WO2023244870A1 - Compositions et procédés de contrôle de gains de copies régiospécifiques transitoires, d'insertions génomiques et de réarrangements associés à une leucémie de lignée mixte - Google Patents

Compositions et procédés de contrôle de gains de copies régiospécifiques transitoires, d'insertions génomiques et de réarrangements associés à une leucémie de lignée mixte Download PDF

Info

Publication number
WO2023244870A1
WO2023244870A1 PCT/US2023/025793 US2023025793W WO2023244870A1 WO 2023244870 A1 WO2023244870 A1 WO 2023244870A1 US 2023025793 W US2023025793 W US 2023025793W WO 2023244870 A1 WO2023244870 A1 WO 2023244870A1
Authority
WO
WIPO (PCT)
Prior art keywords
kdm3b
kmt2a
ctcf
cells
agent
Prior art date
Application number
PCT/US2023/025793
Other languages
English (en)
Inventor
Johnathan R. WHETSTINE
Zach H. GRAY
Damayanti CHAKRABORTY
Reuben R. DUTTWEILER
Sedona E. MURPHY
Gulnaz ALEKBAEVA
Original Assignee
INSTITUTE FOR CANCER RESEARCH d.b.a THE RESEARCH INSTITUTE OF FOX CHASE CANCER CENTER
The General Hospital Corporation
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 INSTITUTE FOR CANCER RESEARCH d.b.a THE RESEARCH INSTITUTE OF FOX CHASE CANCER CENTER, The General Hospital Corporation filed Critical INSTITUTE FOR CANCER RESEARCH d.b.a THE RESEARCH INSTITUTE OF FOX CHASE CANCER CENTER
Publication of WO2023244870A1 publication Critical patent/WO2023244870A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/20Animals treated with compounds which are neither proteins nor nucleic acids
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • C12N15/1137Non-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 enzymes
    • 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
    • C12YENZYMES
    • C12Y104/00Oxidoreductases acting on the CH-NH2 group of donors (1.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y201/00Transferases transferring one-carbon groups (2.1)
    • C12Y201/01Methyltransferases (2.1.1)

