WO2017223268A1 - Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti) - Google Patents

Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti) Download PDF

Info

Publication number
WO2017223268A1
WO2017223268A1 PCT/US2017/038669 US2017038669W WO2017223268A1 WO 2017223268 A1 WO2017223268 A1 WO 2017223268A1 US 2017038669 W US2017038669 W US 2017038669W WO 2017223268 A1 WO2017223268 A1 WO 2017223268A1
Authority
WO
WIPO (PCT)
Prior art keywords
beti
cancer
cells
tgfp
composition
Prior art date
Application number
PCT/US2017/038669
Other languages
English (en)
Inventor
Benjamin Turk
David Calderwood
Joseph Schlessinger
Xiarong SHI
Jamie BATES
David Breckenridge
Saritha KUSAM
Original Assignee
Yale University
Gilead Sciences, Inc.
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 Yale University, Gilead Sciences, Inc. filed Critical Yale University
Publication of WO2017223268A1 publication Critical patent/WO2017223268A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA 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
    • C12N15/1136Non-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 growth factors, growth regulators, cytokines, lymphokines or hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.

Definitions

  • Epigenetic regulation of transcription is central to control of cell fate and proliferation. Enzymatic addition or removal of a variety of specific post-translational modifications of histones support the recruitment of epigenetic "readers," proteins that selectively bind to modified sites and recruit transcriptional activators or repressors. Alterations in this complex epigenetic code contribute to development of a range of diseases, including cancer. Consequently pharmacological modulation of enzymes that generate or remove epigenetic modifications and their readers offer new therapeutic opportunities for cancer treatment.
  • bromodomain and extraterminal domain (BET) proteins are one class of epigenetic readers involved in transcriptional control.
  • the small family of BET proteins (BRD2, BRD3, BRD4 and BRDT) are characterized by tandem
  • BET proteins in particular BRD4, have been implicated as general regulators of transcription through recruitment of the elongation factor, P-TEFb, to gene promoters and through interaction with the mediator complex.
  • P-TEFb elongation factor
  • BET proteins in particular BRD2 and BRD4, in a range of cancers (Baratta et al., 2015, Proc Natl Acad Sci USA 112(l):232-237; Zuber et al., 2011, Nature 478(7370):524-528).
  • BETi BET inhibitors
  • BETi suppress growth and survival of at least some types of cancer cells is by preferentially repressing transcription of the proto-oncogene, MYC, which is often under the control of BRD4 (Dawson et al., 2011, Nature 478(7370):529- 533; Delmore et al., 2011, Cell 146(6):904-917; Mertz et al., 2011, Proc Natl Acad Sci USA 108(40): 16669-16674).
  • BETi may provide a new mechanism to target MYC and other oncogenic transcription factors, which lack obvious binding pockets for small molecules and are thus typically considered to be "undruggable”.
  • the present invention provides a method of increasing anti-tumor sensitivity to a bromodomain and extraterminal domain protein inhibitor (BETi) in a cell in a subject, the method comprising administering a composition comprising a TGFP pathway inhibitor and a BETi to a subject in need thereof, wherein the TGFP pathway inhibitor sensitizes the cell to the BETi.
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the present invention further provides a method of treating a tumor comprising administering to a subject in need thereof a composition comprising a TGFP pathway inhibitor and a BETi, wherein the TGFP pathway inhibitor sensitizes the tumor to the BETi.
  • the present invention further provides a method of increasing anti-tumor sensitivity to a BETi of a cell in a subject, the method comprising administering a composition comprising a BETi and one selected from the group consisting of TRTM33 or a fragment thereof, and a nucleic acid encoding TREVI33 to a subject in need thereof, wherein the TRF 33 sensitizes the cell to the BETi.
  • the present invention further provides a method of treating a tumor comprising administering to a subject in need thereof a composition comprising a BETi and at least one selected from the group consisting of TRF 33 or a fragment thereof, and a nucleic acid encoding TRF 33, wherein TRF 33 sensitizes the tumor to the BETi.
