2 in vitro ubiquitylation reaction products were diluted in a buffer at final concentration of 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM MgCl2, 0.25 mM DTT and 0.1% NP-40. 20 μg MBP or MBP-BRCAl -CC was coupled to amylose resin (New England Biolabs) in the above buffer supplemented with 0.1% BSA prior to addition of the ubiquitylation products. Pulldown reactions were performed at 4°C for 2 h, followed by extensive washing.
Co-immunoprecipitation
[000219] Cells were collected by trypsinization, washed once with PBS and lysed in 500 μΐ, of lysis buffer (20 mM Tris-HCl pH 8.0, 150 mM NaCl, 10% glycerol, 2 mM EDTA, 1 % NP-40, Complete protease inhibitor cocktail (Roche), cocktail of phosphatase inhibitors (Sigma) and N-ethylmaleimide to inhibit deubiquitination) on ice. Lysates were centrifuged at 15 000 g for 10 min at 4°C and protein concentration was evaluated using absorbance at 280 nm. Equivalent amounts of proteins (-0.5-1 mg) were incubated with 2 μg of rabbit anti-PALB2, rabbit anti-USPl l antibody, rabbit anti-GFP antibody or normal rabbit IgG for 5 h at 4°C. A mix of protein A/protein G- Sepharose beads (Thermo Scientific) was added for an additional hour. Beads were collected by centrifugation, washed twice with lysis buffer and once with PBS, and eluted by boiling in 2X Laemmli buffer before analysis by SDS-PAGE and immunoblotting. For MS analysis of Flag-PALB2, 150 x 106 transiently transfected HEK293T cells were lysed in high-salt lysis buffer (50 mM Tris-HCl pH 7.5, 300 mM NaCl, 1 mM EDTA, 1 % Triton X100, 3 mM MgCl2, 3 mM CaCl2), supplemented with Complete protease inhibitor cocktail (Roche), 4 mM 1 , 10-Phenantroline, 50 U benzonase and 50 U micrococcal nuclease. Cleared lysates were incubated with Flag- M2 agarose (Sigma), followed by extensive washing in lysis buffer and 50 mM ammoniumbicarbonate.
Mass spectrometry
[000220] After immunoprecipitation of transiently transfected Flag-PALB2 from siCTRL-transfected or USP1 1 siRNA-depleted 293T cells, cysteine residues were reduced and alkylated on beads using 10 mM DTT (30 min. at 56 °C) and 15 mM 2- chloroacetamide (1 h at room temperature), respectively. Proteins were digested using limited trypsin digestion on beads (1 μg trypsin (Worthington, NJ, USA) per sample, 20 min at 37°C), and dried to completeness. For LCMS/MS analysis, peptides were reconstituted in 5% formic acid and loaded onto a 12 cm fused silica column with pulled tip packed in-house with 3.5 μπι Zorbax CI 8 (Agilent Technologies, CA, USA). Samples were analyzed using an Orbitrap Velos (Thermo Scientific, MA, USA) coupled to an Eksigent nanoLC ultra (AB SCIEX, CA). Peptides were eluted from the column using a 90 min linear gradient from 2% to 35% acetonitrile in 0.1 % formic acid. Tandem MS spectra were acquired in a data-dependent mode for the top two most abundant multiply charged peptides and included targeted scans for five specific N- terminal PALB2 tryptic digest peptides (charge state 1+, 2+, 3+), either in non- modified form or including a diGly-ubiquitin trypsin digestion remnant. Tandem MS spectra were acquired using collision-induced dissociation. Spectra were searched against the human Refseq_V53 database using Mascot, allowing up to 4 missed cleavages and including carbamidomethyl (C), deamidation (NQ), oxidation (M), GlyGly (K) and LeuArgGlyGly (K) [SEQ ID NO: 4] as variable modifications.
[000221] In vitro ubiquitylated HA-PALB2 (1 -103) (50 μί total reaction mix) was run briefly onto an SDS-PAGE gel, followed by total lane excision, in-gel reduction using 10 mM DTT (30 min at 56 °C), alkylation using 50 mM 2-chloroacetamide and trypsin digestion for 16 h at 37 °C. Digested peptides were mixed with 20 μΕ of a mix of 10 unique heavy isotope-labeled N-terminal PALB2 (AQUA) peptides (covering full or partial tryptic digests of regions surrounding Lys 16, 25, 30 or 43, either in non- modified or diG-modified form; 80-1 ,200 fmol μ Γ1 per peptide, based on individual peptide sensitivity testing) before loading 6 μΕ onto a 12 cm fused silica column with pulled tip packed in-house with 3.5 μτη Zorbax C18. Samples were measured on an Orbitrap ELITE (Thermo Scientific, MA, USA) coupled to an Eksigent nanoLC ultra (AB SCIEX, CA, USA). Peptides were eluted from the column using a 180 min linear gradient from 2% to 35% acetonitrile in 0.1% formic acid. Tandem MS spectra were acquired in a data-dependent mode for the top two most abundant multiply charged ions and included targeted scans for ten specific N-terminal PALB2 tryptic digest peptides (charge states 1 +, 2+, 3+), either in light or heavy isotope-labeled form. Tandem MS spectra were acquired using collision induced dissociation. Spectra were searched against the human Refseq_V53 database using Mascot, allowing up to 2 missed cleavages and including carbamidomethyl (C), deamidation (NQ), oxidation (M), GlyGly (K) and LeuArgGlyGly (K) [SEQ ID NO:4] as variable modifications, after which spectra were manually validated.
His-Ubiquitin pull-down
[000222] 293 FLIP-IN cells stably expressing His6-Ub were transfected with the indicated siRNA and treated with doxycycline (DOX) for 24 h to induce HiS6-Ub expression. Cells were pre-treated with 10 mM N-ethylmaleimide for 30 min and lysed in denaturating lysis buffer (6 M guanidinium-HCl, 0.1 M Na2HP04/NaH2P04, 10 mM Tris-HCl, 5 mM imidazole, 0.01 M β-mercaptoethanol, complete protease inhibitor cocktail). Lysates were sonicated on ice twice for 10 sec with 1 min break and centrifuged at 15 000 g for 10 min at 4°C. The supernatant was incubated with Ni- NTA-agarose beads (Qiagen) for 4 h at 4°C. Beads were collected by centrifugation, washed once with denaturating lysis buffer, once with wash buffer (8 M Urea, 0.1 M Na2HP04/NaH2P04, 10 mM Tris-HCl, 5 mM imidazole, 0.01 M β-mercaptoethanol, complete protease inhibitor cocktail), and twice with wash buffer supplemented with 0.1% Triton X-100, and eluted in elution buffer (0.2 M imidazole, 0.15 M Tris-HCl, 30% glycerol, 0.72 M β- mercaptoethanol, 5% SDS) before analysis by SDS-PAGE and immunoblotting.
HR-based repair assays
[000223] Parental U20S cells and U20S cells stably expressing wild-type CtIP or CtIP(T847E) mutant were transfected with the indicated siRNA and the PALB2-KR construct, synchronized with a single thymidine block, treated with doxycycline to induce CtIP expression and subsequently blocked in Gl phase by adding 40 μΜ Lovastatin. Cells were collected by trypsinization, washed once with PBS and electroporated with 2.5 μg of sgRNA plasmid and 2.5 μg of donor template using the Nucleofector technology (Lonza; protocol X-001). Cells were plated in medium supplemented with 40 μΜ Lovastatin and grown for 24 h before flow cytometry analysis.
PALB2 chemical ubiquitylation
[000224] PALB2 (1 -103) polypeptides, engineered with only one cross-linkable cysteine, were ubiquitylated by cross-linking alkylation, as previously described [40, 41], with the following modifications. Purified PALB2 cysteine mutant (final concentration of 600 μΜ) was mixed with Hise-TEV-Ubiquitin G76C (350 μΜ) in 300 mM Tris pH 8.8, 120 mM NaCl and 5% glycerol. Tris(2-carboxyethyl)phosphine (TCEP) (Sigma-Aldrich) reducing agent was added to a final concentration of 6mM to the mixture and incubated for 30 minutes at room temperature. The bi-reactive cysteine cross-linker, 1 ,3-dichloroacetone (Sigma-Aldrich), was dissolved in dimethylformamide and added to the protein mix to a final concentration of 5.25 mM. The reaction was allowed to proceed on ice for 1 h, before being quenched by the addition of 5 mM β-mercaptoethanol. His6-TEV-Ubiquitin-conjugated PALB2 was enriched by passing over Ni-NTA-agarose beads (Qiagen).
[000225] The study and the results of the study are discussed below.
[000226] DNA repair by homologous recombination (HR) [1] is highly suppressed in Gl cells [2,3] to ensure that mitotic recombination occurs solely between sister chromatids [4,5]. Although many HR factors are cell cycle-regulated, the identity of the events that are both necessary and sufficient to suppress recombination in Gl cells is unknown. This study has found that the cell cycle tightly controls the interaction of BRCAl with PALB2-BRCA2 to constrain BRCA2 function to the S/G2 phases. The BRCAl -interaction site on PALB2 is targeted by an E3 ubiquitin ligase composed of KEAP1, a PALB2-interacting protein [6], in complex with CUL3-RBX1 [7]. PALB2 ubiquitylation suppresses its interaction with BRCAl and is counteracted by the deubiquitylase USP1 1 , which is itself under cell cycle control. Restoration of the BRCAl -PALB2 interaction combined with the activation of DNA-end resection is sufficient to induce HR in Gl , as measured by RAD51 recruitment, unscheduled DNA synthesis and a CRISPR/Cas9-based gene targeting assay. The mechanism prohibiting HR in Gl minimally consists of the suppression of DNA-end resection coupled with a multi-step block of the recruitment of BRCA2 to DNA damage sites that involves the inhibition of BRCAl -PALB2-BRCA2 complex assembly. The ability to induce HR in Gl cells with defined factors may be used in gene targeting applications in non- dividing cells.
[000227] The breast and ovarian tumour suppressors BRCAl , PALB2 and BRCA2 promote DNA double-strand break (DSB) repair by HR [8-10]. BRCAl promotes DNA-end resection to produce the single-stranded (ss) DNA necessary for homology search and strand invasion [1], and it interacts with PALB2 [13-15] to direct the recruitment of BRCA2 [13] and RAD51 [16, 17] to DSB sites. The accumulation of BRCAl on the chromatin that flanks DSB sites is suppressed in Gl cells [18], reminiscent of the potent inhibition of HR in this phase of the cell cycle. Since the inhibition of BRCAl recruitment in Gl is dependent on the 53BP1 and RIF1 proteins [18, 19], two inhibitors of end-resection [18-22], this regulation of BRCAl was originally viewed in light of its function in stimulating DNA-end processing.
[000228] However, as BRCAl is also involved in promoting the recruitment of BRCA2 through its interaction with PALB2 [13-15], this study investigated whether inducing BRCAl recruitment to DSB sites in Gl, through mutation of 53BP1 (also known as TP53BP1) by genome editing (53BP1A; Figure 5a-c) also resulted in BRCA2 accumulation into ionizing radiation (IR)-induced foci. In contrast with BRCAl , neither BRCA2 nor PALB2 are recruited to Gl DSB sites in U-2-OS (U20S) cells lacking 53BP1 at IR doses ranging from 2 to 20 Gy (Figure lab and Figure 5d,e). Since BRCAl and PALB2 interact directly [13,14], this result suggested that Gl cells may block BRCA2 recruitment by suppressing the BRCAl -PALB2 interaction. Indeed, while PALB2 interacts with BRCA2 irrespective of cell cycle position, it interacts efficiently with BRCAl only during S phase (Figure lc). The presence of DNA damage led to the loss of the residual PALB2-BRCA1 interaction in Gl whereas it had little impact on the assembly of the BRCAl -PALB2-BRCA2 complex in S phase (Figure lc). Since all proteins were expressed in Gl (Figure lc), the results suggest that the assembly of the BRCA1-PALB2-BRCA2 complex is controlled during the cell cycle, possibly to restrict the accumulation of BRCA2 at DSB sites to the S/G2 phases.
[000229] These results were confirmed using a single-cell assay assessing the co- localization, at an integrated LacO array [23], of a mCherry-tagged LacR-BRCAl fusion protein with GFP -tagged PALB2 (Figure 6a). This LacR LacO system recapitulated the cell cycle-dependent and DNA damage-sensitive BRCA1-PALB2 interaction (Figure 6b) and enabled the finding that sequences on PALB2, located outside its amino-terminal BRC A 1 -interaction domain (residues 1-50) were responsible for the cell cycle-dependent regulation of its association with BRCA1 (Figure 6c, d). Further deletion mutagenesis identified a single region, encompassed within residues 46-103 in PALB2 (Figure 6e,f) responsible for the cell cycle-dependent regulation of the BRCA1-PALB2 interaction. This region corresponds to the interaction site for KEAPl [6], identifying this protein as a candidate regulator of the BRCA1-PALB2 interaction.