Definitions

  • the invention relates to compositions and methods for control of transient site-specific copy gains, amplifications, rearrangements, and genomic insertions associated with mixed lineage leukemia. More specifically, the invention provides methods for reducing MLL/KMT2A copy number, thereby minimizing amplification and formation of breakpoints causing de novo and therapy induced acute myeloid leukemia (AML) and myelodysplasia (MDS). Furthermore, the data establishes that topoisomerase inhibitors (Doxorubicin) promoted MLL/KMT2 amplification and rearrangements, providing a new strategy for treatment of chemotherapy induced leukemia and MDS.
  • topoisomerase inhibitors Doxorubicin
  • Genomic instability is hallmark of cancer. Cancer cells are often associated with copy number changes (e.g., gains or losses of chromosome arms and/or whole chromosomes along, amplification/deletion of short stretches of genomic regions) and structural variation. (Mishra and Whetstine, 2016) Although these events can be genetically stable, focal DNA copy gains can also be extrachromosomal, and transiently appear and disappear (Bailey et al., 2020; Mishra and Whetstine, 2016). These events can also co-exist within a cell or population of cells (Mishra and Whetstine, 2016; Song et al., 2022). A key question remains as to whether the appearance of low or high copy extrachromosomal amplifications are associated with the integrated events that result in genomic rearrangements, and in turn, genetic heterogeneity.
  • TSSGs transient site-specific extrachromosomal copy gains
  • the first documented enzyme shown to promote selective extrachromosomal TSSGs was the Histone 3 lysine 9/36 (H3K9/36) tri- demethylase KDM4A.
  • AML and myelodysplasia are characterized by genomic amplifications and translocations of the 1 lq23 region, which includes MLL/KMT2A and other target genes(Poppe et al., 2004; Schichman et al., 1994; Tang et al., 2015; Walter et al., 2009).
  • Infant acute leukemias MLL/KMT2A rearrangements are a hallmark of greater than 70 percent of infant leukemias (Rice and Roy, 2020; Woo et al., 2014).
  • MLL translocations are also found in adult primary leukemias and therapy related leukemia(Andersen et al., 2001; Meyer et al., 2018; Super et al., 1993; Winters and Bernt, 2017).
  • a vast array of translocation events and fusion partners have been documented for MLL/KMT2A with some being more prevalent than others (Meyer et al., 2018).
  • MLL-AF4 is most common in ALL
  • AML MLL-AF9 is most frequent (Winters and Bernt, 2017).
  • Chromosome 5 Frequent cytogenetic loss of Chromosome 5 (whole chromosome, loss of long arm 5q, and more focal deletions) is observed in malignant myeloid disorders including MDS and AML (Bacher et al., 2015; Rea et al., 2020; Walter et al., 2009; Zahid et al., 2017). Chromosome 5 loss is observed in de novo AML and chemotherapy induced leukemia (Godley and Larson, 2008). Cytogenetic loss of chromosome 5 has been correlated with poor chemotherapy responses and lower survival. Studies have successfully defined minimal chromosomal locations at 5q31 observed in patients with MDS, AML, and secondary AML. (Liu et al., 2007; Stoddart et al., 2014).
  • compositions and methods for reducing MLL/KMT2A amplifications/rearrangements in a patient in need thereof are disclosed.
  • An exemplary method entails administration of an effective amount of one or more agents including i) a first agent which increases KDM3B expression and function and, or ii) a second agent which inhibits expression or activity of H3K9 methyltransferase, wherein the agents are administered in one or more pharmaceutically acceptable carriers and reducing MLL/KMT2A amplification/rearrangements in said patient.
  • the first agent is a KDM3B agonist
  • the second agent is a G9a inhibitor and both agents are administered.
  • the MLL/KMT2A amplification can be caused by administration of a chemotherapeutic agent such as a chemotherapeutic agent that reduces KDM3B expression.
  • a chemotherapeutic agent such as a chemotherapeutic agent that reduces KDM3B expression.
  • the chemotherapeutic agent is a topoisomerase inhibitor, such as a topoisomerase II inhibitor.
  • the topoisomerase II inhibitors include without limitation doxorubicin, daunorubin, and etoposide.
  • the patient has a genetic loss of KDM3B.
  • the method further comprises administration of a chemotherapeutic agent.
  • the administration of agents which reduce MLL/KMT2A amplification can be performed before, during or after administration of said chemotherapeutic agent.
  • the patient has reduced MLL/KMT2A copy numbers after treatment when compared to an untreated control.
  • the patient is treated with a third agent, i.e., a proteosome inhibitor selected from bortezomib, carfilzomib, and ixazomib.
  • a third agent i.e., a proteosome inhibitor selected from bortezomib, carfilzomib, and ixazomib.
  • UNC 0642 an inhibitor of G9a, an siRNA that targets H3K9 methyltransferase, and/or an siRNA that targets the molecules listed in Table 1 are also suitable for use in the invention.
  • Another aspect of the invention includes methods for treating a patient already having cancer patient with MLL/KMT2A amplification, the method comprising administration of an effective amount of the agents described above.
  • Yet another aspect of the invention includes methods for reducing TCF3 or AFF3 amplifications/rearrangements in a patient in need thereof, the method comprising a administration of an effective amount of one or more agents including i) a first agent which increases KDM3B expression and function and, or ii) a second agent which inhibits expression or activity of H3K9 methyltransferase, wherein the agents are administered in one or more pharmaceutically acceptable carriers and reducing MLL/KMT2A amplification/rearrangements in said patient.
  • the first agent is a KDM3B agonist
  • the second agent is a G9a inhibitor and both agents are administered.
  • the MLL/KMT2A amplification can be caused by administration of a chemotherapeutic agent such as a chemotherapeutic agent that reduces KDM3B expression.
  • a chemotherapeutic agent such as a chemotherapeutic agent that reduces KDM3B expression.
  • the chemotherapeutic agent is a topoisomerase inhibitor, such as a topoisomerase II inhibitor.
  • the topoisomerase II inhibitors include without limitation doxorubicin, daunorubin, and etoposide.
  • the patient has a genetic loss of KDM3B.
  • the method further comprises administration of a chemotherapeutic agent.
  • the administration of agents which reduce MLL/KMT2A amplification can be performed before, during or after administration of said chemotherapeutic agent.
  • the patient has reduced MLL/KMT2A copy numbers after treatment when compared to an untreated control.
  • the patient is treated with a third agent, i.e., a proteosome inhibitor selected from bortezomib, carfdzomib, and ixazomib.
  • a third agent i.e., a proteosome inhibitor selected from bortezomib, carfdzomib, and ixazomib.
  • UNC 0642 an inhibitor of G9a, an siRNA that targets H3K9 methyltransferase, and/or an siRNA that targets the molecules listed in Table 1 are also suitable for use in the invention.
  • FIG. 1A TCGA Acute Myeloid Leukemia (TCGA LAML) samples with >50% KDM3B loss. Plot shows most LAML samples with KDM3B loss also have KMT2A copy gain with a p-value of 6.45e-07. Statistical significance was computed by Wilcoxon rank-sum test, which provides a non-parametric hypothesis test on two independent samples.
  • Fig. IB Representative examples with the 5qDel FISH probe (5q probe covers the KDM3B locus) demonstrate that KGla (upper) and HL60 (lower) are LOH for KDM3B (red) (left panels).
  • FIG. 1C A schematic of DNA Fluorescent In Situ Hybridization (FISH) probe genomic locations that are used for KMT2A locus.
  • Fig. ID Representative siCTRL (upper) and siKDM3B (lower) DNA FISH images with the KMT2A-1 probe (orange probe location in schematic C and centromere 11 (11C ⁇ control probe)) in RPE cells.
  • KDM3 family siRNA screen demonstrates that only KDM3B depletion generates KMT2A copy gains (KMT2A-1; orange) but not copy gains of centromere 11 (11C,' grey) in RPE cells.
  • Fig. IF Representative example images with the clinical KMT2A DNA FISH break apart probe (red and green probes in schematic C) that show no copy gain in siCTRL (top panel), DNA copy gains (middle 3 panels) and break apart events (bottom panel) upon KDM3B siRNA depletion in RPE cells. Arrowheads highlight the FISH signal.
  • Fig. 1G Quantification of DNA FISH experiments showing KDM3B siRNA knockdown results in KMT2A copy gains (black) and break aparts (purple).
  • Fig. IL IGV tracks of the region containing KMT2A for H3K9mel-3 ChlP-seq. KDM3B Depletion increased H3K9mel and H3K9me2 at the KMT2A locus. Publicly available ChlP-seq shows KDM3B binding within the BCR in HCT-116 cells which is lost upon shKDM3 treatment (49).
  • Fig. IM Bar graphs of ChlP-seq data demonstrating increased fold enrichments of H3K9mel/2 within the KMT2A locus. Error bars represent the SEM.
  • FISH in Fig. IE two independent Control and KDM3B siRNAs were used and a minimum of 100 nuclei were scored per siRNA.
  • FISH in Figs 1G-1H, 1J- 1K a minimum of 200 nuclei were scored for each independent siRNA per experiment. BA- Break aparts.
  • FIG. 2A KDM3B chemical inhibition (KDM3i) promotes transient KMT2A copy gains and break aparts.
  • Fig. 2A In vitro KDM assay with commercial full length purified KDM3B protein incubated with histones and western blotted for KDM3B, H3K9mel and H3K9me3. Blot demonstrates reduction in H3K9mel levels upon incubation with KDM3B is rescued with KDM3i addition.
  • FIG. 2B Cell growth assay for KDM3i treated RPE cells. Treatment with KDM3i at 25nM does not impact cell growth, whereas 250nM and IpM modestly but significantly suppresses growth.
  • FIG. 2C Western blot demonstrating KDM3B protein levels are not reduced with KDM3i treatment in RPE cells in Fig. 2D.
  • Fig. 2E Cell growth assay for KDM3i treated RPE cells corresponding to Figure 3F. Cell growth is not impacted after 12hrs KDM3i treatment, or after 12hr KDM3i washout. Fig.
  • FIG. 2F Schematic (top) and quantification of DNA FISH (bottom) demonstrating that KMT2A amplification and break apart events occur with KDM3B inhibitor treatment at 25nM and IpM for 72 hours but no change in copy number at the adjacent CD3 locus in RPE cells.
  • Fig. 2G Quantification of DNA FISH showing that KDM3B inhibition at 1 pM for 72 hours results in KMT2A copy gains and break aparts in KGla cells with no change in copy number at the CD 3 locus.
  • Fig. 2H Quantification of DNA FISH showing that KDM3B inhibition at 1 pM for 72 hours results in KMT2A copy gains and break aparts in primary AML organoids with no change in copy number at the CD3 locus.
  • FIG. 3A KDM3B suppression leads to integration and inheritance of KMT2A copy gains and break aparts.
  • Fig. 3A KDM3i treatment schematic and associated passaging of RPE cells. Cells were initially treated for 72hrs with 25nM of KDM3i. Cells were then passaged in media without KDM3i every 3 days for sequential passages.
  • Fig. 3B Quantification of KMT2A and CD 3 DNA FISH at passage 0 and passage 10 after KDM3i treatment, which demonstrates KMT2A copy gains and break aparts are inherited in RPE cells after 10 passages (PIO). No significant change occurred with the CD3 probe.
  • Fig. 3A KDM3i treatment schematic and associated passaging of RPE cells. Cells were initially treated for 72hrs with 25nM of KDM3i. Cells were then passaged in media without KDM3i every 3 days for sequential passages.
  • Fig. 3B Quantification of KMT2A and CD 3 DNA FISH at passage 0 and passage 10 after KDM3i treatment,
  • Example metaphase spreads for KMT2A FISH are shown for vehicle and KDM3i treated cells at passage 10. Arrowheads highlight the FISH signal.
  • Fig. 3D Quantification of the metaphase spreads with KMT2A FISH in KDM3i treated and passage 10 cells demonstrating increased copies o£KMT2A are retained.
  • Fig. 3E A KDM3B siRNA depletion schematic and associated passaging of RPE cells (left). Western blots for KDM3B at cell passages used for DNA FISH are shown, which demonstrates KDM3B protein levels return to baseline by passage 3 (P3; right).
  • FIG. 3F Quantification of KMT2A and CD 3 FISH of KDM3B siRNA passaged cells, which demonstrates inheritance at passage 3, 5 and 15. No significant change occurred with the CD3 probe at any passage.
  • Fig. 3G Example metaphase spreads for KMT2A FISH are shown for siCTRL and siKDM3B cells at passage 3. Arrowheads highlight the FISH signal.
  • Fig. 3H Quantification of the metaphase spreads with KMT2A FISH in cells treated with siCTRL and siKDM3B from two independently propagated siCTRL and siKDM3B cells at passages 3 and 9 demonstrating increased copies of KMT2A are retained.
  • Subtell9 copy gain does not change in KDM3B siRNA depleted cells.
  • Fig. 3L DNA FISH for AFF3/LAF4 after passage KDM3B siRNA depleted RPE cells.
  • AFF3 copy gains occur at PO, but are not retained after 3 passages in KDM3B siRNA depleted cells.
  • NMYC copy gain does not change.
  • Error bars represent the SEM. Asterisk indicates significant difference from indicated (p ⁇ 0.05) by two-tailed Student’s t-test. Scale bar represents 5pm. A minimum of 2 replicates per experiment were conducted. For FISH, a minimum of 200 nuclei were scored per replicate per experiment. BA- Break aparts.
  • FIG. 4A A schematic (upper) and DNA FISH quantification (lower) for co-depletion of KDM3B with EHMT1 or EHMT2/G9a. siRNA depleted G9a but not EHMT1 prevents KMT2A copy gains and break aparts upon KDM3B siRNA depletion. No significant change occurred with the CD3 probe.
  • Fig. 4B A schematic (upper) and DNA FISH quantification (lower) for KDM3i and EHMTi treatment. EHMT1/2 chemical inhibition prevents KMT2A copy gains and break aparts upon KDM3i treatment. No significant change occurred with the CD3 probe.