[000230] KEAPl is a substrate adaptor for a CULLIN 3 -RING ubiquitin (Ub) ligase (CRL3) that targets the antioxidant regulator NRF2 for proteasomal degradation [24] and recognizes an "ETGE" motif on both PALB2 and NRF2 through its KELCH domain [6]. Depletion of KEAPl from 53BP1 cells, or deletion of the ETGE motif in full-length PALB2 (PALB2 AETGE) induced PALB2 IR-induced focus formation in Gl cells (Figure Id and Figure 7a). Furthermore, in cells in which KEAPl was inactivated by genome editing {KEAPl A, Figure 7b) a stable BRC A 1 -PALB2-BRC A2 complex was detected in both Gl and S phases (Figure le). KEAPl is therefore an inhibitor of the BRCA1 -PALB2 interaction.
[000231] CUL3 also interacts with PALB2 (Figure 7c) and its depletion in 53BP1A U20S cells de-repressed PALB2 IR-induced foci in Gl (Figure I d and Figure 7a). Furthermore, in Gl -synchronized KEAP1A cells, expression of a CUL3-binding deficient KEAPl protein that lacks its BTB domain (ΔΒΤΒ) failed to suppress the BRCA1 -PALB2 interaction, unlike its wild type counterpart (Figure 7d). These results suggest that KEAPl recruits CUL3 to PALB2 to suppress its interaction with BRCA1.
[000232] Using the LacR/LacO system and co-immunoprecipitation assays, a mutant of PALB2 lacking all 8 lysine residues in the BRC A 1 -interaction domain (PALB2-KR; Figure 2a) was found to interact with BRCA1 irrespective of cell cycle position (Figure 2b and Figure 7e,f). Further mutagenesis identified residues 20, 25 and 30 in PALB2 as critical for the suppression of the BRCA1-PALB2 interaction since reintroduction of these lysines in the context of PALB2-KR (yielding PALB2-KR/K3; Figure 2a) led to the suppression of BRCA1- PALB2-BRCA2 complex assembly in Gl cells (Figure 2b and Figure 7e). Together, these results suggested a model whereby PALB2-bound KEAP1 forms an active CRL3 complex that ubiquitylates the PALB2 N-terminus to suppress its interaction with BRCA1.
[000233] While PALB2 ubiquitylation can be detected in cells (Figure 8a), the lysine-rich nature of the PALB2 N-terminus has so far precluded unambiguously mapping in vivo ubiquitylation sites on Lys 20, 25 or 30. However, ubiquitylation could be detected on Lys 16 and Lys43 by mass spectrometry, indicating that the PALB2 N- terminus is ubiquitylated (Figure 8b). In a complementary set of experiments, PALB2 targeted to the LacO array induced immunoreactivity to conjugated ubiquitin (Figure 8c-e). Ub co-localization with PALB2 was highest in Gl , and depended on the KEAP1 - interaction motif and the presence of the Lys 20/25/30 residues (Figure 8d-e), consistent with a model that PALB2 is ubiquitylated on those sites in Gl cells. Indeed, ubiquitylation of the N-terminus of PALB2 (residues 1 -103; fused to a haemagglutinin (HA) epitope tag) could be readily reconstituted by recombinant CRL3-KEAP1 , in a manner that depended on the KEAP1 -interaction domain of PALB2 (Figure 2c) and Lys25 and Lys30 were unambiguously identified as being ubiquitylated by KEAP1 in vitro by mass spectrometry.
[000234] Ubiquitylation of PALB2 by CRL3-KEAP1 inhibited its interaction with a BRCA1 fragment comprising residues 1363-1437 (BRCA1-CC), an inhibition that was more obvious with the highly modified forms of PALB2 due to the presence of ubiquitylated lysines outside the BRCA1 -interaction domain (Figure 2d). In order to specifically test whether ubiquitylation of a single lysine residue of the three identified as critical inhibited the interaction with BRCA1, chemical crosslinking was used to install a single ubiquitin moiety at position 20 or 45 (yielding PALB2-Kc20-Ub and PALB2-Kc45-Ub). Ubiquitylation of PALB2 at position 20 completely suppressed its interaction with BRCA1 whereas modification of residue 45 had no impact on the interaction (Figure 9a), echoing the in vivo data (Figure 7e). Together, these results indicate that ubiquitylation of PALB2 at specific sites on its N-terminus prevents its interaction with BRCA1.
[000235] Since neither the activity of the CRL3-KEAP1 E3 ligase (Figure 9b) nor the interaction of CRL3-KEAP1 with PALB2 (Figure 7c) are regulated by the cell cycle, it was possible that eubiquitylation of PALB2 might be regulated in a cell cycle- dependent manner. KEAPl physically interacts with USPl l [25], a deubiquitylase that also interacts with BRCA2 [26] and PALB2 (Figure 9c). USPl l depletion impairs gene conversion [27] (Figure 9d) and results in hypersensitivity to PARP inhibition [27] identifying it as an HR regulator of unknown function. Co-immunoprecipitation experiments confirmed that USP1 1 and its catalytic activity were necessary for the formation of a stable BRCA1 -PALB2-BRCA2 complex, especially in the presence of DNA damage (Figure 3a and Figure 9e,f).
[000236] If USPl l antagonizes PALB2 ubiquitylation by CRL3 -KEAPl , then removal of KEAPl (or CUL3) should reverse the phenotypes imparted by loss of USP1 1. Indeed, deletion of KEAPl restored resistance to PARP inhibitors (PARPi) and the BRCA1 -PALB2 interaction in USPl l knockout cells prepared by genome editing (USP11A) (Figure 3b,c and Figure 9e). Likewise, depletion of CUL3 or KEAPl reversed the gene conversion defect of USP1 1 -depleted cells (Figure 10a). Introduction of the PALB2-KR mutant restored its interaction with BRCA1 and reversed PARPi sensitivity in USPll A cells in a manner that depended on Lys20/25/30 (Figure 10b,c). Since recombinant USPl l can directly de-ubiquitylate PALB2 (1-103) (Figure 3d), these results suggest that USPl l promotes the assembly of the BRCA1 -PALB2- BRCA2 complex by reversing the inhibitory ubiquitylation on the PALB2 Lys20/25/30 residues.
[000237] It was observed that USPl l turns over rapidly in Gl cells and interacts poorly with PALB2 in this phase of the cell cycle (Figure 1 la,b). Furthermore, there is a rapid loss of USPl l upon DNA damage induction, specifically in Gl phase (Figure 3e and Figure 1 lb,c). The destabilization of USP1 1 following IR treatment is dependent on ATM signalling, whereas it is ATR-dependent following UV irradiation (Figure l ld,e). The drop in USPl l steady-state levels in Gl is the result of proteasomal degradation (Figure l l f). A CRL4 E3 Ub ligase is most likely responsible for controlling the stability of USPl l as treatment with MLN4924, a pan-CRL inhibitor [28] (Figure l l g), or depletion of CUL4 (Figure 3f> protected USPl l from DNA damage-induced degradation. CUL4 depletion led to BRCA2 and PALB2 IR-induced focus formation in Gl 53BP1A cells (Figure 3g and Figure 12a), consistent with the regulation of USP1 1 by a CRL4 complex acting as the upstream signal that ultimately controls BRCA1 -PALB2-BRCA2 complex assembly. [000238] While deletion of 53BP1 produces low levels of ssDNA in Gl cells [29], combining the 53BP1 mutation with depletion of KEAP1 did not produce extraction- resistant RAD51 IR induced foci, suggesting little-to-no RAD51 nucleofilament formation (Figure 12b). ssDNA formation remained insufficient in those cells and thus took advantage of the phosphomimetic T847E mutant of CtIP that promotes resection in Gl cells [30]. Unlike wild type CtIP, introduction of CtIP-T847E into 53BP1A cells depleted of KEAP1 induced RAD51 IR-induced focus formation in Gl cells (Figure 4a,b and Figure 12b,c) along with unscheduled DNA synthesis (Figure 12d). These results suggested that the steps downstream of RAD51 nucleofilament formation, i.e. strand invasion, D-loop formation and DNA synthesis, could be activated in Gl .
[000239] To test whether productive HR could also be activated in Gl a CRISPR/Cas9-stimulated gene targeting assay ( Pinder J. et al, Nuclei Acids Res. 43, 9379-9392, 2015) was employed in which the insertion of the coding sequence for the mClover fluorescent protein at the 5' of the lamin A (LMNA) or PML genes was monitored by microscopy or flow cytometry (Figure 4c and Figure 12e,f), with the latter method enabling the gating of cells with a defined DNA content (such as Gl cells). Synchronization protocols were also established in which Gl cells obtained after release from a thymidine block were arrested in Gl by lovastatin treatment [2] for 24 h (Figure 12g,h). Using this system, a concentration of donor template in the linear range of the assay was determined, and it was ascertained that gene targeting at the LMNA locus was dependent on BRCA1 -PALB2-BRCA2 complex assembly (Figure 13a,b). It was also confirmed that gene targeting by HR was highly suppressed in Gl (Figure 4d).
[000240] The combined activation of resection and BRCA1 recruitment to DSB sites (i.e. in 53 BP 1 cells expressing CtIP(T847E) was insufficient to stimulate gene targeting at either the LMNA or the PML locus in Gl cells (Figure 4e and Figure 13c). However, when the BRCA1-PALB2 interaction was restored in resection-competent Gl cells using either KEAP1 depletion or expression of the PALB2-KR mutant, a robust increase in gene targeting events at both loci was detected (Figure 4e and Figure 13c). However, the gene -targeting frequencies of Gl cells remained lower than those of asynchronously dividing cells, suggesting an incomplete activation of HR. 53BP1 inactivation and the expression of CtIP(T847E) were both necessary for G 1 HR (Figure 13d,e), indicating that the simultaneous activation of end resection and BRCA2 recruitment to DSB sites were both necessary and sufficient to activate unscheduled recombination in this phase of the cell cycle. [000241] In conclusion, the regulation of BRCA1 -PALB2-BRCA2 complex assembly is a key node in the cell cycle control of DSB repair by HR. This regulation converges on the BRCA1 -interaction site on PALB2 and is enforced by the opposing activities of the E3 ligase CRL3-KEAP1 and the deubiquitylase USPl l , with the latter being antagonized in Gl by a CRL4 complex (Figure 4f). In this model, the stabilization of USPl l in S phase licenses the recruitment of PALB2-BRCA2 and the subsequent loading of RAD51 at DSB sites. The studies also demonstrate that the suppression of HR in Gl cells is largely reversible and that it involves the combined suppression of end resection and BRCA2 recruitment to DSB sites (Figure 41). As most cells in the human body are not actively cycling and are thus refractory to HR, the manipulations described herein give rise to the development of genome editing methods that enable therapeutic gene targeting in a wider variety of tissues.
EXAMPLE 2
Identification of DCAF10 as a substrate adaptor for the degradation of USPll.
[000242] CUL4-RING-Ligase (CRL4) complexes are composed of CULLIN4 (CUL4), RBXl , DDB1 , DDA1 and a substrate adaptor called a DCAF [42]. To search for the substrate adaptor that mediates the ubiquitylation of USPl l , a focused siRNA library was assembled that depletes known and predicted DCAFs along with other CUL4-interacting proteins. This library was screened in a high content microscopy assay where USPl l levels were evaluated by immunofluorescence microscopy. Cells were treated either with ultraviolet light (UV) or ionizing radiation (IR) to induce USPl l degradation. Data was normalized to the non-irradiated condition and the mean of two independent experiments was used to plot values after UV- and IR-treatment. The data, shown in Figure 14a shows that in addition to the expected stabilization of USP1 1 after CUL4 depletion, the depletion of DCAF10, DCA F15 and DCAF 17 also led to USPl l stabilization. Since siRNA-mediated knockdown is prone to off-target effects, it was then assessed whether the knockdown of DCAF 10, DCAF 15 or DCAF 17 by two independent siRNAs could stabilize USPl l in immunoblotting experiments. It was found that while stabilization of USPl l could only be observed with a single siRNA (Figure 14b), depletion of DCAF 10 with both siRNAs led to a robust stabilization of USPl l (Figure 14b). Since CRL4 substrate adaptors bind to their substrates [42], it was next assessed whether DCAF10 or DCAF15 could interact with USPl l in co-immunoprecipitation assays. It was found that when Flag-tagged USPl l was immunoprecipitated from HEK293 cell extracts, it interacted with DCAF 10 but not DCAF15 (Figure 15 a), strongly suggesting that DCAF10 is a bona fide substrate adaptor that targets USP11 for degradation. To further assess whether DCAF10 is indeed involved in the regulation of USP1 1 , mouse embryo fibroblasts (MEFs) were generated from congenic wild type (Deaf 1 CT/+), heterozygote (Deaf 10"/+) and Deaf 10_/" mice and immunoblotted for USP1 1. Loss of DCAF10 resulted in higher steady-state levels of USP11 in mouse cells (Figure 15b), consistent with DCAF10 being the adaptor of a CRL4 complex targeting USP11. Finally, it was assessed whether DCAF10 over-expression can suppress homologous recombination in a dominant manner using the direct repeat (DR)-GFP assay [43]. Overexpression of DCAF10, but not of DCAF15 led to a decrease in HR in the same magnitude as the depletion of USP1 1 and other core HR factors (Figure 15c). Collectively, these data suggest that DCAF10 regulates HR through the control of USP1 1.
EXAMPLE 3
A genetically-encoded inhibitor of KEAP1 can promote homologous recombination in Gl cells.
[000243] The activation of gene targeting in Gl cells requires the removal of 53BP1 , the introduction of CtIP-T847E and the interaction between PALB2 and BRCA1 , which can be achieved by the removal of KEAP1. To develop a system that would enable activation of HR in G 1 and non-dividing cells, it was determined whether KEAP1 siRNAs could be replaced with inhibitors of KEAP1. A recently described high-affinity genetically encoded inhibitor of KEAP1 , named Rl , which is based on fibronectin-3 (FN3) scaffold was selected [44]. The LMNA gene targeting assay [45] was carried out in 53ΒΡ1Δ U20S cells synchronized in Gl phase and it was found that transfection of the Rl KEAP1 inhibitor, but not its FN3 control, led to a robust activation of gene-targeting, albeit less, and KEAP1 depletion (Figure 16). Inhibition of KEAP1 can be a propitious route for the activation of HR in non-dividing cells.
[000244] The present invention is not to be limited in scope by the specific embodiments described herein, since such embodiments are intended as but single illustrations of one aspect of the invention and any functionally equivalent embodiments are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims. [000245] All publications, patents and patent applications referred to herein are incorporated 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. All publications, patents and patent applications mentioned herein are incorporated herein by reference for the purpose of describing and disclosing the methodologies, reagents, etc. which are reported therein which might be used in connection with the invention. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
Table 1
Protein NCBI Accession SEQ ID
No. NO.
USPl 1 (ubiquitin carboxyl -terminal hydrolase 1 1) NP_004642.2 5
PALB2 (partner and localizer of BRCA2) NP_078951.2 6
BRCA1 (Homo sapiens breast cancer 1) NG_005905.2 7
BRCA2 (Homo sapiens breast cancer 2) NG_012772.3 8
KEAP1 (kelch-like ECH-associated protein 1) NP_036421.2 9
53BP1 (tumor suppressor p53 -binding protein 1 NP_001135452.1 10 isoform 1)
53BP1 (tumor suppressor p53-binding protein 1 NP_001135451.1 1 1 isoform 2)
53BP1 (tumor suppressor p53-binding protein 1 NP_005648.1 12 isoform 3)
DCAF 10 (DDB l - and CUL4-associated factor 10 NP_077321.3 13 isoform a)
DCAF (DDBl - and CUL4-associated factor 10 isoform NP_001273739.1 14 b)
RBX1(E3 ubiquitin-protein ligase RBX1 ) NP_055063.1 15
NG_032169.1 16
CUL3 (Homo sapiens cullin 3)
CtIP/RBBP8 (Homo sapiens retinoblastoma binding NG_012121.1 17 protein 8)
Full citations for publications
1. Jasin, M. & Rothstein, R. Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol 5, a012740, doi: 10.1 101/cshperspect.a012740 (2013).
2. Hartlerode, A., Odate, S., Shim, I., Brown, J. & Scully, R. Cell cycle-dependent induction of homologous recombination by a tightly regulated I-Scel fusion protein.
PLoS One 6, el 6501 , doi: 10.1371/journal.pone.0016501 (201 1).
3. Rothkamm, K., Kruger, I., Thompson, L. H. & Lobrich, M. Pathways of DNA doublestrand break repair during the mammalian cell cycle. Mol Cell Biol 23, 5706- 5715 (2003).
4. Kasparek, T. R. & Humphrey, T. C. DNA double-strand break repair pathways, chromosomal rearrangements and cancer. Semin Cell Dev Biol 22, 886-897, doi: 10.1016/j .semcdb.201 1.10.007 (201 1 ).
5. Panier, S. & Durocher, D. Push back to respond better: regulatory inhibition of the DNA double-strand break response. Nature reviews. Molecular cell biology, doi: 10.1038/nrm3659 (2013).
6. Ma, J. et al. PALB2 interacts with KEAP1 to promote NRF2 nuclear accumulation and function. Mol Cell Biol 32, 1506-1517, doi: 10.1 128/MCB.06271 -1 1 (2012).
7. Genschik, P., Sumara, I. & Lechner, E. The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): cellular functions and disease implications. EMBO J 32, 2307-2320, doi: 10.1038/emboj.2013.173 (2013).
8. Roy, R., Chun, J. & Powell, S. N. BRCAl and BRCA2: different roles in a common pathway of genome protection. Nat Rev Cancer 12, 68-78, doi: 10.1038/nrc3181 nrc3181 [pii] (201 1).
9. Li, M. L. & Greenberg, R. A. Links between genome integrity and BRCAl tumor suppression. Trends Biochem Sci, doi: S0968-0004( 12)00092-8 [pii]
10.1016/j.tibs.2012.06.007 (2012).
10. Park, J. Y., Zhang, F. & Andreassen, P. R. PALB2: The hub of a network of tumor suppressors involved in DNA damage responses. Biochim Biophys Acta 1846, 263-275, doi: 10.1016/j.bbcan.2014.06.003 (2014).
1 1. Schlegel, B. P., Jodelka, F. M. & Nunez, R. BRCAl promotes induction of ssDNA by ionizing radiation. Cancer Res 66, 5181-5189, doi:66/10/5181 [pii] 10.1 158/0008- 5472.CAN-05-3209 (2006). 12. Stark, J. M., Pierce, A. J., Oh, J., Pastink, A. & Jasin, M. Genetic steps of mammalian homologous repair with distinct mutagenic consequences. Mol Cell Biol 24, 9305-9316, doi:24/21/9305 [pii] 10.1 128/MCB.24.21.9305-93 16.2004 (2004).
13. Zhang, F. et al. PALB2 links BRCAl and BRCA2 in the DNA-damage response. Curr Biol 19, 524-529, doi: S0960-9822(09)00723-4 [pii] 10.1016/j.cub.2009.02.018
(2009).
14. Sy, S. M., Huen, M. S. & Chen, J. PALB2 is an integral component of the BRCA complex required for homologous recombination repair. P Natl Acad Sci USA 106, 7155-7160, doi: 10.1073/pnas.081 1 159106 (2009).
15. Simhadri, S. et al. Male Fertility Defect Associated with Disrupted BRCA1 -PALB2 Interaction in Mice. J Biol Chem 289, 24617-24629, doi: 10.1074/jbc.Ml 14.566141 (2014).
16. Bhattacharyya, A., Ear, U. S., Roller, B. H., Weichselbaum, R. R. & Bishop, D. K. The breast cancer susceptibility gene BRCAl is required for subnuclear assembly of Rad51 and survival following treatment with the DNA cross-linking agent cisplatin. J Biol Chem 275, 23899-23903, doi: 10.1074/jbc.C000276200 [pii] (2000).
17. Zhang, F., Bick, G., Park, J. Y. & Andreassen, P. R. MDC1 and RNF8 function in a pathway that directs BRCAl -dependent localization of PALB2 required for homologous recombination. J Cell Sci 125, 6049-6057, doi: 10.1242/jcs.1 1 1872 (2012). 18. Escribano-Diaz, C. et al. A Cell Cycle-Dependent Regulatory Circuit Composed of 53BP1 -RIF1 and BRCAl -CtIP Controls DNA Repair Pathway Choice. Molecular cell 49, 872-883, doi: 10.1016/j.molcel.2013.01.001 (2013).
19. Feng, L., Fong, K. W., Wang, J., Wang, W. & Chen, J. RIF1 counteracts BRCA 1 - mediated end resection during DNA repair. The Journal of biological chemistry 288, 1 1 135-1 1 143, doi: 10.1074/jbc.Ml 13.457440 (2013).
20. Chapman, J. R. et al. RIF1 Is Essential for 53 BP 1 -Dependent Nonhomologous End Joining and Suppression of DNA Double-Strand Break Resection. Mol Cell, doi: S I 097- 2765( 13)00003-8 [pii] 10.1016/j.molcel.2013.01.002 (2013).
21. Bunting, S. F. et al. 53BP 1 inhibits homologous recombination in Brcal -deficient cells by blocking resection of DNA breaks. Cell 141 , 243-254, doi:S0092-
8674(10)00285-0[pii] 10.1016/j.cell.2010.03.012 (2010).
22. Zimmermann, M., Lottersberger, F., Buonomo, S. B., Sfeir, A. & de Lange, T. 53BP1 Regulates DSB Repair Using Rifl to Control 5' End Resection. Science, dokscience.1231573 [pii] 10.1 126/science. l231573 (2013). 23. Tang, J. et al. Acetylation limits 53BP1 association with damaged chromatin to promote homologous recombination. Nature structural & molecular biology, doi: 10.1038/nsmb.2499 (2013).
24. Taguchi, K., Motohashi, H. & Yamamoto, M. Molecular mechanisms of the Keapl- Nrf2 pathway in stress response and cancer evolution. Genes Cells 16, 123-140, doi: 10.1 1 1 l/j. l365-2443.2010.01473.x (201 1).
25. Sowa, M. E., Bennett, E. J., Gygi, S. P. & Harper, J. W. Defining the human deubiquitinating enzyme interaction landscape. Cell 138, 389-403, doi: 10.1016/j.cell.2009.04.042 (2009).
26. Schoenfeld, A. R., Apgar, S., Dolios, G., Wang, R. & Aaronson, S. A. BRCA2 is ubiquitinated in vivo and interacts with USP1 1 , a deubiquitinating enzyme that exhibits prosurvival function in the cellular response to DNA damage. Mol Cell Biol 24, 7444- 7455, doi: 10.1 128/MCB.24.17.7444-7455.2004 24/17/7444 [pii] (2004).
27. Wiltshire, T. D. et al. Sensitivity to poly(ADP-ribose) polymerase (PARP) inhibition identifies ubiquitin- specific peptidase 11 (USP1 1) as a regulator of DNA double-strand break repair. J Biol Chem 285, 14565-14571 , doi:Ml 10.104745 [pii] 10.1074/jbc.M 110.104745 (2010).
28. Enchev, R. I., Schulman, B. A. & Peter, M. Protein neddylation: beyond cullin- RING ligases. Nat Rev Mol Cell Biol 16, 30-44, doi: 10.1038/nrni3919 (2015).
29. Yamane, A. et al. RPA accumulation during class switch recombination represents 5'-3' DNA-end resection during the S-G2/M phase of the cell cycle. Cell Rep 3, 138- 147,doi: 10.1016/j.celrep.2012.12.006 (2013).
30. Huertas, P. & Jackson, S. P. Human CtIP mediates cell cycle control of DNA end resection and double strand break repair. The Journal of biological chemistry 284, 9558-9565, doi: 10.1074/jbc.M808906200 (2009).
31. Fradet-Turcotte, A. et al. 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature 499, 50-54, doi: 10.1038/naturel2318 (2013).
32. Enchev, R. I., Schreiber, A., Beuron, F. & Morris, E. P. Structural insights into the COP9 signalosome and its common architecture with the 26S proteasome lid and eIF3. Structure 18, 518-527, doi: 10.1016/j .str.2010.02.008 (2010).
33. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protocols 8,2281-2308, doi: 10.1038/nprot.2013.143 (2013).
34. Xia, B. et al. Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2. Mol Cell 22, 719-729, doi: 10.1016/j.molcel.2006.05.022 (2006). 35. Orthwein, A. et al. Mitosis inhibits DNA double-strand break repair to guard against telomere fusions. Science 344, 189-193, doi: 10.1 126/science.1248024 (2014).
36. Panier, S. et al. Tandem protein interaction modules organize the ubiquitin- dependent response to DNA double-strand breaks. Molecular cell 47, 383-395, doi: 10.1016/j.molcel.2012.05.045 (2012).
37. Juang, Y. C. et al. OTUB 1 co-opts Lys48-linked ubiquitin recognition to suppress E2 enzyme function. Mol Cell 45, 384-397, doi: 10.1016/j.molcel.2012.01.01 1 S1097-2765(12)00077-9 [pii] (2012).
38. Cui, J. et al. Glutamine deamidation and dysfunction of ubiquitin/NEDD8 induced by abacterial effector family. Science 329, 1215-1218, doi: 10.1 126/science.1 193844 (2010).
39. Hendriks, I. A., Schimmel, J., Eifler, K., Olsen, J. V. & Vertegaal, A. C. Ubiquitin- specific Protease 1 1 (USP11 ) Deubiquitinates Hybrid Small Ubiquitin-like Modifier (SUMO)-Ubiquitin Chains to Counteract RING Finger Protein 4 (RNF4). J Biol Chem 290, 15526-15537, doi: 10.1074/jbc.Ml 14.618132 (2015).
40. Long, L., Furgason, M. & Yao, T. Generation of nonhydrolyzable ubiquitin- histone mimics. Methods 70, 134-138, doi: 10.1016/j.ymeth.2014.07.006 (2014).
41. Yin, L., Krantz, B., Russell, N. S., Deshpande, S. & Wilkinson, K. D. Nonhydrolyzable diubiquitin analogues are inhibitors of ubiquitin conjugation and deconjugation. Biochemistry 39, 10001 -10010 (2000).
42. Jackson, S., and Xiong, Y. (2009). CRL4s: the CUL4-RING E3 ubiquitin ligases. Trends Biochem Sci 34, 562-570.
43. Moynahan, M.E., Chiu, J.W., Koller, B.H., and Jasin, M. (1999). Brcal controls homology-directed DNA repair. Mol Cell 4, 51 1-518.
44. Guntas, G., Lewis, S.M., Mulvaney, K.M., Cloer, E.W., Tripathy, A., Lane, T.R., Major, M.B., and Kuhlman, B. (2016). Engineering a genetically encoded competitive inhibitor of the KEAP1 -NRF2 interaction via structure -based design and phage display. Protein engineering, design & selection : PEDS 29, 1-9.
45. Pinder, J., Salsman, J., and Dellaire, G. (2015). Nuclear domain 'knock-in' screen for the evaluation and identification of small molecule enhancers of CRISPR-based genome editing. Nucleic Acids Res. Oct 30;43(19):9379-92. doi: 10.1093/nar/gkv993. Epub 2015 Oct 1.