  • Fig. 4A A schematic (upper) and DNA FISH quantification (lower) for KDM3i and EHMTi treatment. EHMT1/2 chemical inhibition prevents KMT2A copy gains and break aparts upon KDM3i treatment. No significant change occurred with the CD3 probe.
  • Fig. 4A A schematic (upper) and DNA FISH quantification (lower) for KDM
  • FIG. 4C A schematic (upper) and DNA FISH quantification (lower) that shows G9a overexpression promotes KMT2A copy gains and break aparts. Halo-EV- Halo empty vector. No significant change occurred with the 11C probe.
  • Fig. 4DA schematic (upper) and DNA FISH quantification (lower) for depletion of G9a in HL60 cells (KDM3B LOH cell line). G9a depletion modestly but significantly suppresses KMT2A copy gains in HL60 cells. No significant change occurred with the CD3 probe.
  • Fig. 4E A schematic (upper) and DNA FISH quantification (lower) for depletion of G9a in RPE-WT or RPE-inherited KMT2A cells.
  • Fig. 4F Genomic tracks in the vicinity of KMT2A gene for input- normalized ChlP-seq densities of H3K9me 1/2/3 upon siKDM3B or siG9a alone or in combination in RPE cells.
  • Fig. 4G Bar graphs representing H3K9 methylation ChlP-seq fold enrichment over input in three parts of KMT2A gene shown in (4F).
  • Fig. 4H A model depicting interplay between KDM3B-G9a regulating H3K9mel/2 and modulating KMT2A amplifications/rearrangements.
  • FIGS. 5A - 5L Reduced CTCF occupancy leads to KMT2A copy gains and break aparts.
  • Fig. 5A Genomic tracks of publicly available ENCODE input-normalized ChlP-seq densities of CTCF in multiple ENCODE cell lines or tissues at the KMT2A locus. CTCF binding at exon 11 of KMT2A is conserved in multiple cell lines and directly overlaps with KDM3B binding in HCT116 cells (Li etal., 2017).
  • Fig. 5B DNA FISH quantification demonstrating single and cosiRNA depletion of KDM3B and CTCF promotes KMT2A copy gains and break aparts. No significant change occurred with the CD3 probe.
  • Fig. 5D Genomic tracks of publicly available input- normalized ChlP-seq densities ofKDM3B (Li et al., 2017) in control and shKDM3 cells. KDM3B binds at exon 11 and is lost upon shKDM3 (green tracks). Lower tracks: input- normalized ChlP-seq densities of CTCF showing that siKDM3B reduced CTCF binding at exon 11 in RPE cells (lower tracks). Fig.
  • FIG. 5E ChlP-qPCR demonstrating suppression of CTCF occupancy at KMT2A exon 11 ⁇ KMT2A ex 77; black) or a negative control for CTCF binding ⁇ CTCF negative site)' is shown in yellow following KDM3B siRNA depletion.
  • Fig. 5G A total of 17,077 CTCF sites out of 46,340 (36.9%) had reduced occupancy with KDM3B depletion.
  • Fig. 5J DNA FISH quantification demonstrating siRNA depletion of G9a prevents KMT2A copy gains and break aparts upon CTCF siRNA depletion. No significant change occurred with the CD 3 probe.
  • Fig. 5K DNA FISH quantification demonstrating EHMT1/2 chemical inhibition prevents KMT2A copy gains and break aparts upon CTCF siRNA depletion. No significant change occurred with the CD3 probe.
  • Fig. 5L A model depicting interplay between KDM3B-G9a-CTCF upon H3K9mel/2 modulation.
  • Fig. 6B A schematic of human KMT2A and adjacent CD3 DNA FISH probes (top). Graph of the DNA FISH quantification for Primary Hematopoietic Stem and Progenitor Cells (HSPC) treated with Dox for 72hrs (bottom). Dox treatment causes significant copy gains and break aparts at the KMT2A locus; while the control region (CD3) changes were not significant.
  • Fig. 6C A schematic of DNA FISH probe genomic locations that are used for mouse Kmt2a/Con9 locus (top).
  • Fig. 6D RT-qPCR analysis demonstrating that Dox significantly reduced KDM3B transcript levels relative to B-actin after 72 hours of exposure in RPE cells.
  • Fig. 6E Representative western blot illustrating Dox reducing KDM3B protein levels after 72 hours of exposure in RPE cells.
  • FIG. 6G RT-qPCR analysis demonstrating that Dox significantly reduced CTCF transcript levels relative to B-actin after 72 hours of exposure in RPE cells.
  • FIG. 6H Representative western blot illustrating Dox reducing CTCF protein levels after 72 hours of exposure in RPE cells.
  • Fig. 6J Graph of the quantification of western blots in panel S6F showing a significant reduction in KDM3B (black) and CTCF (blue) protein levels following Dox treatment in KGla cells.
  • FIG. 6K Western blot illustrating etoposide dose-dependently reducing KDM3B (upper) and CTCF (lower) protein levels after 72 hours of exposure in RPE cells.
  • Fig. 6L Western blot illustrating MG132 partially rescuing KDM3B protein levels in the presence of Dox treatment in RPE cells.
  • Fig. 6M Western blot illustrating MG132 rescuing CTCF protein levels in the presence of Dox treatment. Protein levels were quantified using ImageJ and CTCF was normalized to a-Actinin. Error bars represent the SEM. Asterisk indicates significant difference from indicated (p ⁇ 0.05) by two-tailed Student’s t-test. A minimum of 2 replicates per experiment were conducted. For FISH, a minimum of 200 nuclei were scored per replicate per experiment. BA- Break aparts. NS- not significant to control.
  • FIG. 7A ChlP-qPCR demonstrating increase of H3K9mel at KMT2A exon 11 (KMT2A CTCF site) following KDM3B siRNA depletion (left; upper) and Dox treatment at Ipg/pL for 24hr (right; upper).
  • Fig. 7A ChlP-qPCR demonstrating increase of H3K9mel at KMT2A exon 11 (KMT2A CTCF site) following KDM3B siRNA depletion (left; upper) and Dox treatment at Ipg/pL for 24hr (right; upper).
  • FIG. 7B A treatment schematic (upper) and DNA FISH quantification (lower) demonstrating that Dox treatment causes KMT2A amplification and rearrangements. KMT2A amplifications generated by Dox treatment are significantly rescued with CTCF overexpression by DNA FISH.
  • Fig. 7C A treatment schematic (upper) and DNA FISH quantification (lower) demonstrating that Dox treatment causes KMT2A amplification and rearrangement. KMT2A amplifications and rearrangements generated by Dox treatment that are significantly rescued with G9a depletion by DNA FISH.
  • FIG. 7D A treatment schematic (upper) and DNA FISH quantification (lower) demonstrating that Dox treatment causes KMT2A amplification and rearrangements that are significantly rescued with EHMT1/2 inhibition by DNA FISH.
  • Fig. 7E A treatment schematic (upper) and DNA FISH (lower) demonstrating that Dox treatment causes KMT2A amplification and rearrangements that are significantly rescued with KDM3B overexpression by DNA FISH.
  • Fig. 7F A first model summarizing the data described herein. The model illustrates that KDM3B and CTCF are suppressed with Dox treatment, leading to increased H3K9 mono- and dimethylation and reducing CTCF occupancy, which in turn promotes KMT2A amplification and rearrangements (BA).
  • FIG. 7G A second schematic depicting the regulation of KMT2A DNA copy gains and rearrangements.
  • KDM3B and/or CTCF control KMT2A copy gains and break aparts, while G9a promotes these events (I; Figures 1, 2, 4, and 5).
  • KDM3B and/or CTCF control KMT2A copy gains and break aparts, while G9a promotes these events (I; Figures 1, 2, 4, and 5).
  • KMT2A extrachromosomal DNA copy gains and break aparts recover and return to baseline (II; Figure 2K).
  • KMT2A copy gains and rearrangement events become inherited in the genome (Bottom; Figure 3).
  • KDM3B e.g., KDM3B LOH
  • KDM3B LOH transient extrachromosomal and inherited KMT2A amplifications
  • Figure 8 A table showing the different domains and regions of the KDM3B amino acid sequence that provide therapeutic targets for the treatment of MLL.
  • Peptide mimetics can be designed which mimic the enzymatic regions and/or the intrinsic disordered domains of the KDM3B protein.
  • Another region of interest is the LXLL motif mimics and along with the regions containing the mutations in KDM3B correlated with cancer and de novo in individuals.
  • KDM3B mimics or agonists are used with G9a inhibitor(s) as described herein below.
  • AML and myelodysplasia are characterized by genomic amplifications and translocations of the 1 lq23 region, which includes MLL/KMT2A and other target genes (Cox et al., 2003; Dolan et al., 2002; Maitta et al., 2009; Poppe et al., 2004; Schichman et al., 1994; Sperling et al. , 2017; Tang et rz/., 2015; Walter et al., 2009). MLL/KMT2A rearrangements are a hallmark of greater than 70 percent of infant leukemias (Rice and Roy, 2020; Woo et al., 2014).
  • MLL translocations are also found in adult primary and therapy -related leukemias (Andersen et al., 2001; Dulak etal., 2012; Meyer et al., 2023; Pedersen-Bj ergaard et al., 1998; Super et al., 1993; Winters and Bernt, 2017).
  • KMT2A rearrangement partners More than 100 known KMT2A rearrangement partners have been documented (Meyer et al., 2023)
  • KMT2A rearrangements result in the fusion of the gene to any of the partner genes, leading to protein chimeras.
  • KMT2A can also rearrange to several noncoding regions throughout the genome (Meyer etal., 2023).
  • topo II inhibitors have been shown to promote non-leukemia and leukemia-associated KMT2A rearrangements in hematopoietic stem cells and a host of other non-hematologic cell types (Gomez-Herreros etal., 2017; Gothe et ⁇ l, 2019; Libura et aL, 2005).
  • the H3K9mel/2 lysine demethylase KDM3B originally named 5qNCA, resides in the frequently deleted region of 5q31 associated with loss of heterozygosity (LOH) (Ebert, 2010; Hu etal., 2001; Yoo et al., 2020). MLL/KMT2A copy gains often occur with 5q LOH (Herry et al., 2006; Schoch et al., 2005).
  • KDM3B has been implicated as a myeloid leukemia tumor suppressor through oncogene regulation and is shown to contribute to genome stability; however, a full appreciation for the role KDM3B plays in genome regulation is understudied (Hu et al., 2001; Kim et al., 2012; MacKinnon etal., 2011; Saavedra et al., 2020; Xu etal., 2018).
  • KDM3B-G9a balance controls CTCF occupancy in the region enriched in H3K9 methylation, and in turn, the ability of the KMT2A/MLL locus to undergo site-specific copy gains and genomic rearrangement.
  • Dox chemotherapeutic agent
  • the chemotherapeutic agent Doxorubicin (Dox) reduces KDM3B and CTCF protein levels, and as a consequence, promotes KMT2A copy gains and rearrangements.
  • Dox chemotherapeutic agent
  • KDM3B rescued the Dox-induced KMT2A changes.
  • knockdown or chemical inhibition of G9a rescued the MLL/KMT2A amplification and break apart events in Dox-treated cells.
  • a or “an” entity refers to one or more of that entity; for example, "a cDNA” refers to one or more cDNA or at least one cDNA.
  • a cDNA refers to one or more cDNA or at least one cDNA.
  • the terms “a” or “an,” “one or more” and “at least one” can be used interchangeably herein.
  • the terms “comprising,” “including,” and “having” can be used interchangeably.
  • a compound “selected from the group consisting of' refers to one or more of the compounds in the list that follows, including mixtures (i.e. combinations) of two or more of the compounds.
  • phrases "consisting essentially of' when referring to a particular nucleotide or amino acid means a sequence having the properties of a given SEQ ID NO.
  • the phrase when used in reference to an amino acid sequence, the phrase includes the sequence per se and molecular modifications that would not affect the functional and novel characteristics of the sequence.
  • a “derivative" of a polypeptide, polynucleotide or fragments thereof means a sequence modified by varying the sequence of the construct, e.g. by manipulation of the nucleic acid encoding the protein or by altering the protein itself.
  • “Derivatives" of a gene or nucleotide sequence refers to any isolated nucleic acid molecule that contains significant sequence similarity to the gene or nucleotide sequence or a part thereof.
  • “derivatives” include such isolated nucleic acids containing modified nucleotides or mimetics of naturally-occurring nucleotides.
  • nucleic acid refers to any DNA or RNA molecule, either single or double stranded and, if single stranded, the molecule of its complementary sequence in either linear or circular form.
  • nucleic acid molecules a sequence or structure of a particular nucleic acid molecule may be described herein according to the normal convention of providing the sequence in the 5' to 3' direction. With reference to nucleic acids of the invention, the term “isolated nucleic acid” is sometimes used.
  • an "isolated nucleic acid” may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a prokaryotic or eukaryotic cell or host organism.
  • a vector such as a plasmid or virus vector
  • this term may refer to a DNA that has been sufficiently separated from (e.g., substantially free of) other cellular components with which it would naturally be associated.
  • isolated nucleic acid refers primarily to an RNA molecule encoded by an isolated DNA molecule as defined above. Alternatively, the term may refer to an RNA molecule that has been sufficiently separated from other nucleic acids with which it would be associated in its natural state (i.e., in cells or tissues). An isolated nucleic acid (either DNA or RNA) may further represent a molecule produced directly by biological or synthetic means and separated from other components present during its production.
  • an isolated or biologically pure molecule or cell is a compound that has been removed from its natural milieu.
  • isolated and biologically pure do not necessarily reflect the extent to which the compound has been purified.
  • An isolated compound of the present invention can be obtained from its natural source, can be produced using laboratory synthetic techniques or can be produced by any such chemical synthetic route.
  • the method of treatment effectively suppresses symptoms associated with cancer. Symptoms of vary according to the location and type of cancer being treated. In certain embodiments, symptoms of cancer include, fatigue, weight loss, lumps, swelling, pain, coughing, wheezing, new or unusual growth, discoloration, and no symptoms at all. In certain embodiments, the treatment reduces the risk of relapse.