Claims

WHAT IS CLAIMED IS:
1. A method for activating or modulating homologous recombination in a cell comprising:
(a) promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1 -PALB2-BRCA2 complexes in the cell;
(b) contacting the cell with BRCA1-PALB2 or BRCA1-PALB2-BRCA2 complexes;
(c) activating or stimulating BRCA1 recruitment to DNA double-strand break (DSB) sites;
(d) inhibiting or removing KEAP1 or CRL3-KEAP1 ;
(e) inhibiting the degradation of USP11 or promoting USP11 activity; and/or
(f) inhibiting DCAF10.
2. The method of claim 1 comprising inhibiting KEAP1 or DCAF10.
3. The method of claim 1 or 2 further comprising reconstituting or activating DNA end resection.
4. The method for activating homologous recombination in a cell in which DNA end resection is activated or reconstituted generating single-stranded DNA, comprising promoting or stimulating the assembly or occurrence of BRCA1 -PALB2 or BRCA1- PALB2-BRCA2 complexes in the cell.
5. The method of claim 3 or 4 wherein DNA end resection is activated by inhibiting 53BP1 expression or activity and upregulating or expressing CtlP.
6. The method of claim 5 wherein CtlP is upregulated or expressed by administering CtlP or an analog of CtlP.
7. The method of claim 3 comprising inhibiting or removing KEAP1 and 53BP1 and upregulating or expressing CtlP or an analog of CtlP.
8. The method of claim 5, 6, or 7 wherein 53BP1 is inhibited by administrating
53BP1 short interfering (si) RNA, short hairpin (sh) RNA or microRNAs (miRNAs).
9. A method of any one of claims 1 to 8 further comprising introducing into the cell a gene editing system.
10. A method of claim 9 wherein the gene editing system is a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR associated (Cas) system, a Transcription Activator-Like Effector Nucleases (TALEN) system or a zinc finger nucleases system.
11. A method for altering a genetic disorder associated with a target polynucleotide sequence in a cell comprising: (1) activating homologous recombination in the cell in accordance with the method of any one of claims 1 to 8; and (2) contacting the target polynucleotide sequence with a CRISPR-associated (Cas) protein and from one to two ribonucleic acids, wherein the ribonucleic acids direct Cas protein to and hybridize to a selected motif of the target polynucleotide sequence, wherein the target polynucleotide sequence is cleaved whereby the expression of the target polynucleotide sequence is reduced, knocked-out, or corrected from an undesired sequence to a desired sequence
12. A method of any one of claims 1 to 1 1 wherein the cell is in Gl phase of the cell cycle (Gl) or GO phase of the cell cycle (GO).
13. A method of claim 12 wherein the cell is in Gl phase of the cell cycle (Gl).
14. A method for monitoring activity or expression of USP1 1 in a sample comprising (i) isolating complexes of (a) BRCA1 and PALB2; (b) BRCA1 , PALB2, and BRCA2; (c) USP11 and PALB2; and/or (d) USP11 and DCAF10 in the sample; (ii) measuring the levels of the complexes; and (iii) detecting an increase or decrease in the activity or expression of the complexes as compared to a control as an indication of the activity or expression of USP1 1.
15. A method of predicting a response or categorizing a response to a PARP inhibitor in an individual comprising assaying complexes of BRCA1 , PALB2 and
BRCA2 in a sample from the subject and comparing to a control to determine if the individual is sensitive to the PARP inhibitor.
16. A kit for stimulating or activating homologous recombination or gene editing in a cell comprising (a) an inliibitor of 53BP1 ; (b) an inhibitor of KEAPl ; and (c) CtIP or an analog of CtlP.
PCT/CA2016/000057 2015-03-02 2016-03-01 Homologous recombination factors WO2016138574A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA2977685A CA2977685C (en) 2015-03-02 2016-03-01 Homologous recombination factors
EP16758390.5A EP3265560B1 (en) 2015-03-02 2016-03-01 Homologous recombination factors
CN201680017382.6A CN109121418A (en) 2015-03-02 2016-03-01 Homologous recombination factors
US15/554,042 US11261466B2 (en) 2015-03-02 2016-03-01 Homologous recombination factors
JP2017546726A JP6871169B2 (en) 2015-03-02 2016-03-01 Homologous recombination factor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562127013P 2015-03-02 2015-03-02
US62/127,013 2015-03-02
US201562222542P 2015-09-23 2015-09-23
US62/222,542 2015-09-23

Publications (1)

Publication Number Publication Date
WO2016138574A1 true WO2016138574A1 (en) 2016-09-09

Family

ID=56849143

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2016/000057 WO2016138574A1 (en) 2015-03-02 2016-03-01 Homologous recombination factors

Country Status (6)