  • treatment or inhibition may be assessed by inhibition of disease progression, inhibition of tumor growth, reduction of primary tumor, relief of tumor-related symptoms, inhibition of tumor secreted factors, delayed appearance of primary or secondary tumors, slowed development of primary or secondary tumors, decreased occurrence of primary or secondary tumors, slowed or decreased severity of secondary effects of disease, arrested tumor growth and regression of tumors, increased Time To Progression (TTP), increased Progression Free Survival (PFS), increased Overall Survival (OS), among others.
  • OS as used herein means the time from treatment onset until death from any cause.
  • TTP as used herein means the time from treatment onset until tumor progression; TTP does not include deaths.
  • Time to Remission means the time from treatment onset until remisison, for example, complete or partial remission.
  • PFS means the time from treatment onset until tumor progression or death.
  • PFS rates will be computed using the Kaplan-Meier estimates.
  • Event-free survival means the time from study entry until any treatment failure, including disease progression, treatment discontinuation for any reason, or death.
  • Relapse-free survival means the length of time after the treatment ends that the patient survives without any signs or symptoms of that cancer.
  • ORR Overall response rate
  • ORR means the sum of the percentage of patients who achieve complete and partial responses.
  • Complete remission rate refers to the percentage of patients achieving complete remission (CR).
  • Duration of response is the time from achieving a response until relapse or disease progression.
  • Duration of remission is the time from achieving remission, for example, complete or partial remission, until relapse.
  • complete inhibition is referred to herein as prevention or chemoprevention.
  • prevention includes either preventing the onset of clinically evident cancer altogether or preventing the onset of a preclinically evident stage of a cancer.
  • chromosomal translocation is defined as a genome abnormality in which a chromosome breaks and either the whole or a portion of it reattaches to a different chromosome. Depending on the location of the breaks, translocations may lead to the formation of fusion genes, or may disrupt a gene or its regulatory sequences, and in this way cause gene misregulation.
  • MLL1-4 or KMT2A-D are histone methyltransferases that methylate lysine 4 on the histone H3 tail and regulate crucial functions of the genome by modulating the chromatin structure and DNA accessibility.
  • G9a/EHMT-2 is a histone methyltransferase that specifically mono- and dimethylates 'Lys-9' of histone H3 (H3K9mel and H3K9me2, respectively) in euchromatin.
  • H3K9me represents a specific tag for epigenetic transcriptional repression by recruiting HP1 proteins to methylated histones. Also mediates monomethylation of 'Lys-56' of histone H3 (H3K56mel) in G1 phase, thereby promoting interaction between histone H3 and PCNA and regulating DNA replication. Also weakly methylates 'Lys-27' of histone H3 (H3K27me).
  • CTCF is an insulator protein, that along with cohesin, controls domain location by folding domains into loop structures.
  • CTCF and cohesin co-occupy the same sites and physically interact as a complex.
  • the cohesin complex is a multi-subunit ring-like structure composed of SMC1A, SMC3, RAD21, and STAG! or STAG2 proteins and functions in the processes of anaphase sister chromatid exchange (mitotic checkpoint), DNA repair regulation, and transcription control.
  • Agents which increase binding functions of CTCF and or RAD21 can be included with the other MLL/DKMTA amplification reducing agents described herein. Studies show that both CTCF and RAD21 are mutated in a variety of cancers and other diseases See for example, Antony et al.
  • CTCF a ssociated with disease include, for example, in order of occurrence, p.R377, p.R448, p.R457, p.H284, and p.S354.
  • Zinc finger I amino acids 260-288) and zinc finger 2, (amino acids 294-316) are also frequently mutated.
  • Peptide mimics of these regions which stabilize or restore mutated zinc finger sequences in CTCF are also within the scope of the invention.
  • a protein mimetic is a molecule such as a peptide, a modified peptide or any other molecule that biologically mimics the action or activity of some other protein.
  • Protein mimetics are commonly used in drug design and discovery. Types of mimetics include without limitation, Antibody mimetics, e .g., molecules that mimic antigen binding activity of antibodies; peptidomimetics - small protein-like chains designed to mimic larger peptide and phosphomimetics - An amino acid substitution or modification which mimic the effect of protein phosphorylation.
  • the design and generation of molecules capable of mimicking the binding and/or functional sites of proteins are used to advantage for the exploration and modulation of protein function through controlled interference with the underlying molecular interactions.
  • Synthetic peptides are effective mimics of native protein sites because such peptides can be generated as exact copies of protein fragments and can also comprise diverse chemical modifications, which includes the incorporation of a large range of non-proteinogenic amino acids as well as the modification of the peptide backbone. Apart from extending the chemical and structural diversity presented by peptides, such modifications also increase the proteolytic stability of the molecules, enhancing their utility for biological applications. Peptide mimetics of KDM3B and CTCF could provide therapeutic benefit to subjects having or being a risk for MLL.
  • break-apart or translocation probes refer to probes which target two areas of a specific gene sequence. Usually, a green fluorescent label is used on one end of a gene sequence and a red fluorescent label is used on the other end of the gene sequence. When the gene sequences are intact (still close together), the green and red signals will usually fluoresce as a yellow signal, known as a fusion signal. The width of the green and red signals are determined. If the green and red signals are closer than the width of one signal, they are said to be intact. When a break in the gene sequence occurs, the green and red signal will not be close together anymore and will thus appear as separate green and red signals.
  • a “break point” is where a precise area a break occurs. The “break apart” is analyzed by the FISH technique, refers to separate areas on the gene and is typically characterized as a rearrangement.
  • extrachromosomal DNA or “ecDNA” refer to any DNA that is found off the chromosomes, either inside or outside the nucleus. Multiple forms of ecDNA exist and can play an important role in diseases such as cancer. ecDNA has been identified in the nuclei of various cancer cells and has been shown to cary many copies of driver oncogenes. ecDNA is considered to be a primary mechanism of gene amplification, resulting in many copies of driver oncogenes and very aggressive cancers.
  • Proteasome inhibitors induce the accumulation of unfolded and misfolded proteins, leading to apoptosis and cell death through ER stress, reactive oxygen species production, JNK and p53 activation, cyclin-dependent kinase inhibitors, and pro-apoptotic proteins induction.
  • Pls Proteasome inhibitors
  • miRNA refers to about 10-35 nt, preferably about 15-30 nt, and more preferably about 19-26 nt, non-coding RNAs derived from endogenous genes encoded in the genomes of plants and animals. They are processed from longer hairpin-like precursors termed pre-miRNAs that are often hundreds of nucleotides in length. MicroRNAs assemble in complexes termed miRNPs and recognize their targets by antisense complementarity. These highly conserved, endogenously expressed RNAs are believed to regulate the expression of genes by binding to the 3 '-untranslated regions (3'-UTR) of specific mRNAs as well as other regions on targeted mRNAs.
  • 3'-UTR 3 '-untranslated regions
  • a possible mechanism of action assumes that if the microRNAs match 100% their target, i.e. the complementarity is complete, the target mRNA is cleaved, and the miRNA acts like a siRNA. However, if the match is incomplete, i.e. the complementarity is partial, then the translation of the target mRNA is blocked.
  • the manner by which a miRNA base-pairs with its mRNA target correlates with its function: if the complementarity between a mRNA and its target is extensive, the RNA target is cleaved; if the complementarity is partial, the stability of the target mRNA in not affected but its translation is repressed.
  • RNA interference refers generally to a process or system in which a RNA molecule changes the expression of a nucleic acid sequence with which RNA molecule shares substantial or total homology.
  • RNAi agent refers to an RNA sequence that elicits RNAi.
  • siRNA refers to a molecule involved in the RNA interference process for a sequence-specific post-transcriptional gene silencing or gene knockdown by providing small interfering RNAs (siRNAs) that has homology with the sequence of the targeted gene.
  • small interfering RNAs can be synthesized in vitro or generated by ribonuclease III cleavage from longer dsRNA and are the mediators of sequence-specific mRNA degradation.
  • the siRNA of the invention are chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
  • the siRNA can be synthesized as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • Commercial suppliers of synthetic RNA molecules or synthesis reagents include Applied Biosystems (Foster City, Calif., USA), Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, Colo., USA), Pierce Chemical (part of Perbio Science, Rockford, Ill., USA), Glen Research (Sterling, Va., USA), ChemGenes (Ashland, Mass., USA) and Cruachem (Glasgow, UK).
  • a "small nucleic acid inhibitor” refers to any sequence based nucleic acid molecule which, when introduced into a cell expressing the target nucleic acid, is capable of modulating expression of that target.
  • siRNA, antisense, miRNA, shRNA and the like may be utilized in the methods of the invention.
  • delivery refers to the introduction of foreign molecule (i.e., miRNA containing nanoparticle) into cells.
  • administration means the introduction of a foreign molecule into a cell.
  • delivery means the introduction of a foreign molecule into a cell. The term is intended to be synonymous with the term “delivery”.
  • construct means a recombinant nucleic acid, generally recombinant DNA, which has been generated for the purpose of the expression or propagation of a nucleotide sequence(s) of interest or is to be used in the construction of other recombinant nucleotide sequences.
  • promoter or “promoter polynucleotide” is understood to mean a regulatory sequence/ element or control sequence/element that is capable of binding/recruiting an RNA polymerase and initiating transcription of sequence downstream or in a 3’ direction from the promoter.
  • a promoter can be, for example, constitutively active, or always on, or inducible in which the promoter is active or inactive in the presence of an external stimulus.
  • Example of promoters include T7 promoters or U6 promoters.
  • operably linked can mean the positioning of components in a relationship which permits them to function in their intended manner.
  • a promoter can be linked to a polynucleotide sequence to induce transcription of the polynucleotide sequence.
  • complementarity refers to the ability of a nucleic acid to form hydrogen bond(s) with another nucleic acid sequence by either traditional Watson-Crick or other non-traditional types.
  • a percent complementarity indicates the percentage of residues in a nucleic acid molecule which can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 4, 5, and 6 out of 6 being 66.67%, 83.33%, and 100% complementary).
  • Perfectly complementary means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence.
  • substantially complementary refers to a degree of complementarity that is at least 40%, 50%, 60%, 62.5%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 100%, or percentages in between over a region of 4, 5, 6, 7, and 8 nucleotides, or refers to two nucleic acids that hybridize under stringent conditions.
  • the invention provides methods comprising delivering one or more polynucleotides, such as or one or more vectors as described herein (e.g., encoding siRNA, antisense oligonucleotides or other type of inhibitory nucleic acid), to a host cell.
  • the invention further provides cells produced by such methods, and organisms or cells comprising or produced from such cells.
  • Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids in mammalian cells or target tissues. Such methods can be used to administer nucleic acids encoding inhibitory compounds to cells in culture, or in a host organism.
  • Non-viral vector delivery systems include DNA plasmids, RNA (e.g.
  • Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell.
  • Methods of non-viral delivery of nucleic acids include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipidmucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA.
  • Lipofection is described in e.g., U.S. Pat. Nos. 5,049,386, 4,946,787; and 4,897,355) and lipofection reagents are sold commercially (e.g., TransfectamTM and LipofectinTM).
  • Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those of Feigner, WO 91/17424; WO 91/16024. Delivery can be to cells (e.g. in vitro or ex vivo administration) or target tissues (e.g. in vivo administration).
  • lipidmucleic acid complexes including targeted liposomes such as immunolipid complexes
  • crystal Science 270:404-410 (1995); Blaese et al., Cancer Gene Then 2:291-297 (1995); Behr et al., Bioconjugate Chem. 5:382-389 (1994); Remy et al., Bioconjugate Chem. 5:647-654 (1994); Gao et al., Gene Therapy 2:710-722 (1995); Ahmad et al., Cancer Res. 52:4817-4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).
  • RNA or DNA viral based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus.
  • Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro, and the modified cells may optionally be administered to patients (ex vivo).
  • Conventional viral based systems could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.
  • Lentiviral vectors are retroviral vectors that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression.
  • Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian Immuno deficiency virus (SIV), human immuno deficiency virus (HIV), and combinations thereof (see, e.g., Buchscher et al., J. Virol. 66:2731-2739 (1992); Johann et al., J. Virol. 66: 1635-1640 (1992); Sommnerfelt et al., Virol. 176:58-59 (1990); Wilson et al., J. Virol. 63:2374-2378 (1989); Miller et al., J. Virol.
  • MiLV murine leukemia virus
  • GaLV gibbon ape leukemia virus
  • SIV Simian Immuno deficiency virus
  • HAV human immuno deficiency virus
  • adenoviral based systems may be used.
  • Adenoviral based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system.
  • Adeno-associated virus vectors may also be used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acids and peptides, and for in vivo and ex vivo gene therapy procedures (see, e.g., West et al., Virology 160:38-47 (1987); U.S. Pat. No. 4,797,368; WO 93/24641;
  • Packaging cells are typically used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and ⁇
  • Viral vectors used in gene therapy are usually generated by producing a cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences being replaced by an expression cassette for the polynucleotide(s) to be expressed. The missing viral functions are typically supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome which are required for packaging and integration into the host genome.
  • Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences.
  • the cell line may also be infected with adenovirus as a helper.
  • the helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid.
  • the helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV.
  • the phrase "effective amount" of a compound or pharmaceutical composition refers to an amount sufficient to modulate tumor growth or metastasis in an animal, especially a human, including without limitation decreasing tumor growth or size or preventing formation of tumor growth in an animal lacking any tumor formation prior to administration, i.e., prophylactic administration
  • the term "pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers, for example to a diluent, adjuvant, excipient, auxiliary agent or vehicle with which an active agent of the present invention is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water or aqueous saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin.
  • a pharmaceutical composition of the present invention can be administered by any suitable route, for example, by injection, by oral, pulmonary, nasal or other forms of administration.
  • pharmaceutical compositions contemplated to be within the scope of the invention comprise, inter alia, pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers.
  • compositions can include diluents of various buffer content (e.g., Tris HC1, acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc., or into liposomes.
  • buffer content e.g., Tris HC1, acetate, phosphate
  • additives e.g., Tween 80, Polysorbate 80
  • anti oxidants e.g., ascorbic acid, sodium metabisulfite
  • preservatives e.g., Thimersol, benzyl alcohol
  • compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of components of a pharmaceutical composition of the present invention. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, Pa. 18042) pages 1435 1712 which are herein incorporated by reference.
  • a pharmaceutical composition of the present invention can be prepared, for example, in liquid form, or can be in dried powder, such as lyophilized form. Particular methods of administering such compositions are described infra.
  • a pharmaceutical composition of the present invention can be delivered in a controlled release system, such as using an intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used [see Langer, supra; Sefton, CRC Crit. Ref Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)].
  • polymeric materials can be used [see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Press: Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the target tissues of the animal, thus requiring only a fraction of the systemic dose [see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115 138 (1984)].
  • a controlled release device can be introduced into an animal in proximity of the site of inappropriate immune activation or a tumor. Other controlled release systems are discussed in the review by Langer [Science 249:1527 1533 (1990)].
  • biomarker refers to a characteristic that is objectively measured and evaluated as an indicator of normal biologic processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention.
  • the terms “modulate”, “modulating” or “modulation” refer to changing the rate at which a particular process occurs, inhibiting or promoting a particular process, reversing a particular process, and/or preventing the initiation of a particular process. Accordingly, if the particular process is tumor growth or metastasis, the term “modulation” includes, without limitation, decreasing the rate at which tumor growth and/or metastasis occurs; inhibiting tumor growth and/or metastasis; reversing tumor growth and/or metastasis (including tumor shrinkage and/or eradication) and/or preventing tumor growth and/or metastasis.
  • a compound that increases a known activity, e.g., tumor growth or metastasis is an “agonist”.
  • One that decreases, or prevents, an undesirable malignant phenotype is an “antagonist” or “inhibitor”.
  • tumor As used herein, the terms “tumor”, “tumor growth” or “tumor tissue” can be used interchangeably, and refer to an abnormal growth of tissue resulting from uncontrolled progressive multiplication of cells and serving no physiological function.
  • a solid tumor can be malignant, e.g. tending to metastasize and being life threatening, or benign.
  • treatment induced cancer refers to a new cancer or tumor in a patient with a preexisting cancer that is developed in response to treatment of the preexisting cancer.
  • “Chemo- induced cancer” or “chemotherapy induced cancer” refers to a treatment induced cancer that was developed in response to chemotherapy treatment.
  • chromosomal instability refers to a higher than normal rate of mis-segregation of chromosomes or parts of chromosomes during mitosis due to defective cell cycle quality control mechanisms, resulting in copy number alterations (CNAs) or aneuploidy.
  • gene amplification or “copy number amplification” or “DNA copy gain” refers to an increase in the number of copies of a gene sequence. In certain embodiments, these phrases refer to any number of copies greater than diploid. There may also be an increase in the RNA and protein made from that gene. Gene amplification is common in cancer cells, and some amplified genes may cause cancer cells to grow or become resistant to anticancer drugs. Gene amplification of oncogenes on ecDNA is a frequent event in cancer.
  • Epigenetic state or Epigenetic phenomena means changes produced in gene expression or other DNA-dependent processes caused by mechanisms other than changes in the underlying DNA sequence. For example, methylation of cytosines (Cs) or histone modifications can affect expression of a gene. These molecular modifications of the DNA are often called “epigenetic marks.” For example, increased or decreased methylation of Cs in a genome are part of normal biology but can also be associated with disease. In a similar fashion, post translational modifications (PTMs) occur on histones and impact DNA-dependent processes.
  • epigenetic state refers to a gene or region in a genome that reflects particular epigenetic phenomena.
  • a gene in a particular disease cohort, can be found that causes disease through multiple mechanisms, including, but not limited to, impairment of protein function by a SNV, deletion of the gene via a CNV, little or no expression of the gene due to a change in the epigenetic state of the gene itself and/or regulatory region(s) in the genome controlling expression of the gene.
  • an “inhibitor” (interchangeably termed “antagonist”) of a polypeptide of interest is an agent that interferes with activation or function of the polypeptide of interest, e.g., partially or fully blocks, inhibits, or neutralizes a biological activity mediated by a polypeptide of interest.
  • an antagonist of G9a may refers to any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity mediated by G9a.
  • Examples of inhibitors include antibodies; ligand antibodies; small molecule antagonists; antisense and inhibitory RNA (e.g., siRNA) molecules. siRNAs described above “modified nucleotides”.
  • nucleotides comprising non- naturally occurring moieties that confer increased nuclease resistance or thermodynamic stability during hybridization as compared with a polynucleotide or polyribonucleotide that differs from the inhibitory nucleic acid only by having a natural nucleotide in place of the modified nucleotide.
  • the ribose moiety of a nucleotide is modified with an extra bridge connecting the 2' oxygen and 4' carbon. Numerous chemical modifications are commonly used for the synthesis of oligonucleotides for a variety of reasons.
  • Modified sugar moieties are also being incorporated into therapeutic oligonucleotides. Changing the sugar moiety generally increases nuclease resistance and binding affinity to a complementary target.
  • BNA “Bridged nucleic acid” refers to 2'-O,4'-C-methylene-modified nucleic acids.
  • BNA where the 2' oxygen and 4' carbon are bridged by a methylene group are used.
  • 2'-O,4'-C-ethylene-bridged nucleic acids ENA
  • BNA can include, but are not limited to, 2',4'-BNA NC [NH], 2',4'-BNA NC [NMe], and 2',4'-BNA NC [NBn], (s)-cEt (S- constrained Ethyl).
  • tcDNA (tricycloDNA) modifications can also be used to constrain nucleotides.
  • LNA nucleotide refers to a modified RNA nucleotide that provides the polynucleotide with greater thermodynamic stability during hybridization as compared with a polynucleotide that differs from the LNA only by having a natural ribonucleotide in place of the modified RNA nucleotide.
  • the ribose moiety of a modified RNA nucleotide is modified with an extra bridge connecting the 2' oxygen and 4' carbon.
  • LNA nucleotides can comprise any type of extra bridge between the 2'-0 and 4'-C of the RNA that increases the thermodynamic stability of the duplex between the LNA and its complement.
  • Oligonucleotide backbone configurations that demonstrate particularly high binding affinities to the target (measured by melting temperature or Tm) are preferred for implementing the steric hindrance mechanism.
  • BNA, LNA, FANA, 2'-fluoro, 2’-O-methoxyethyl (2’ -MOE), 2’-NH2, 2’-F-RNA, morpholino and piperazine containing backbones are particularly well suited for this purpose.
  • oligonucleotide ribose include, are not limited to, FHNA (Fluoro Hexitol Nucleic Acid), (s)-5’-C-methyl, UNA (Unlocked Nucleic Acid), 4’-thio-RNA, cyclohexene nucleic acid. Modified backbone linkages are sometimes used instead of phosphodi ester linkage to minimize oligonucleotide degradation by nucleases.
  • Some examples include, are not limited to, phosphorothioate, boranophosphonate, phosphoramidate, methyl phosphonate, (SC5’ Rp)-a,p- CNA (Dioxaphosphorinane-Constrained Nucleic Acid), PNA (Peptide Nucleic Acid), PMO (Phosphorodiamidate Morpholino Oligonucleotide), phosphoryl guanidine.
  • 5’ modifications to increase phosphate stability include, are not limited to, E-VP ((E)-VinylPhosphonate), 5’ methyl phosphonate, 5 ’-phosphorothioate, (s)-5 ’ -methyl with phosphate, 5’-methoxy.
  • 3’ modifications to increase phosphate stability include, are not limited to, 2-hydroxy ethylphosphate AND, 3’-ddc (dideoxyCytosine), 3 ’-amino.
  • Base modifications to improve 3’ stability include, are not limited to, 2’-thio-dT.
  • oligonucleotides with mixed linkages such as boranophosphate and phosphate linkages has been accomplished by several solid phase methods including one involving the use of bis(trimethylsiloxy)cyclododecyloxysilyl as the 5'-0-protecting group (Brummel and Caruthers, Tetrahedron Lett 43: 749, 2002).
  • bis(trimethylsiloxy)cyclododecyloxysilyl as the 5'-0-protecting group
  • the 5'-hydroxyl is initially protected with a benzhydroxybis-(trimethylsilyloxy)silyl group and then deblocked by Et3N:HF before the next cycle (McCuen et al., J Am Chem Soc 128: 8138, 2006).
  • This method can result in a 99% coupling yield and can be applied to the synthesis of oligos with pure boranophosphate linkages or boranophosphate mixed with phosphodiester, phosphorothioate, phosphorodithioate or methyl phosphonate linkages.
  • the boranophosphorylating reagent 2-(4-nitrophenyl)ethyl ester of boranophosphoramidate can be used to produce boranophosphate linked oligoribonucleotides
  • This reagent readily reacts with a hydroxyl group on the nucleosides in the presence of 1H- tetrazole as a catalyst.
  • the 2-(4-nitrophenyl)ethyl group can be removed by 1,4- diazabicyclo[5.4.0]undec-7-ene (DBU) through beta-elimination, producing the corresponding nucleoside boranomonophosphates (NMPB) in good yield.
  • DBU 1,4- diazabicyclo[5.4.0]undec-7-ene
  • Nucleobase modifications to increase binding affinity include, are not limited to, 5’- methylcytidine, 5-methyluridine (ribothymidine), and abasic RNA.
  • MLL inhibitor refers to any compound naturally occurring or synthesized, having the ability of inhibiting MLL/KMT2A amplification.
  • Agents that reduce MLL/KMT2A break aparts, and amplifications include, without limitation, a G9a inhibitor or KDM3B agonist.
  • G9a inhibitor refers to any compound natural occurring or synthesized, having the ability of inhibiting G9a activity, stability or expression.