Country Link
US (1) US11261466B2 (en)
EP (1) EP3265560B1 (en)
JP (1) JP6871169B2 (en)
CN (1) CN109121418A (en)
CA (1) CA2977685C (en)
WO (1) WO2016138574A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107119077A (en) * 2017-05-16 2017-09-01 上海交通大学 The new application of CtIP inhibitor and a kind of accurately genomic DNA fragment edit methods
US10208317B2 (en) 2013-12-11 2019-02-19 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse embryonic stem cell genome
US10385359B2 (en) 2013-04-16 2019-08-20 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10457960B2 (en) 2014-11-21 2019-10-29 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification using paired guide RNAs
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
EP3864169A4 (en) * 2018-10-12 2022-07-06 Children's Hospital Medical Center Modular expression systems for gene expression and methods of using same
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110499305A (en) * 2018-05-18 2019-11-26 南京理工大学 Expression vector, construction method and its expression and purification of nuclease protein Cas9
EP3826675A1 (en) * 2018-07-24 2021-06-02 January Therapeutics, Inc. Nanoparticle compositions
US20210277421A1 (en) * 2018-08-06 2021-09-09 NemaMetrix, Inc Homologous Recombination Reporter Construct and Uses Thereof
EP3965831A4 (en) * 2019-05-07 2022-12-21 Board of Regents, The University of Texas System Targeting otub1 in immunotherapy
JP2022542359A (en) * 2019-07-22 2022-10-03 ザ・トラスティーズ・オブ・コロンビア・ユニバーシティ・イン・ザ・シティ・オブ・ニューヨーク Increased genomic stability and reprogramming efficiency of induced pluripotent stem cells
CN114736917B (en) * 2022-03-27 2023-08-01 苏州大学 Application of USP11 in inhibiting degradation of cytokine IL6

Citations (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5756510A (en) 1994-03-09 1998-05-26 Newcastle University Ventures Limited Benzamide analogs useful as PARP (ADP-ribosyltransferase, ADPRT) DNA repair enzyme inhibitors
WO1999011623A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Thioalkyl compounds, methods, and compositions for inhibiting parp activity
WO1999011622A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Amino-substituted compounds, methods, and compositions for inhibiting parp activity
WO1999011628A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhi biting parp activity
WO1999011644A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
WO1999011649A2 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
WO1999011945A1 (en) 1997-09-03 1999-03-11 Yang Ge Industrial Co., Ltd. Hub structure
USRE36397E (en) 1994-02-04 1999-11-16 The John Hopkins University Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
WO1999059975A1 (en) 1998-05-15 1999-11-25 Guilford Pharmaceuticals Inc. Fused tricyclic compounds which inhibit parp activity
WO1999059973A1 (en) 1998-05-15 1999-11-25 Guilford Pharmaceuticals Inc. Carboxamide compounds, compositions, and methods for inhibiting parp activity
WO2000039070A1 (en) 1998-12-31 2000-07-06 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
WO2000039104A1 (en) 1998-12-31 2000-07-06 Guilford Pharmaceuticals, Inc. Phenazine compounds, methods and pharmaceutical compositions for inhibiting parp
WO2000042040A1 (en) 1999-01-11 2000-07-20 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(adp-ribose) polymerases
US6346536B1 (en) 1997-09-03 2002-02-12 Guilford Pharmaceuticals Inc. Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same
US6380193B1 (en) 1998-05-15 2002-04-30 Guilford Pharmaceuticals Inc. Fused tricyclic compounds, methods and compositions for inhibiting PARP activity
US6395749B1 (en) 1998-05-15 2002-05-28 Guilford Pharmaceuticals Inc. Carboxamide compounds, methods, and compositions for inhibiting PARP activity
US6413725B1 (en) 1998-08-07 2002-07-02 California Institute Of Technology Biochemical assay to monitor the ubiquitin ligase activities of cullins
US6444676B1 (en) 1999-12-20 2002-09-03 Iok-Hou Pang Use of PARP inhibitors in the treatment of glaucoma
EP1268847A2 (en) 2000-04-03 2003-01-02 Rigel Pharmaceuticals, Inc. Ubiquitin ligase assay
US6737421B1 (en) 1999-04-22 2004-05-18 Abbott Gmbh & Co. Kg Cyclo-alkyl substituted benzimidazoles and their use as PARP inhibitors
US6924284B2 (en) 2001-08-15 2005-08-02 Icos Corporation PARP inhibitors
US7041675B2 (en) 2000-02-01 2006-05-09 Abbott Gmbh & Co. Kg Heterocyclic compounds and their use as PARP inhibitors
US7087637B2 (en) 2000-05-11 2006-08-08 Basf Ag Substituted indoles which are PARP inhibitors
US7462724B2 (en) 2005-11-15 2008-12-09 Abbott Laboratories Substituted 1H-benzimidazole-4-carboxamides are potent PARP inhibitors
US7601719B2 (en) 2003-05-28 2009-10-13 Eisai Corporation Of North America Compounds, methods and pharmaceutical compositions for inhibiting PARP
US7652014B2 (en) 2003-12-10 2010-01-26 Janssen Pharmaceutica Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US7728026B2 (en) 2005-04-11 2010-06-01 Abbott Laboratories, Inc. 2-substituted-1 h-benzimidazile-4-carboxamides are PARP inhibitors
US7732491B2 (en) 2007-11-12 2010-06-08 Bipar Sciences, Inc. Treatment of breast cancer with a PARP inhibitor alone or in combination with anti-tumor agents
US7820668B2 (en) 2005-01-19 2010-10-26 Eisai Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP
US7875621B2 (en) 2004-06-30 2011-01-25 Janssen Pharmaceutica Nv Substituted 2-alkyl quinazolinone derivatives as PARP inhibitors
US7956064B2 (en) 2006-09-01 2011-06-07 Cylene Pharmaceuticals, Inc. Fused tricyclic compounds as serine-threonine protein kinase and PARP modulators
US20110145940A1 (en) 2009-12-10 2011-06-16 Voytas Daniel F Tal effector-mediated dna modification
US7999117B2 (en) 2006-05-02 2011-08-16 Abbott Lab Substituted 1H-benzimidazole-4-carboxamides are potent PARP inhibitors
US8008491B2 (en) 2003-09-04 2011-08-30 Aventis Pharmaceuticals, Inc. Substituted AZA-indoles as inhibitors of poly(ADP-ribose) polymerase (PARP)
US8012976B2 (en) 2008-08-06 2011-09-06 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US8071623B2 (en) 2007-01-10 2011-12-06 Instituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Amide substituted indazoles as poly(ADP-ribose)polymerase(PARP) inhibitors
US8080557B2 (en) 2004-06-30 2011-12-20 Janssen Pharmaceutica, Nv Quinazolinedione derivatives as PARP inhibitors
US8088760B2 (en) 2008-02-06 2012-01-03 Biomarin Pharmaceutical Inc. Benzoxazole carboxamide inhibitors of poly(ADP-ribose)polymerase (PARP)
US8173682B2 (en) 2004-03-30 2012-05-08 Aventis Pharmaceuticals Inc. Substituted pyridones as inhibitors of poly(ADP-ribose) polymerase (PARP)
US8183250B2 (en) 2006-06-20 2012-05-22 Abbott Laboratories Potent PARP inhibitors
US8188084B2 (en) 2006-05-31 2012-05-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa. Pyridinone and pyridazinone derivatives as inhibitors of poly (ADP-ribose) polymerase (PARP)
US8236802B2 (en) 2007-10-03 2012-08-07 Eisai Inc. PARP inhibitor compounds, compositions and methods of use
US8362030B2 (en) 2008-03-14 2013-01-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.R.L. Tricyclic derivatives as inhibitors of poly(ADP-ribose) polymerase (PARP)
US20130116152A1 (en) 2010-04-29 2013-05-09 Jonathan Peter Clark Ubiquitination assay
US8541417B2 (en) 2009-07-30 2013-09-24 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
US8541403B2 (en) 2010-02-03 2013-09-24 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP) for use in treatment of diseases associated with a PTEN deficiency
US8546368B2 (en) 2006-02-15 2013-10-01 Abbvie Inc. Pyrazoloquinolones are potent PARP inhibitors
US8592416B2 (en) 2009-05-21 2013-11-26 Nerviano Medical Sciences S.R.L. Isoquinolin-1 (2H)-one derivatives as PARP-1 inhibitors
WO2014018423A2 (en) 2012-07-25 2014-01-30 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
US8669249B2 (en) 2009-03-27 2014-03-11 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
WO2014093718A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
WO2014093622A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
WO2014093701A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
WO2014093595A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
WO2014093655A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
WO2014093709A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
WO2014093635A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
WO2014093712A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093694A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
US8778966B2 (en) 2007-03-08 2014-07-15 Janssen Pharmaceutica, Nv Quinolinone derivatives as PARP and tank inhibitors
US20140287938A1 (en) 2013-03-15 2014-09-25 The Broad Institute, Inc. Recombinant virus and preparations thereof
WO2014204725A1 (en) * 2013-06-17 2014-12-24 The Broad Institute Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
US8946221B2 (en) 2004-06-30 2015-02-03 Janssen Pharmaceutica, Nv Phthalazine derivatives as PARP inhibitors
US20150232883A1 (en) 2013-12-12 2015-08-20 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160040155A1 (en) * 2013-04-16 2016-02-11 University Of Washington Through Its Center For Commercialization Activating an alternative pathway for homology-directed repair to stimulate targeted gene correction and genome engineering