  • KDM3B agonist refers to any compound natural occurring or synthesized, having the ability of promoting KDM3B expression or overexpression.
  • a KDM3B agonist is for example any molecule that can increase activity or stabilize the protein such as an RNA aptamer.
  • drug response means any biological response in an organism that is the result of exposure to the drug.
  • Drug responses can be favorable, such as when a patient's disease is eradicated by treatment with the drug, or unfavorable, such as when a patient enters a coma upon treatment with a drug.
  • Retinal pigment epithelial (RPE) cells were cultured in DMEM-high glucose (Sigma) media supplemented with 10% heat-inactivated fetal bovine serum (FBS), lOOU/ml penicillin, lOOpg/ml streptomycin, and 2mM L-glutamine.
  • FBS heat-inactivated fetal bovine serum
  • U937 cells were cultured in RPMI 1640 media supplemented with 10% heat-inactivated FBS, lOOU/ml penicillin, lOOpg/ml streptomycin and 2mM L-glutamine.
  • HL60 and KGla cells were cultured in RPMI 1640 media supplemented with 20% heat-inactivated FBS, lOOU/ml penicillin, lOOpg/ml streptomycin and 2mM L-glutamine.
  • Cell line identities were authenticated by short tandem repeat analysis and Mycoplasma tested using the MycoAlert Detection Kit (Lonza, LT07-218).
  • Human primary patient-derived AML cells were obtained and generated as described previously (Duy et al., 2019). Primary patient-derived AML cells were maintained on 30 Gy- irradiated OP9 feeder layer cells in complete in Iscove’s modified Dulbecco’s medium (IMDM; Thermo Fisher Scientific, Waltham, MA) containing 20% fetal bovine serum (Corning Premium FBS), 100 TU/ml penicillin, 100 pg/ml streptomycin, 50 pM 2-mercaptoethanol and supplemented with AML-maintaining cytokines (50 ng/ml SCF, 20 ng/ml GM-CSF, 50 ng/ml FLT3 ligand, 20 ng/ml IL-3, 20 ng/ml IL-6, and 20 ng/ml G-CSF). Isolation of HSPCs:
  • HSPCs hematopoietic stem and progenitor cells
  • Tables 3A- H Reagents and Materials For co-transfection experiments, both the siRNAs were transfected at the same time and collected at 72 hr from transfection (FIG. 9B-9C, 10A-10D).
  • Figures 8A and 7A-7F cells were first transfected with siEHTMl/EHMT2 siRNA’s for 24 hr followed by KDM3B siRNA transfection for 48hr.
  • U937 were transfected using Neon System (Invitrogen) following manufacturer’s instructions. Cells were mixed with siRNA constructs in lOpl of supplied buffer. Cell mixture was loaded in Neon syringe and submerged in electrode buffer. For U937 cells, 3 pulses of 1400V with a width of 10ms was applied for 500,000 cells. Cells were immediately transferred into fresh media in 6 well plates.
  • Neon System Invitrogen
  • the membranes were blocked for at least 1 hr in 5% BSA- PBST (IX PBS with 0.5% Tween-20) or 5% milk-PBST and probed over night with specific antibodies as follows at the following dilutions: anti-KDM3B (Cell Signaling) (1: 1000); anti-P- Actin (1 : 10,000); anti-G9A (Sigma) (1: 1000); anti-Actinin (Santacruz) (1: 1000). Catalog numbers for all antibodies used in this study can be found in the tables above.
  • Membranes were washed three times in PBST the next day, incubated with goat antimouse IgG peroxidase conjugated secondary antibody (170-6516, Biorad) or goat anti-rabbit peroxidase conjugated secondary antibody (A00167, GenScript) at 1 :2500 in 5% milk-PBST for at least Cup at room temperature, washed 3 times with PBST and incubated in Lumi-Light western blotting substrate (12015200001, Roche) Imin. Membranes were developed with Lumi- Film Chemiluminiscent detection fdm (11666657001, Roche). The western blot images displayed in the figures have been cropped and auto-contrasted.
  • FISH DNA Fluorescent In situ Hybridization
  • the FISH protocol was performed as described previously in Black et al. (2013). Briefly, cell suspensions were fixed in ice-cold methanol :glacial acetic acid (3: 1) solution for a minimum of four hours, before being centrifuged onto 8 Chamber Polystyrene vessel tissue culture treated glass slides (Falcon, Fisher Scientific) at 900rpm. The slides were air-dried and incubated in 2X SSC buffer for 2 min, followed by serial ethanol dilution (70%, 85% and 100%) incubations for 2 min each, for a total of 6 min. Air-dried slides were hybridized with probes that were diluted in appropriate buffer overnight at 37°C.
  • the slides were washed the next day for 3 to 4 mins in appropriate wash buffers at 69°C with 0.4X SSC for Cytocell probes or Agilent Bufferl for Agilent probes, followed by washing in 2X SSC with 0.05% Tween-20 (Cytocell probes) or Agilent Buffer 2 for Agilent probes.
  • the slides were incubated in Img/mL DAPI solution made in 1% BSA-PBS, followed by a final IX PBS wash. After the wash, the slides were mounted with ProLong Gold antifade reagent (Invitrogen).
  • FISH images were acquired using an Olympus 1X81 or Olympus 1X83 spinning disk microscope at 40X magnification and analyzed using Slidebook 6.0 softwares. A minimum of 20 z-planes with 0.5um step size was acquired for each field. Copy number gains for MLL1,11C, NMYC/LAF4 were scored in RPE cells as three or more foci. For MEL breakapart probe, copy gains were scored as 3 or more foci for the N terminus flanking probe (green) and C terminus flanking probe (red). Complete separation of red and green probe with no overlap was called breakapart for the MLL locus, TCF3 locus and any other locus FISHed with dual breakapart probe. A minimum of 100 nuclei are scored for each independent experiment. Extended list of probes used are provided in the tables above.
  • RPE cells were transfected with siRNAs and passed 3 times. Cells were seeded for 48 hours. The cells were treated with KaiyoMAX colcemid solution (Gibco) at a final concentration of 2pg/mL for 4 hour and were collected by mitotic shake off, washed with IX PBS followed by 0.59% KC1 (w/v) hypotonic solution for 1 hour 30 minute for expansion and swelling. The reaction was terminated by addition of 3: 1 solution of cold methanol: acetic acid, followed by 4 washes. The cells were then resuspended in fixative solution. The cells were pipetted and dropped on a glass slide from a height of 12-15 inches to make the metaphase spread. FISH was performed for the indicated probes post drying of the slides. The images were taken with 25 z- planes with 0.5 pm step size using the Olympus 1X83 microscope. The images were analyzed for FISH using Slidebook 6.0 software.
  • Doxorubicin treatment RPE cells were plated in 10 cm tissue culture plates at a density of 2.5xl0 5 Cells were allowed to adhere for approximately 16 hours before Doxorubicin (Sigma) (dissolved in DMSO) was supplemented to media in different concentrations. Final concentrations used were 5, 2.5 and 1 pg/pl . Cells were cultured in Doxorubicin for a total of 72 hours before harvesting. For Figures 12D and 14F-14G, cells were transfected with KDM3B plasmid for 4 hr. After removal of transfection mixture, cells were supplemented with 1 pg/pl Doxorubicin supplemented media and cultured for 20 hours before harvesting. For Figures 12C and 14C-14D, Doxorubicin supplemented media was added after removal of EHMT2 siRNA transfection mix and cultured for 72 hours before harvesting.
  • RPE cells were plated in 10cm tissue culture plates at a density of 3x10 5 cells. Cells were allowed to adhere to the plate for a minimum of 24h before JDI12/KDM3i (dissolved in DMSO) was supplemented to media at 25nM unless specified differently. Cells were cultured for a total of 72 hours before harvesting. For washout experiment (Fig. 4C), cells were allowed to adhere for 48h or 60h before treatment with JDI12/KDM3i. Cells were treated for 12h before media was removed, plates were washed with lx PBS, and cells were either harvested or fresh complete media was added back to the plate without JDI12. For passage experiments, RPE cells were plated at 1.5xl0 5 cells.
  • JDI12 + EHMTi (UNC0642) experiments cells were seeded at 3xlO 5 in 10cm tissue culture plates and allowed to adhere for 60h before media was supplemented with JDH2 (25nM) and G9ai (2.5pM). Cells were harvested 12 hours post treatment.
  • siCTCF + G9ai experiments cells were seeded in 10cm tissue culture plates at 2. IxlO 5 and allowed to adhere for 24h before following the transfection procedure described above. 24h post-transfection, media was supplemented with 1 ,5uM G9ai. Cells were cultured for a further 48h (72h total transfection) before being harvested.
  • EHMTi For Dox+EHMTi experiments, cells were plated at 1.5xl0 5 and allowed to adhere for 24h. EHMTi was supplemented to media at a final concentration of 1.5uM. 48h later DOX was supplemented to the media at Ipg/ul. 24h after Dox supplement (72h total EHMTi, 24h total Dox), cells were harvested.
  • KDM3A, KDM3B and KDM5A were incubated with I pg bulk histones(Histone from calf thymus) or 3.3 pM H3K9me2 peptide in 30pl reaction system at 27 degree, 5 hours.
  • Reaction buffer Hepes(PH7.5) 50mM, 2-OG 50pM, Fe(NH4)2(SO4)2 50pM, Sodium L- Acorbate 400pM, TCEP ImM.
  • Recombinant human KDM3B/JMJD1B protein (abeam ab271569) was incubated with lpM KDM3i, IM Tris, 5M NaCl, 10mM Asorbic Acid, lOmM a-Ketoglutarate, lOmM Fe(NH4)2(SO4)2(H2O)6 at 27 C, 30 minutes.
  • IpL Histone from Calf Thymus (Img/mL) in H2O was added to make reaction lOOpL then incubated at 27 C for 5 hrs.
  • 4X Laemmli Loading buffer with 5% P -Mercaptoethanol was added to reaction and then heated 95 C for 10 mins. Samples were snap frozen and then used for western blots.
  • the Digital Droplet PCR was performed using 10 pL of 2 * ddPCR Supermix for Probes (no dUTP) (Bio-Rad), 900 nM of each primer, 250 nM probe, 50 ng of digested DNA template using Hindlll restriction enzyme (NEB) and r2.1 Buffer (NEB), and nuclease free water to a total volume of 20 pl.
  • the QX200 droplet generator (Bio-Rad) was used to generate the droplet mixture.
  • the droplet mixture was then transferred to a PCR reaction plate and amplified with the following conditions: denaturation of 95 °C for 10 min, followed by 40 cycles of a two- step thermal profile consisting of 95 °C for 15 s and 60 °C for 60 s, then incubated at 98 °C for 10 min and cooled to 8°C until the droplets were read.
  • the plate was transferred to the QX200 droplet reader (Bio-Rad) and analyzed for copy number variation (CNV). The number of positive (high level of fluorescence) and negative (low and constant level of fluorescence) droplets obtained were analyzed using QuantaSoft software (Bio-Rad, Pleasanton, CA, USA). Ratios of KMT2A to CD3E gene were used to determine copy number.
  • Primer and probe sequences are provided in Table 2.
  • 3xl0 5 control or KMT2A inherited cells were seeded in a 10 cm plate and grown asynchronously for 72 hours. Cells were treated with 25nM of KDM3i for 12, 6, or 3 hours before prior to harvesting. Collected cells were processed using ALEXA FLUOR 488 conjugated Annexin V and PI staining following the manufacturer’s instructions (Life Technologies). Data was collected on a BD Biosciences Symphony A5 flow cytometer and analyzed using FACSDiva software.
  • Chromatin was prepared and ChIP were performed as described in Mishra et al 2018 and Black el al 2013. Sonication of chromatin was done with the Qsonica Q800R2 system (Qsonica).
  • Qsonica Q800R2 system
  • H3K9Me3, H3K36Me3 and H3K27Me3 ChIP 0.3x10 6 RPE cells were seeded in 10cm plates. Cross linking of the cells were done by adding 1% formaldehyde to the media for 13 min at 37°C and stopped with 0.125.M glycine, pH2.6. Plates were washed with ice cold PBS and scraped off. followed by centrifugation at 800 rpm for 5 min at 4°C.
  • the pellet was resuspended in cellular lysis buffer (5mM PIPES pH8.00, 85mM KC1, 0.5% NP40) supplemented with protease and phosphatase inhibitors, incubated 5min on ice and centrifuged at 800 rpm, 5 min at 4°C.
  • the pellet was resuspended in nuclear lysis buffer (NI..B, 50mM Tris, pH 8.0, 1.0% SDS).
  • Chromatin was sonicated at 70% amplitude 15 s on 45 sec off setting for 35 min. 4 pL of chromatin was reverse cross-linked overnight at 65°C in presence of proteinase K. After RNase treatment, DNA was isolated with phenol: chloroform extraction and checked on 1% agarose gel for a smear below 300bp. 1 -10 ⁇ g of chromatin was precleared by centrifugation at 14,000rpm for 1 Omin at 4°C.
  • chromatin was immunoprecipitated with 0.2-2pg of antibody in dilution IP buffer (16.7mM Tris pH 8.0, 1.2mM EDTA pH 8.0, 167mM NaCl, 0.2% SDS, 0.24% or 1.84% Triston-X-100) at 4°C overnight. Chromatin was precleared for 2 hr each with protein A agarose and magnetic protein A or protein G beads (Invitrogen; to match antibody isotype) before immunoprecipitation.
  • the immunoprecipitated material was washed 2 times in dilution IP buffer, 1 time in TSE buffer (20mM Tris pH 8.0, 2mM EDTA pH8.0, 500mM NaCl, 1% Triton X-100, 0.1% SDS), 1 time in LiCl buffer (lOOmM Tris pH 8.0, 500mM LiC1, 1% deoxycholic acid, 1% NP40) and 2 times in TE (lOmM Tris pH 8.0, ImM EDTA pH8.0) before elution in elution buffer (50niM NaHCCh, 140mM NaCl, 1% SDS) with lOug proteinase K at 1 hr 55°C 1000 rpm.
  • TSE buffer 20mM Tris pH 8.0, 2mM EDTA pH8.0, 500mM NaCl, 1% Triton X-100, 0.1% SDS
  • LiCl buffer lOOmM Tris pH 8.0, 500mM LiC1,
  • Chip sequencing libraries were prepped using the TruSeq ChlP Sample Preparation kit (Illumina). Libraries were single-end sequenced (75 cycles) using a NextSeq500 (Illumina). ChlP-seq analysis:
  • ChlP-seq analysis was performed as previously described (Clarke et al., 2020; Van Rechem et al., 2021; Van Rechem et al., 2020). Sequencing reads were aligned against the human hgl9 reference genome using BWA (Li and Durbin, 2010). Alignments were filtered for uniquely mapped reads and duplicates were removed. Input-normalized ratio coverage tracks were generated using Deeptools (Ramirez et al., 2016).
  • Loss of KDM3B causes site-specific copy gains of MLL/KMT2A locus.
  • KDM3B is a H3K9mel/2 demethylase located in the 5q31.1 region that is associated with KMT2A copy gains and rearrangements (Herry et al., 2006; Hu et al., 2001; Xu et al., 2018; Zatkova el al., 2009).