Patent Citations (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE36397E (en) 1994-02-04 1999-11-16 The John Hopkins University Inhibitors of poly(ADP-ribose) synthetase and use thereof to treat NMDA neurotoxicity
US5756510A (en) 1994-03-09 1998-05-26 Newcastle University Ventures Limited Benzamide analogs useful as PARP (ADP-ribosyltransferase, ADPRT) DNA repair enzyme inhibitors
US6197785B1 (en) 1997-09-03 2001-03-06 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
WO1999011623A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Thioalkyl compounds, methods, and compositions for inhibiting parp activity
WO1999011622A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Amino-substituted compounds, methods, and compositions for inhibiting parp activity
WO1999011628A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Alkoxy-substituted compounds, methods, and compositions for inhi biting parp activity
WO1999011644A1 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Di-n-heterocyclic compounds, methods, and compositions for inhibiting parp activity
WO1999011649A2 (en) 1997-09-03 1999-03-11 Guilford Pharmaceuticals Inc. Parp inhibitors, pharmaceutical compositions comprising same, and methods of using same
WO1999011945A1 (en) 1997-09-03 1999-03-11 Yang Ge Industrial Co., Ltd. Hub structure
US6635642B1 (en) 1997-09-03 2003-10-21 Guilford Pharmaceuticals Inc. PARP inhibitors, pharmaceutical compositions comprising same, and methods of using same
US6380211B1 (en) 1997-09-03 2002-04-30 Guilford Pharmaceutical Inc. Alkoxy-substituted compounds, methods, and compositions for inhibiting PARP activity
US6346536B1 (en) 1997-09-03 2002-02-12 Guilford Pharmaceuticals Inc. Poly(ADP-ribose) polymerase inhibitors and method for treating neural or cardiovascular tissue damage using the same
WO1999059975A1 (en) 1998-05-15 1999-11-25 Guilford Pharmaceuticals Inc. Fused tricyclic compounds which inhibit parp activity
WO1999059973A1 (en) 1998-05-15 1999-11-25 Guilford Pharmaceuticals Inc. Carboxamide compounds, compositions, and methods for inhibiting parp activity
US6380193B1 (en) 1998-05-15 2002-04-30 Guilford Pharmaceuticals Inc. Fused tricyclic compounds, methods and compositions for inhibiting PARP activity
US6395749B1 (en) 1998-05-15 2002-05-28 Guilford Pharmaceuticals Inc. Carboxamide compounds, methods, and compositions for inhibiting PARP activity
US6413725B1 (en) 1998-08-07 2002-07-02 California Institute Of Technology Biochemical assay to monitor the ubiquitin ligase activities of cullins
WO2000039104A1 (en) 1998-12-31 2000-07-06 Guilford Pharmaceuticals, Inc. Phenazine compounds, methods and pharmaceutical compositions for inhibiting parp
WO2000039070A1 (en) 1998-12-31 2000-07-06 Guilford Pharmaceuticals, Inc. Ortho-diphenol compounds, methods and pharmaceutical compositions for inhibiting parp
US6977298B2 (en) 1999-01-11 2005-12-20 Agouron Pharmacetucals, Inc. Tricyclic inhibitors of poly(ADP-ribose) polymerases
US6495541B1 (en) 1999-01-11 2002-12-17 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(ADP-ribose) polymerases
WO2000042040A1 (en) 1999-01-11 2000-07-20 Agouron Pharmaceuticals, Inc. Tricyclic inhibitors of poly(adp-ribose) polymerases
US6737421B1 (en) 1999-04-22 2004-05-18 Abbott Gmbh & Co. Kg Cyclo-alkyl substituted benzimidazoles and their use as PARP inhibitors
US6444676B1 (en) 1999-12-20 2002-09-03 Iok-Hou Pang Use of PARP inhibitors in the treatment of glaucoma
US7041675B2 (en) 2000-02-01 2006-05-09 Abbott Gmbh & Co. Kg Heterocyclic compounds and their use as PARP inhibitors
EP1268847A2 (en) 2000-04-03 2003-01-02 Rigel Pharmaceuticals, Inc. Ubiquitin ligase assay
US7087637B2 (en) 2000-05-11 2006-08-08 Basf Ag Substituted indoles which are PARP inhibitors
US6924284B2 (en) 2001-08-15 2005-08-02 Icos Corporation PARP inhibitors
US7750008B2 (en) 2003-05-28 2010-07-06 Eisai Inc. Compounds, methods and pharmaceutical compositions for inhibiting PARP
US7601719B2 (en) 2003-05-28 2009-10-13 Eisai Corporation Of North America Compounds, methods and pharmaceutical compositions for inhibiting PARP
US8129382B2 (en) 2003-05-28 2012-03-06 Eisai Inc. Compounds, methods and pharmaceutical compositions for inhibiting PARP
US8008491B2 (en) 2003-09-04 2011-08-30 Aventis Pharmaceuticals, Inc. Substituted AZA-indoles as inhibitors of poly(ADP-ribose) polymerase (PARP)
US7652014B2 (en) 2003-12-10 2010-01-26 Janssen Pharmaceutica Substituted 6-cyclohexylalkyl substituted 2-quinolinones and 2-quinoxalinones as poly(ADP-ribose) polymerase inhibitors
US8173682B2 (en) 2004-03-30 2012-05-08 Aventis Pharmaceuticals Inc. Substituted pyridones as inhibitors of poly(ADP-ribose) polymerase (PARP)
US8623884B2 (en) 2004-06-30 2014-01-07 Janssen Pharmaceutica, Nv Quinazolinedione derivatives as PARP inhibitors
US8188103B2 (en) 2004-06-30 2012-05-29 Janssen Pharmaceutica Nv Substituted 2-alkyl quinazolinone derivatives as PARP inhibitors
US7875621B2 (en) 2004-06-30 2011-01-25 Janssen Pharmaceutica Nv Substituted 2-alkyl quinazolinone derivatives as PARP inhibitors
US8946221B2 (en) 2004-06-30 2015-02-03 Janssen Pharmaceutica, Nv Phthalazine derivatives as PARP inhibitors
US8080557B2 (en) 2004-06-30 2011-12-20 Janssen Pharmaceutica, Nv Quinazolinedione derivatives as PARP inhibitors
US8058275B2 (en) 2005-01-19 2011-11-15 Eisai Inc. Diazabenzo[de] anthracen-3-one compounds and methods for inhibiting PARP
US7820668B2 (en) 2005-01-19 2010-10-26 Eisai Inc. Diazabenzo[de]anthracen-3-one compounds and methods for inhibiting PARP
US7728026B2 (en) 2005-04-11 2010-06-01 Abbott Laboratories, Inc. 2-substituted-1 h-benzimidazile-4-carboxamides are PARP inhibitors
US8217070B2 (en) 2005-04-11 2012-07-10 Abbott Laboratories 2-substituted-1H-benzimidazole-4-carboxamides are PARP inhibitors
US7462724B2 (en) 2005-11-15 2008-12-09 Abbott Laboratories Substituted 1H-benzimidazole-4-carboxamides are potent PARP inhibitors
US8546368B2 (en) 2006-02-15 2013-10-01 Abbvie Inc. Pyrazoloquinolones are potent PARP inhibitors
US7999117B2 (en) 2006-05-02 2011-08-16 Abbott Lab Substituted 1H-benzimidazole-4-carboxamides are potent PARP inhibitors
US8188084B2 (en) 2006-05-31 2012-05-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa. Pyridinone and pyridazinone derivatives as inhibitors of poly (ADP-ribose) polymerase (PARP)
US8183250B2 (en) 2006-06-20 2012-05-22 Abbott Laboratories Potent PARP inhibitors
US7956064B2 (en) 2006-09-01 2011-06-07 Cylene Pharmaceuticals, Inc. Fused tricyclic compounds as serine-threonine protein kinase and PARP modulators
US8071623B2 (en) 2007-01-10 2011-12-06 Instituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Amide substituted indazoles as poly(ADP-ribose)polymerase(PARP) inhibitors
US8778966B2 (en) 2007-03-08 2014-07-15 Janssen Pharmaceutica, Nv Quinolinone derivatives as PARP and tank inhibitors
US8236802B2 (en) 2007-10-03 2012-08-07 Eisai Inc. PARP inhibitor compounds, compositions and methods of use
US8894989B2 (en) 2007-10-03 2014-11-25 Eisai Inc. PARP inhibitor compounds, compositions and methods of use
US7732491B2 (en) 2007-11-12 2010-06-08 Bipar Sciences, Inc. Treatment of breast cancer with a PARP inhibitor alone or in combination with anti-tumor agents
US8088760B2 (en) 2008-02-06 2012-01-03 Biomarin Pharmaceutical Inc. Benzoxazole carboxamide inhibitors of poly(ADP-ribose)polymerase (PARP)
US8362030B2 (en) 2008-03-14 2013-01-29 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.R.L. Tricyclic derivatives as inhibitors of poly(ADP-ribose) polymerase (PARP)
US8420650B2 (en) 2008-08-06 2013-04-16 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US8012976B2 (en) 2008-08-06 2011-09-06 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US8669249B2 (en) 2009-03-27 2014-03-11 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
US8592416B2 (en) 2009-05-21 2013-11-26 Nerviano Medical Sciences S.R.L. Isoquinolin-1 (2H)-one derivatives as PARP-1 inhibitors
US8541417B2 (en) 2009-07-30 2013-09-24 Takeda Pharmaceutical Company Limited Poly (ADP-ribose) polymerase (PARP) inhibitors
US20110145940A1 (en) 2009-12-10 2011-06-16 Voytas Daniel F Tal effector-mediated dna modification
US8541403B2 (en) 2010-02-03 2013-09-24 Biomarin Pharmaceutical Inc. Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP) for use in treatment of diseases associated with a PTEN deficiency
US20130116152A1 (en) 2010-04-29 2013-05-09 Jonathan Peter Clark Ubiquitination assay
WO2014018423A2 (en) 2012-07-25 2014-01-30 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
EP2764103A2 (en) 2012-12-12 2014-08-13 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
WO2014093709A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
WO2014093655A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140242664A1 (en) 2012-12-12 2014-08-28 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093635A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140170753A1 (en) 2012-12-12 2014-06-19 Massachusetts Institute Of Technology Crispr-cas systems and methods for altering expression of gene products
WO2014093712A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093694A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
WO2014093661A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US20140179006A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Crispr-cas component systems, methods and compositions for sequence manipulation
US20140179770A1 (en) 2012-12-12 2014-06-26 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20140189896A1 (en) 2012-12-12 2014-07-03 Feng Zhang Crispr-cas component systems, methods and compositions for sequence manipulation
US20140186843A1 (en) 2012-12-12 2014-07-03 Massachusetts Institute Of Technology Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US20140186919A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140186958A1 (en) 2012-12-12 2014-07-03 Feng Zhang Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140242699A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
WO2014093701A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
US8795965B2 (en) 2012-12-12 2014-08-05 The Broad Institute, Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
WO2014093622A2 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US20140227787A1 (en) 2012-12-12 2014-08-14 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US20150247150A1 (en) 2012-12-12 2015-09-03 The Broad Institute Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2014093595A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
US8771945B1 (en) 2012-12-12 2014-07-08 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
US20140242700A1 (en) 2012-12-12 2014-08-28 Massachusetts Institute Of Technology Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP2771468A1 (en) 2012-12-12 2014-09-03 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140248702A1 (en) 2012-12-12 2014-09-04 The Broad Institute, Inc. CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US20140256046A1 (en) 2012-12-12 2014-09-11 Massachusetts Institute Of Technology Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US20140273234A1 (en) 2012-12-12 2014-09-18 The Board Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US20140273232A1 (en) 2012-12-12 2014-09-18 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140273231A1 (en) 2012-12-12 2014-09-18 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
US20140234972A1 (en) 2012-12-12 2014-08-21 Massachusetts Institute Of Technology CRISPR-CAS Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
EP2784162A1 (en) 2012-12-12 2014-10-01 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US8865406B2 (en) 2012-12-12 2014-10-21 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8871445B2 (en) 2012-12-12 2014-10-28 The Broad Institute Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
US8889418B2 (en) 2012-12-12 2014-11-18 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8889356B2 (en) 2012-12-12 2014-11-18 The Broad Institute Inc. CRISPR-Cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
WO2014093718A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US8895308B1 (en) 2012-12-12 2014-11-25 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
US20140287938A1 (en) 2013-03-15 2014-09-25 The Broad Institute, Inc. Recombinant virus and preparations thereof
WO2014204725A1 (en) * 2013-06-17 2014-12-24 The Broad Institute Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
US20150232883A1 (en) 2013-12-12 2015-08-20 The Broad Institute Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components

Non-Patent Citations (98)