  • LAML Acute Myeloid Leukemia
  • DNA FISH DNA in situ hybridization
  • RPEs retinal pigment epithelial cells
  • KDM3 family members have comparable H3K9mel/2 activity in vitro, only KDM3B siRNA depletion caused a significant increase in KMT2A copy gains with no significant changes to the 11C control region (Figs. 1D-1E).
  • CD 3 adjacent and partially overlapping FISH probe
  • KDM3B knockdown caused a significant increase in KMT2A gains (black bars) as well as break apart (BA; purple bars) events (Figs. 1F-1G).
  • the copy gains did not appear to have N-terminal (green) or C-terminal (red) bias for the KMT2A gene, including the whole locus of the KMT2A gene (both probes- pseudo-colored yellow) (Fig. IF). These events are focal as the adjacent FISH probe CD3 (Fig. 1C; grey) did not significantly change upon KDM3B depletion (Fig. 1H), emphasizing the site-specific control of KDM3B depletion.
  • KDM3B depletion alters H3K9mel/2 across the KMT2A genomic locus
  • KDM3B is an H3K9mel/2 demethylase (Kim et al., 2012; Wang et al., 2019). Therefore, we performed chromatin immunoprecipitation (ChIP) sequencing for H3K9mel/2/3 methylation marks in control and KDM3B siRNA transfected RPE cells. KDM3B depletion produced genome-wide H3K9mel and H3K9me2 changes that were preferentially skewed towards an increase of these markers.
  • ChIP chromatin immunoprecipitation
  • H3K9mel/2 increased upon KDM3B depletion across the KMT2A gene body, particularly H3K9mel within the 8.3kb breakpoint cluster region (BCR) spanning exons 8-14, which is enriched for the KMT2A rearrangements (data not shown) (Broeker et al., 1996).
  • BCR 8.3kb breakpoint cluster region
  • H3K9mel at BCR was lower than in the adjacent regions, corresponding to the trough of H3K9mel density.
  • KDM3B knockdown led to a strong increase of H3K9mel, so that this trough was reduced and H3K9mel density became more uniform across the whole BCR vicinity.
  • H3K9me2 was also increased on the flank to the BCR.
  • KDM3B binds across KMT2A, with a strong peak within the BCR that was lost upon shRNA-mediated KDM3B depletion. Furthermore, KDM3B also binds across TCF3 and AFF3.
  • KDM3B depletion alters H3K9mel/2 methylation landscape of the KMT2A locus, especially H3K9mel at the BCR region, and contributes to the KMT2A copy gains and break apart events, which is consistent with other regulated regions.
  • KDM3B Inhibition or depletion of KDM3B causes inherited KMT2A copy gains and genomic alterations
  • KDM3i a recently identified KDM3 family inhibitor (JDI-12, referred to as KDM3i (Xu et al., 2020)
  • KDM3i a recently identified KDM3 family inhibitor
  • KDM3B inhibition was tested on a panel of primary and cancer cell lines, which included KGla cells, a primary AML derived cell line and a primary AML organoid model as well as primary Hematopoietic Stem and Progenitor Cells (HSPC).
  • KDM3i treatment significantly increased KMT2A copy gains and genomic rearrangements in all lines tested (Figs. 2F-2J) This demonstrates that KDM3B inhibition promotes conserved KMT2A DNA amplification and rearrangements across cancer and non-cancer cells as well as primary human cells.
  • KMT2A amplifications occur as both extrachromosomal and integrated events (Herry et al., 006; Streubel et al., 2000).
  • KDM3i transient site-specific extrachromosomal copy gains
  • TSSGs transient site-specific extrachromosomal copy gains
  • KMT2A DNA copy gains and break apart events occur as soon as 12 hours after KDM3i treatment (Fig. 2K, KDM3i 12hrs), but are no longer observed upon KDM3i removal from the cells (Fig.
  • G9a is a Vietnamese H3K9mel/2 KMT that regulates replication and is loaded in a ternary complex with PCNA (Esteve et al., 2006).
  • EHMTi a dual inhibitor for G9a/EHMT2 and EHMT1 (EHMTi) (Liu et al, 2013).
  • H3K9mel increase was rescued across 81% of regions genome-wide (88 Mb out of total 109 Mb), whereas H3K9me2 increase was rescued across 75% of regions genome-wide (219 Mb out of total 292 Mb), suggesting that maintaining a KDM3B-G9a balance is critical for controlling H3K9mel/2 levels genome-wide.
  • HL60 cells have a 5q allelic loss (Hejlik and Nagarajan, 2005) and appeared to have lower CTCF occupancy when compared to the other cells that were assessed. Therefore, we hypothesized that KDM3B depletion could disrupt CTCF binding and promote KMT2A amplification and genomic rearrangement.
  • KDM3B depletion could disrupt CTCF binding and promote KMT2A amplification and genomic rearrangement.
  • CTCF depletion alone was sufficient to promote significant KMT2A site-specific copy gains and genomic rearrangements that were not enhanced by KDM3B depletion (Fig. 5B). Since the CTCF peak within exon 11 of KMT2A directly overlapped with the KDM3B peak (Fig.
  • H3K9mel levels increased at least 1.5-fold in the 5 Kb proximity of approximately 1,000 CTCF peaks genome-wide (Figure 5H).
  • Figure 5H Upon the double knockdown of KDM3B and G9a, this increase was rescued at the majority (-80%) of these peaks, where H3K9mel no longer showed this increase (Figure 5H).
  • Figure 51 the scatterplots in Figure 51 show comparisons of H3K9mel levels for the proximal regions of the 17,077 CTCF sites after KDM3B knockdown (left), G9a knockdown (middle), and the double knockdown of KDM3B and G9a (right).
  • KDM3B knockdown ( Figure 51, left plot) resulted in a preferential increase of H3K9mel (the part above the upper red line corresponding to > 1.5-fold increase).
  • G9a knockdown ( Figure 51, middle plot) resulted in decreased H3K9mel (the part below the upper red line corresponding to > 1.5-fold decrease).
  • H3K9mel changes were strongly reduced, with smaller extent of differences from control in either direction (above and below red lines).
  • CTCF is known to regulate gene expression (Phillips and Corces, 2009).
  • CTCF depletion regulated KDM3B expression levels While depletion of CTCF modestly suppressed KDM3B transcript levels, both KDM3B and G9a protein levels were not significantly reduced, suggesting that CTCF is likely a downstream effector of KDM3B loss and H3K9mel/2 disruption.
  • We tested this by depleting or chemically inhibiting G9a in combination with CTCF depletion. In fact, G9a depletion/inhibition was sufficient to block CTCF-induced amplifications (Figs. 5J-5K).
  • Doxorubicin promotes KMT2A amplification and rearrangement as well as reduces KDM3B and CTCF protein levels
  • KMT2A amplification and a host of genomic rearrangements are observed in pediatric leukemia and therapy-induced leukemia when conventional chemotherapy is used to treat several cancer types (Felix, 1998; Pedersen-Bjergaard et al., 1998; Sanjuan-Pla et al., 2015).
  • KMT2A amplified and rearranged MDS and AML are generated after topoisomerase II (topo II) inhibitor treatment (e.g., Doxorubicin, Dox)(Godley and Larson, 2008).
  • topoisomerase II topo II
  • topo II inhibitor treatment e.g., Doxorubicin, Dox
  • Dox treatment promoted KMT2A, AFF3 and TCF3 copy gains and genomic alterations in RPE cells with no significant impact on control regions as determined by DNA FISH (Fig. 6A).
  • lower doses of Dox 1 pg/pl and 5pg/pl
  • Dox treatment also promoted KMT2A copy gains and rearrangements in primary HSPCs (Fig. 6B).
  • KDM3B and CTCF regulation controls Doxorubicin-induced KMT2A amplification and rearrangement
  • KDM3B-G9a balance controls CTCF occupancy, and in turn, the ability of the KMT2A locus to undergo sitespecific copy gains and genomic rearrangement.
  • KDM3B depletion or inhibition as well as G9a overexpression, promotes the amplification of specific sites, suggesting these proteins are maintaining the methylation balance and capacity for amplification of the affected region.
  • KMT2A gene locus G9a is facilitating the amplification and rearrangement
  • KDM3B and CTCF are counterbalancing these genomic alterations, illustrating the critical requirement for histone methylation regulation and the control of the transient amplification events ( Figure 7F).
  • Doxorubicin treatment reduced KDM3B and CTCF protein levels, at least in part through activation of the UPS, which resulted in KMT2A DNA copy gains and rearrangement.
  • KMT2A rearrangement partners There are currently more than 100 known KMT2A rearrangement partners documented (Meyer et al., 2023). These KMT2A rearrangements result in the fusion of the gene to any of the partner genes, leading to protein chimeras. KMT2A can also rearrange to several noncoding regions throughout the genome (Meyer et al., 2023). Therefore, not all rearrangement events generate functional fusion proteins (Apian, 2006b). These data suggest that the molecular mechanism(s) leading to the generation of KMT2A rearrangements, including those that do not generate translatable products, could be key to understanding tumors containing amplifications and rearrangements.
  • KDM3B suppression or loss alone generates inheritable rearrangements of KMT2A, it is likely just the first step necessary to promote or allow the selection of the rearrangement events resulting in functional fusion proteins that provide a cellular growth advantage.
  • Mouse knock-in studies of the major KMT2A fusion genes demonstrate that even the most common oncogenic associated fusions have a varying range of latency before developing into leukemia [reviewed in (Liu et al., 2009)].
  • the resulting fusion proteins generated clearly provide differential levels of cellular fitness advantage before developing into homogenous rearrangements and promoting leukemia. Furthermore, studies show that nonhom ologous end-joining is required for topo II inhibitor driven leukemia-associated KMT2A rearrangements (Gomez-Herreros et al., 2017; Gothe et al., 2019), suggesting that mis-regulated DNA damage responses are likely another factor involved in generating/selecting for the oncogenic fusion events observed in leukemia. However, additional influences could also potentiate the driver fusion events to emerge, including without limitation, cellular ageing, stress exposures, and/or acquired mutations.
  • del(5q) regions contain KDM3B (Eisenmann etal., 2009). However, patients with del(5q) alone have a better prognosis compared to those presenting with del(5q) as well as other mutations or abnormalities (Giagounidis et al., 2004). Since KDM3B loss was not responsible for gains or rearrangements of a panel of other KMT2A fusion partners (data not shown), we suspect that additional gene mutations and/or the dysregulation of additional epigenetic regulators are likely required to promote copy gains and rearrangements of the oncogenic fusion partners, providing the secondary hit(s) necessary.
  • KDM3B and CTCF as a bridge to Dox-induced KMT2A amplification and rearrangement
  • topo II inhibitor treatment e.g., Doxorubicin, Dox
  • MDS and AML occur and are accompanied by amplification and rearrangement of the KMT2A locus (Godley and Larson, 2008).
  • Prior studies demonstrate that topo II inhibitors promote non-leukemia and leukemia associated KMT2A rearrangements in various cell types (Gothe et al., 2019; Libura et al., 2005), which suggest a more universal regulatory mechanism controlling KMT2A alterations.
  • a population-based study demonstrated that younger individuals developing secondary leukemia have a significantly worse prognosis compared to de novo (Hulegardh el al., 2015).
  • CTCF occupancy m KMT2A was reduced with Dox treatment, while overexpression of CTCF prevented the Dox-induced KMT2A copy gain events.
  • Appendix I Sequences encoding the genes and proteins useful for practicing the invention and regions of interest for peptide mimetic binding and siRNA targeting.
  • KDM4A lysine demethylase induces site-specific copy gain and rereplication of regions amplified in tumors.
  • TDP2 suppresses chromosomal translocations induced by DNA topoisomerase II during gene transcription. Nat Commun 8, 233. 10.1038/s41467-017-00307-y.
  • a novel nuclear protein, 5qNCA (LOC51780) is a candidate for the myeloid leukemia tumor suppressor gene on chromosome 5 band q31. Oncogene 20, 6946-6954. 10.1038/sj. one.1204850.
  • KDM3B is the H3K9 demethylase involved in transcriptional activation of lmo2 in leukemia. Mol Cell Biol 32, 2917-2933. 10.1128/mcb.00133-12.
  • a novel transgenic mouse model reveals deregulation of the ubiquitin- proteasome system in the heart by doxorubicin. Faseb j 19, 2051-2053. 10.1096/fj.05-3973fje.
  • KDM3 epigenetically controls tumorigenic potentials of human colorectal cancer stem cells through Wnt/p-catenin signalling. Nature Communications 8, 15146. 10.1038/ncommsl5146.
  • MLL fusions pathways to leukemia. Cancer Biol Ther 8, 1204-1211. 10.4161/cbt.8.13.8924.
  • CTCF master weaver of the genome. Cell 137, 1194-1211. 10.1016/j. cell.2009.06.001.
  • Acute myeloid leukemia with a complex aberrant karyotype is a distinct biological entity characterized by genomic imbalances and a specific gene expression profile.
  • KDM3B suppresses APL progression by restricting chromatin accessibility and facilitating the ATRA- mediated degradation of PML/RARalpha. Cancer Cell Int 19, 256. 10.1186/sl2935-019-0979-7.
  • KDM3B shows tumor-suppressive activity and transcriptionally regulates HOXA1 through retinoic acid response elements in acute myeloid leukemia. Leuk Lymphoma 59, 204-213. 10.1080/10428194.2017.1324156.
  • AML/MDS with 1 Iq/MLL amplification show characteristic gene expression signature and interplay of DNA copy number changes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des compositions et des procédés de contrôle de gains de copies régiospécifiques transitoires et d'insertion génomique associés à une leucémie de lignée mixte.
PCT/US2023/025793 2022-06-17 2023-06-20 Compositions et procédés de contrôle de gains de copies régiospécifiques transitoires, d'insertions génomiques et de réarrangements associés à une leucémie de lignée mixte WO2023244870A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263353521P 2022-06-17 2022-06-17
US63/353,521 2022-06-17