* Cited by examiner, † Cited by third party
Title
"Methods in Enzymology", vol. 304, 1999, ACADEMIC PRESS, article "Chromatin"
"Methods in Molecular Biology", vol. 119, 1999, HUMANA PRESS, article "Chromatin Protocols"
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1987, JOHN WILEY & SONS
BANASIK ET AL.: "Inhibitors and Activators of ADP-Ribosylation Reactions", MOLEC. CELL. BIOCHEM., vol. 138, 1994, pages 185 - 97, XP009050157, DOI: 10.1007/BF00928461
BANASIK ET AL.: "Specific Inhibitors of Poly(ADP-Ribose) Synthetase and Mono(ADP-Ribosyl)-Transferase", J. BIOL. CHEM., vol. 267, no. 3, 1992, pages 1569 - 75
BHATTACHARYYA, A.EAR, U. S.KOLLER, B. H.WEICHSELBAUM, R. R.BISHOP, D. K.: "The breast cancer susceptibility gene BRCA1 is required for subnuclear assembly of Rad51 and survival following treatment with the DNA cross-linking agent cisplatin", J BIOL CHEM, vol. 275, 2000, pages 23899 - 23903, XP002992275, DOI: 10.1074/jbc.C000276200
BIBIKOVA M. ET AL., GENETICS, vol. 161, no. 3, 2002, pages 1169 - 1175
BUNTING, S. F. ET AL.: "53BP1 inhibits homologous recombination in Brcal-deficient cells by blocking resection of DNA breaks", CELL, vol. 141, 2010, pages 243 - 254, XP055430025, DOI: 10.1016/j.cell.2010.03.012
CANNING P. ET AL., ACTA PHARM SIN B., vol. 4, 2015, pages 285 - 99
CHAPMAN, J. R. ET AL.: "RIF1 Is Essential for 53BP1-Dependent Nonhomologous End Joining and Suppression of DNA Double-Strand Break Resection", MOL CELL, 2013
CONG, L. ET AL., SCIENCE, vol. 339, no. 6121, 15 February 2013 (2013-02-15), pages 819 - 23
CUI, J. ET AL.: "Glutamine deamidation and dysfunction of ubiquitin/NEDD8 induced by abacterial effector family", SCIENCE, vol. 329, 2010, pages 1215 - 1218
DOENCH ET AL., NATURE BIOTECHNOLOGY, 3 September 2014 (2014-09-03)
ENCHEV, R. I.SCHREIBER, A.BEURON, F.MORRIS, E. P.: "Structural insights into the COP9 signalosome and its common architecture with the 26S proteasome lid and eIF3", STRUCTURE, vol. 18, 2010, pages 518 - 527, XP026999369
ENCHEV, R. I.SCHULMAN, B. A.PETER, M.: "Protein neddylation: beyond cullin-RING ligases", NAT REV MOL CELL BIOL, vol. 16, 2015, pages 30 - 44
ESCRIBANO-DIAZ ET AL.: "A Cell Cycle Dependent Regulatory Circuit Composed of 53BP1-RF1 and BRCA1-CtIP Controls DNA Repair Pathway Choice", MOLECULAR CELL, vol. 49, 7 March 2013 (2013-03-07), pages 872 - 883, XP028590116, ISSN: 1097-2765, DOI: doi:10.1016/j.molcel.2013.01.001 *
ESCRIBANO-DIAZ, C. ET AL.: "A Cell Cycle-Dependent Regulatory Circuit Composed of 53BP1-RIF1 and BRCAl-CtIP Controls DNA Repair Pathway Choice", MOLECULAR CELL, vol. 49, 2013, pages 872 - 883, XP028590116, DOI: 10.1016/j.molcel.2013.01.001
FENG, L.FONG, K. W.WANG, J.WANG, W.CHEN, J.: "RIF1 counteracts BRCA1-mediated end resection during DNA repair", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 288, 2013, pages 11135 - 11143
FRADET-TURCOTTE, A. ET AL.: "53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark", NATURE, vol. 499, 2013, pages 50 - 54
FUKUDA T. ET AL., CANCER SCI., vol. 106, no. 8, August 2015 (2015-08-01), pages 1050 - 6
GAJ T. ET AL., TRENDS BIOTECHNOL., vol. 31, no. 7, July 2013 (2013-07-01), pages 397 - 405
GENSCHIK, P.SUMARA, I.LECHNER, E.: "The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): cellular functions and disease implications", EMBO J, vol. 32, 2013, pages 2307 - 2320
GUNTAS, G.LEWIS, S.M.MULVANEY, K.M.CLOER, E.W.TRIPATHY, A.LANE, T.R.MAJOR, M.B.KUHLMAN, B.: "Engineering a genetically encoded competitive inhibitor of the KEAP1-NRF2 interaction via structure-based design and phage display", PROTEIN ENGINEERING, DESIGN & SELECTION : PEDS, vol. 29, 2016, pages 1 - 9
HARTLERODE, A.ODATE, S.SHIM, I.BROWN, J.SCULLY, R.: "Cell cycle-dependent induction of homologous recombination by a tightly regulated I-Scel fusion protein", PLOS ONE, vol. 6, 2011, pages e16501
HENDRIKS, I. A.SCHIMMEL, J.EIFLER, K.OLSEN, J. V.VERTEGAAL, A. C.: "Ubiquitin- specific Protease 11 (USP11) Deubiquitinates Hybrid Small Ubiquitin-like Modifier (SUMO)-Ubiquitin Chains to Counteract RING Finger Protein 4 (RNF4", J BIOL CHEM, vol. 290, 2015, pages 15526 - 15537
HSU ET AL., CELL, vol. 157, 5 June 2014 (2014-06-05), pages 1262 - 1278
HSU, P. ET AL., NAT BIOTECHNOL, 2013
HUERTAS, P, NAT STRUCT MOL BIOL, vol. 17, no. 10, pages 11 - 16
HUERTAS, P.JACKSON, S. P.: "Human CtIP mediates cell cycle control of DNA end resection and double strand break repair", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 284, 2009, pages 9558 - 9565
JACKSON, S.XIONG, Y.: "CRL4s: the CUL4-RING E3 ubiquitin ligases", TRENDS BIOCHEM SCI, vol. 34, 2009, pages 562 - 570, XP026736004, DOI: 10.1016/j.tibs.2009.07.002
JARVIUS M. ET AL., MOL. CELL. PROTEOMICS, vol. 6, no. 9, September 2007 (2007-09-01), pages 1500 - 9
JASIN, M.ROTHSTEIN, R.: "Repair of strand breaks by homologous recombination", COLD SPRING HARB PERSPECT BIOL, vol. 5, 2013, pages a012740, XP055269842, DOI: 10.1101/cshperspect.a012740
JIANG W. ET AL., NAT BIOTECHNOL, vol. 31, no. 3, March 2013 (2013-03-01), pages 233 - 9
JIMENO S. ET AL., NUCL. ACIDS RES, 2015
JUANG, Y. C. ET AL.: "OTUB 1 co-opts Lys48-linked ubiquitin recognition to suppress E2 enzyme function", MOL CELL, vol. 45, 2012, pages 384 - 397, XP028455128, DOI: 10.1016/j.molcel.2012.01.011
KASPAREK, T. R.HUMPHREY, T. C.: "DNA double-strand break repair pathways, chromosomal rearrangements and cancer", SEMIN CELL DEV BIOL, vol. 22, 2011, pages 886 - 897, XP028116410, DOI: 10.1016/j.semcdb.2011.10.007
KONERMANN S ET AL., NATURE, vol. 500, no. 7463, 22 August 2013 (2013-08-22), pages 472 - 6
KONSTANTIN ET AL.: "Synthetic lethality of PARP inhibition in cancers lacking BRCA1 and BRCA2 mutations", CELL CYCLE, vol. 10, no. 8, 15 April 2011 (2011-04-15), pages 1192 - 1199, XP055482764, ISSN: 1538-4101 *
LI, M. L.GREENBERG, R. A.: "Links between genome integrity and BRCA1 tumor suppression", TRENDS BIOCHEM SCI, 2012
LOMBARDO A. ET AL., NAT. METHODS., vol. 8, no. 10, 2011, pages 861 - 869
LONG, L.FURGASON, M.YAO, T.: "Generation of nonhydrolyzable ubiquitin-histone mimics", METHODS, vol. 70, 2014, pages 134 - 138
MA, J. ET AL.: "PALB2 interacts with KEAP1 to promote NRF2 nuclear accumulation and function", MOL CELL BIOL, vol. 32, 2012, pages 1506 - 1517, XP055487139, DOI: 10.1128/MCB.06271-11
MONTONI ET AL., FRONTIERS IN PHARMACOLOGY, vol. 4, pages 1 - 7
MOYNAHAN, M.E.CHIU, J.W.KOLLER, B.H.JASIN, M.: "Brcal controls homology-directed DNA repair", MOL CELL, vol. 4, 1999, pages 511 - 518
NEMUDRYI A.A. ET AL., ACTA NATURAE, vol. 6, no. 3, July 2014 (2014-07-01), pages 19 - 40
NIMONKAR A.V. ET AL., GENES & DEVELOPMENT, vol. 25, 2011, pages 350 - 362
NISHIMASU, H. ET AL., CELL, vol. 156, no. 5, 27 February 2014 (2014-02-27), pages 935 - 49
O'CONNOR T.P.R.G. CRYSTAL, NATURE REVIEWS/GENETICS VOLUME, 7 April 2006 (2006-04-07), pages 261 - 276
ORTHWEIN ET AL.: "A mechanism for the suppression of homologous recombination in G 1 cells", NATURE, vol. 528, 17 December 2015 (2015-12-17), pages 422 - 426, XP055404677, ISSN: 0028-0836 *
ORTHWEIN, A. ET AL.: "Mitosis inhibits DNA double-strand break repair to guard against telomere fusions", SCIENCE, vol. 344, 2014, pages 189 - 193, XP055447969, DOI: 10.1126/science.1248024
PANIER, S. ET AL.: "Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks", MOLECULAR CELL, vol. 47, 2012, pages 383 - 395
PANIER, S.DUROCHER, D.: "Push back to respond better: regulatory inhibition of the DNA double-strand break response", NATURE REVIEWS. MOLECULAR CELL BIOLOGY, 2013
PARK, J. Y.ZHANG, F.ANDREASSEN, P. R.: "PALB2: The hub of a network of tumor suppressors involved in DNA damage responses", BIOCHIM BIOPHYS ACTA, vol. 1846, 2014, pages 263 - 275
PINDER J. ET AL., NUCLEI ACIDS RES., vol. 43, 2015, pages 9379 - 9392
PINDER, J.SALSMAN, J.DELLAIRE, G.: "Nuclear domain 'knock-in' screen for the evaluation and identification of small molecule enhancers of CRISPR-based genome editing", NUCLEIC ACIDS RES., vol. 43, no. 19, 30 October 2015 (2015-10-30), pages 9379 - 92, XP055342317, DOI: 10.1093/nar/gkv993
PLATT ET AL., CELL, vol. 159, no. 2, 2014, pages 440 - 455
PROVASI E. ET AL., J.. NAT. MED., vol. 18, no. 5, 2012, pages 807 - 8151
RAN, F A. ET AL., CELL, 28 August 2013 (2013-08-28)
RAN, F A. ET AL., NATURE PROTOCOLS, vol. 8, no. 11, November 2013 (2013-11-01), pages 2281 - 308
RAN, F. A. ET AL.: "Genome engineering using the CRISPR-Cas9 system", NAT. PROTOCOLS, vol. 8, 2013, pages 2281 - 2308, XP009174668, DOI: 10.1038/nprot.2013.143
ROTHKAMM, K.KRUGER, I.THOMPSON, L. H.LOBRICH, M.: "Pathways of DNA doublestrand break repair during the mammalian cell cycle", MOL CELL BIOL, vol. 23, 2003, pages 5706 - 5715
ROY, R.CHUN, J.POWELL, S. N.: "BRCA1 and BRCA2: different roles in a common pathway of genome protection", NAT REV CANCER, vol. 12, 2011, pages 68 - 78, XP055487148, DOI: 10.1038/nrc3181
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
SAN FILIPPO ET AL., ANNU. REV. BIOCHEM., vol. 77, 2008, pages 229 - 57
SCHLEGEL, B. P.JODELKA, F. M.NUNEZ, R.: "BRCA1 promotes induction of ssDNA by ionizing radiation", CANCER RES, vol. 66, 2006, pages 5181 - 5189
SCHOENFELD, A. R.APGAR, S.DOLIOS, G.WANG, R.AARONSON, S. A.: "BRCA2 is ubiquitinated in vivo and interacts with USP11, a deubiquitinating enzyme that exhibits prosurvival function in the cellular response to DNA damage", MOL CELL BIOL, vol. 24, 2004, pages 7444 - 7455, XP055182826, DOI: 10.1128/MCB.24.17.7444-7455.2004
See also references of EP3265560A4
SHALEM, O. ET AL., SCIENCE, 12 December 2013 (2013-12-12)
SHIRLEY ET AL.: "PALB2 is an integral component of the BRCA complex required for homologous recombination repair", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 106, no. 7, 28 April 2009 (2009-04-28), pages 7155 - 7160, XP055482759, ISSN: 1091-6490 *
SIMHADRI, S. ET AL.: "Male Fertility Defect Associated with Disrupted BRCA1-PALB2 Interaction in Mice", J BIOL CHEM, vol. 289, 2014, pages 24617 - 24629
SODERBERG O. ET AL., NAT. METHODS., vol. 3, no. 12, December 2006 (2006-12-01), pages 995 - 1000
SOWA, M. E.BENNETT, E. J.GYGI, S. P.HARPER, J. W.: "Defining the human deubiquitinating enzyme interaction landscape", CELL, vol. 138, 2009, pages 389 - 403, XP055608782, DOI: 10.1016/j.cell.2009.04.042
STARK, J. M.PIERCE, A. J.OH, J.PASTINK, A.JASIN, M.: "Genetic steps of mammalian homologous repair with distinct mutagenic consequences", MOL CELL BIOL, vol. 24, 2004, pages 9305 - 9316, XP055224116, DOI: 10.1128/MCB.24.21.9305-9316.2004
STORRS, THE SCIENTIST, 1 March 2014 (2014-03-01)
SWIECH ET AL., NATURE BIOTECHNOLOGY, 19 October 2014 (2014-10-19)
SY, S. M.HUEN, M. S.CHEN, J.: "PALB2 is an integral component of the BRCA complex required for homologous recombination repair", P NATL ACAD SCI USA, vol. 106, 2009, pages 7155 - 7160, XP055482759, DOI: 10.1073/pnas.0811159106
TAGUCHI, K.MOTOHASHI, H.YAMAMOTO, M.: "Molecular mechanisms of the Keapl-Nrf2 pathway in stress response and cancer evolution", GENES CELLS, vol. 16, 2011, pages 123 - 140
TANG, J. ET AL.: "Acetylation limits 53BP1 association with damaged chromatin to promote homologous recombination", NATURE STRUCTURAL & MOLECULAR BIOLOGY, 2013
TORIKAI H. ET AL., BLOOD, vol. 119, no. 24, 2012, pages 5697 - 5705
TOWNSEND J.A. ET AL., NATURE, vol. 459, no. 7245, 2009, pages 442 - 445
TURITZ COX D.B. ET AL., NATURE MEDICINE, vol. 21, 2015, pages 121 - 131
WANG ET AL., SCIENCE, vol. 343, no. 6166, 3 January 2014 (2014-01-03), pages 80 - 84
WANG H. ET AL., CELL, vol. 153, no. 4, 9 May 2013 (2013-05-09), pages 910 - 8
WELLS, G., BIOCHEM SOC TRANS., vol. 43, no. 4, 2015, pages 674 - 9
WILTSHIRE ET AL., JBC, vol. 285, no. 19, 2010, pages 14565 - 14571
WILTSHIRE ET AL.: "Sensitivity to Poly(ADP-ribose) Polymerase (PARP) Inhibition Identifies Ubiquitin-specific Peptidase 11 ( USP 11) as a Regulator of DNA Double-strand Break Repair", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 285, no. 19, 7 May 2010 (2010-05-07), pages 14565 - 14571, XP002613116, ISSN: 0021-9258, DOI: doi:10.1074/JBC.M110.104745 *
WILTSHIRE, T. D. ET AL.: "Sensitivity to poly(ADP-ribose) polymerase (PARP) inhibition identifies ubiquitin-specific peptidase 11 (USP11) as a regulator of DNA double-strand break repair", J BIOL CHEM, vol. 285, 2010, pages 14565 - 14571, XP002613116, DOI: 10.1074/JBC.M110.104745
WOLFFE: "Chromatin Structure and Function", 1998, ACADEMIC PRESS
WU X. ET AL., NAT BIOTECHNOL., 20 April 2014 (2014-04-20)
XIA, B. ET AL.: "Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2", MOL CELL, vol. 22, 2006, pages 719 - 729, XP002474133, DOI: 10.1016/j.molcel.2006.05.022
YAMANE, A. ET AL.: "RPA accumulation during class switch recombination represents 5'-3' DNA-end resection during the S-G2/M phase of the cell cycle", CELL REP, vol. 3, 2013, pages 138 - 147
YIN, L.KRANTZ, B.RUSSELL, N. S.DESHPANDE, S.WILKINSON, K. D.: "Nonhydrolyzable diubiquitin analogues are inhibitors of ubiquitin conjugation and deconjugation", BIOCHEMISTRY, vol. 39, 2000, pages 10001 - 10010, XP055165402, DOI: 10.1021/bi0007019
ZHANG ET AL.: "PALB2 Links BRCA1 and BRCA2 in the DNA-Damage Response", CURRENT BIOLOGY, vol. 19, no. 6, 24 March 2009 (2009-03-24), pages 524 - 529, XP026027209, ISSN: 0960-9822, DOI: doi:10.1016/j.cub.2009.02.018 *
ZHANG F. ET AL., PROC. NATL. ACAD. SCI. USA., vol. 107, no. 26, 2010, pages 12028 - 12033
ZHANG, F. ET AL.: "PALB2 links BRCA1 and BRCA2 in the DNA-damage response", CURR BIOL, vol. 19, 2009, pages 524 - 529, XP026027209, DOI: 10.1016/j.cub.2009.02.018
ZHANG, F.BICK, G.PARK, J. Y.ANDREASSEN, P. R.: "MDC1 and RNF8 function in a pathway that directs BRCA1-dependent localization of PALB2 required for homologous recombination", J CELL SCI, vol. 125, 2012, pages 6049 - 6057
ZIMMERMANN, M.LOTTERSBERGER, F.BUONOMO, S. B.SFEIR, A.DE LANGE, T.: "53BPlRegulates DSB Repair Using Rifl to Control 5' End Resection", SCIENCE, 2013