Publications (1)

Publication Number Publication Date
WO2023244870A1 true WO2023244870A1 (fr) 2023-12-21

Family

ID=89191873

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/025793 WO2023244870A1 (fr) 2022-06-17 2023-06-20 Compositions et procédés de contrôle de gains de copies régiospécifiques transitoires, d'insertions génomiques et de réarrangements associés à une leucémie de lignée mixte

Country Status (1)

Country Link
WO (1) WO2023244870A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140161785A1 (en) * 2012-12-10 2014-06-12 Feiyan Liu Verticillin A Inhibition of Histone Methyltransferases
US20180045727A1 (en) * 2015-03-03 2018-02-15 Caris Mpi, Inc. Molecular profiling for cancer
WO2018102736A1 (fr) * 2016-12-01 2018-06-07 The General Hospital Corporation Procédés de traitement du cancer
US20190211036A1 (en) * 2016-09-14 2019-07-11 Janssen Pharmaceutica Nv Spiro bicyclic inhibitors of menin-mll interaction
CN110680816A (zh) * 2018-07-05 2020-01-14 潍坊医学院 化合物在制备治疗肿瘤的药物中的用途

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140161785A1 (en) * 2012-12-10 2014-06-12 Feiyan Liu Verticillin A Inhibition of Histone Methyltransferases
US20180045727A1 (en) * 2015-03-03 2018-02-15 Caris Mpi, Inc. Molecular profiling for cancer
US20190211036A1 (en) * 2016-09-14 2019-07-11 Janssen Pharmaceutica Nv Spiro bicyclic inhibitors of menin-mll interaction
WO2018102736A1 (fr) * 2016-12-01 2018-06-07 The General Hospital Corporation Procédés de traitement du cancer
CN110680816A (zh) * 2018-07-05 2020-01-14 潍坊医学院 化合物在制备治疗肿瘤的药物中的用途

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LIBURA JOLANTA, SLATER DIANA J., FELIX CAROLYN A., RICHARDSON CHRISTINE: "Therapy-related acute myeloid leukemia–like MLL rearrangements are induced by etoposide in primary human CD34+ cells and remain stable after clonal expansion", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 105, no. 5, 1 March 2005 (2005-03-01), US , pages 2124 - 2131, XP093122917, ISSN: 0006-4971, DOI: 10.1182/blood-2004-07-2683 *

Similar Documents

Publication Publication Date Title
US8283331B2 (en) Methods to regulate miRNA processing by targeting Lin-28
EP2688595B1 (fr) Régulation par lin28 de la biogenèse de let-7
WO2014077354A1 (fr) Arn non codant à longue chaîne mis en œuvre dans un traitement contre le cancer
TW201402813A (zh) 對單核苷酸kras突變具專一性之雙功能性短髮夾rna(bi-shrna)
JP7392954B2 (ja) トリプルネガティブ乳癌の治療方法
US20180320187A1 (en) TOXIC RNAi ACTIVE SEED SEQUENCES FOR KILLING CANCER CELLS
US20220275374A1 (en) Oligonucleotide interference treatments of prostate cancer
US20170035795A1 (en) Methods and reagents for the diagnosis and treatment of acute leukemia
US10161008B2 (en) miR-193a-3p and associated genes predict tumorigenesis and chemotherapy outcomes
US11013754B2 (en) Compositions and methods for treating cancer
JP7406278B2 (ja) 腫瘍マーカー及び治療ターゲットとしての長鎖ノンコーディングrna letn
TW201119681A (en) Compositions and methods for inhibiting expression of KIF10 genes
WO2023244870A1 (fr) Compositions et procédés de contrôle de gains de copies régiospécifiques transitoires, d'insertions génomiques et de réarrangements associés à une leucémie de lignée mixte
WO2021108546A1 (fr) Inhibition de microarn-224 pour traiter l'hypertension pulmonaire
EP4058604A1 (fr) Compositions et méthodes pour réguler l'amplification de l'egfr dans des cellules cancéreuses pour améliorer un agent anticancéreux ciblant l'egfr
US20200157537A1 (en) Modulating RNA Interactions with Polycomb Repressive Complex 1 (PRC1)
US11987795B2 (en) Methods of modulating SLC7A11 pre-mRNA transcripts for diseases and conditions associated with expression of SLC7A11
KR102143701B1 (ko) nc886 및/또는 PKR 저해제를 포함하는 항암 보조제, 및 암 치료를 위한 약물의 정보 제공 방법
US20240110190A1 (en) Compositions and methods for treating pulmonary arterial hypertension (pah) and other disorders
WO2023196944A2 (fr) Traitements du cancer de la prostate par interférence d'oligonucléotides
WO2016201438A1 (fr) Nouvelles cibles modulées par un variant occasionnel du diabète de type 2 (t2d) incorporé dans le gène tcf7l2 et procédés d'utilisation de celles-ci pour l'identification d'agents présentant une efficacité dans le traitement du diabète de type 2 et d'autres troubles métaboliques
Macha et al. Alternative Splicing and Cancer
Boujnouni et al. Block or degrade? Balancing on-and off-target effects of antisense strategies against transcripts with expanded triplet repeats in DM1.
WO2022241165A2 (fr) Compositions et méthodes d'utilisation de hotair muté dans le traitement de cancers
AU2022384267A1 (en) Lncrna transcripts in melanomagenesis

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: 23824692

Country of ref document: EP

Kind code of ref document: A1