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10385359B2 (en) 2013-04-16 2019-08-20 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10975390B2 (en) 2013-04-16 2021-04-13 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10208317B2 (en) 2013-12-11 2019-02-19 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse embryonic stem cell genome
US10711280B2 (en) 2013-12-11 2020-07-14 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse ES cell genome
US11820997B2 (en) 2013-12-11 2023-11-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US10457960B2 (en) 2014-11-21 2019-10-29 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification using paired guide RNAs
US11697828B2 (en) 2014-11-21 2023-07-11 Regeneran Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification using paired guide RNAs
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
CN107119077A (en) * 2017-05-16 2017-09-01 上海交通大学 The new application of CtIP inhibitor and a kind of accurately genomic DNA fragment edit methods
CN107119077B (en) * 2017-05-16 2021-09-21 上海交通大学 New application of CtIP inhibitor and accurate genomic DNA fragment editing method
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
EP3864169A4 (en) * 2018-10-12 2022-07-06 Children's Hospital Medical Center Modular expression systems for gene expression and methods of using same

Also Published As

Publication number Publication date
EP3265560A1 (en) 2018-01-10
US20180073039A1 (en) 2018-03-15
CA2977685A1 (en) 2016-09-09
CN109121418A (en) 2019-01-01
CA2977685C (en) 2024-02-20
EP3265560B1 (en) 2021-12-08
JP2018512393A (en) 2018-05-17
JP6871169B2 (en) 2021-05-12
US11261466B2 (en) 2022-03-01
EP3265560A4 (en) 2018-08-01

Similar Documents

Publication Publication Date Title
CA2977685C (en) Homologous recombination factors
Orthwein et al. A mechanism for the suppression of homologous recombination in G1 cells
Kwasna et al. Discovery and characterization of ZUFSP/ZUP1, a distinct deubiquitinase class important for genome stability
Zhang et al. HDAC6 deacetylates and ubiquitinates MSH2 to maintain proper levels of MutSα
Chen et al. A PARP1–BRG1–SIRT1 axis promotes HR repair by reducing nucleosome density at DNA damage sites
Gallina et al. The ubiquitin ligase RFWD3 is required for translesion DNA synthesis
US10808017B2 (en) Ubiquitin variants and uses therof as 53BP1 inhibitors
Emanuelli et al. Smurf2-mediated stabilization of DNA topoisomerase IIα controls genomic integrity
Hu et al. The ARK assay is a sensitive and versatile method for the global detection of DNA-protein crosslinks
Duan et al. E3 ligase RFWD3 is a novel modulator of stalled fork stability in BRCA2-deficient cells
Zhou et al. TBK1 phosphorylation activates LIR-dependent degradation of the inflammation repressor TNIP1
Strauss et al. MDC1 is ubiquitylated on its tandem BRCT domain and directly binds RAP80 in a UBC13-dependent manner
Salas-Lloret et al. BRCA1/BARD1 ubiquitinates PCNA in unperturbed conditions to promote replication fork stability and continuous DNA synthesis
Bao et al. A di-acetyl-decorated chromatin signature couples liquid condensation to suppress DNA end synapsis
US11897925B2 (en) Treatment of homologous recombination deficient cancers
Nowsheen Regulation of DNA Double Strand Break Repair
Papke Signaling Alterations in PPP2R5D-Variant Disorder
Lyons Using a Chemical Proteomics Approach to Identify Regulatory Mechanisms of the Phosphoprotein Phosphatases
Thakar Assessing the Role of Gap-Avoidance Mechanisms and Nucleosome Assembly Pathways as Guardians of DNA Replication Fork Stability
Brickner Activation and Regulation of the ALKBH3-ASCC Alkylation Repair Pathway
Zhou et al. Dynamics of the DYNLL1/MRE11 complex regulates DNA end resection and recruitment of the Shieldin complex to DSBs
WO2023235313A2 (en) Systems, compositions and methods for identifying e3 ligase substrates by ubiquitin biotinylation
Bei et al. Amplicon structure creates collateral therapeutic vulnerability in cancer
Ting Characterization of Ubiquitin Ligase Targeting by Anticancer Sulfonamides
WO2022241228A2 (en) Variants of sirt6 for use in preventing and/or treating age-related diseases

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16758390

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2977685

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 15554042

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2017546726

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2016758390

Country of ref document: EP