  • the TGFP pathway inhibitor is selected from the group consisting of a small molecule inhibitor, an inhibitory nucleic acid, neutralizing antibody, and an antagonist.
  • the small molecule inhibitor is selected from the group consisting of SB431542, A83-01, RepSox, SB208, SB505124, LY364947, LY2157299, R268712, D4476, SB525334, GW788388, TEW-7197, and any combination thereof.
  • the antagonist is selected from the group consisting of HtrAl, decorin, biglycan, fibromodulin, lumican, endoglin, somatostatin, follistatin, RAP-1332, pirfenidone (5-methyl-l-phenyl-2(lH)-pyridone), soluble ectodomains of TGFp receptor type II (RII) or betaglycan (BG), and any combination thereof.
  • the neutralizing antibody specifically binds one selected from the group consisting of TGF and a TGF receptor.
  • the nucleic acid encoding TREVI33 is an expression vector comprising a TRIM33 gene.
  • the vector is a viral vector.
  • the BETi is selected from the group consisting of JQ1, GS-626510, GS-5829, BMS 986158, RVX2135, CPI203, CPI-0610, ABBV-075, BAY1238097, INCB054329, FT-1101, PFI-1, 1-BET151, ZEN-3365, 1-BET762, OTX015, TEN-010, and any combination thereof.
  • the subject has a cancer.
  • the cancer is selected from the group consisting of breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, and lung cancer.
  • the method treats a tumor wherein the tumor is a cancer selected from the group consisting of breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, and lung cancer.
  • the cell in the subject is BETi resistant. In other embodiments, the tumor is BETi resistant.
  • the invention further provides a composition comprising a TGFP pathway inhibitor and a bromodomain and extraterminal domain protein inhibitor (BETi), wherein the TGFP pathway inhibitor sensitizes cells to the BETi.
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the TGFP pathway inhibitor is selected from the group consisting of a small molecule inhibitor, an inhibitory nucleic acid, a neutralizing antibody, and an antagonist.
  • the small molecule inhibitor is selected from the group consisting of SB431542, A83-01, RepSox, SB208, SB505124, LY364947, LY2157299, R268712, D4476, SB525334, GW788388, TEW-7197, and any combination thereof.
  • the neutralizing antibody specifically binds one selected from the group consisting of TGF and a TGF receptor.
  • the antagonist is selected from the group consisting of HtrAl, decorin, biglycan, fibromodulin, lumican, endoglin, somatostatin, follistatin, RAP-1332, pirfenidone (5-methyl-l-phenyl-2(lH)-pyridone), soluble ectodomains of TGFP receptor type II (RII) or betaglycan (BG), a neutralizing antibody against TGFP or a TGFP receptor, and any combination thereof.
  • the invention also provides a composition comprising a BETi and at least one selected from the group consisting of TREVI33 or a fragment thereof, and a nucleic acid encoding TREVI33, wherein the TGF pathway inhibitor sensitizes cells to the BETi.
  • the nucleic acid encoding TRIM33 is a vector comprising a TRIM33 gene. In other embodiments, the vector is a viral vector.
  • the BETi is selected from the group consisting of JQl, GS-626510, GS-5829, BMS 986158, RVX2135, CPI203, CPI-0610, ABBV-075, BAY1238097, INCB054329, FT-1101, PFI-1, 1-BET151, ZEN-3365, 1-BET762, OTX015, TEN-010, and any combination thereof.
  • compositions of the invention can be formulated in a pharmaceutical composition further comprising a pharmaceutically acceptable carrier.
  • pharmaceutical composition can be used in the
  • Figures 1 A-1H are images showing that the shRNA screening revealed TREVI33 as a regulator of BETi resistance in cancer cells.
  • Figure 1A shows the structures of the two different BETi used in the study, JQl and GS-626510.
  • Figure IB shows the ⁇ D values of GS-626510 for 40 bromodomains (Table 3) determined with a
  • Figure 1C shows the dose- dependent inhibition of RKO cell proliferation by JQl and GS-626510 in a 5-day assay. Relative viable cell number was determined by CellTiter Glo assay. Figure ID shows that both GS-626510 and JQl down-regulate MYC protein levels. RKO cells were treated with increasing concentrations of BETi for 3 h and MYC levels in whole cell lysates were assessed by immunoblotting. Actin was used as a loading control.
  • Figure IE shows gene expression changes induced by JQl and GS-626510 in shCTRL cells were highly correlated.
  • FIG. IF shows the scheme of the shRNA screening procedure.
  • Cells infected by the pooled shRNA library were propagated through 8 doublings in presence of either DMSO vehicle control or different concentrations of JQl or GS-626510.
  • Genomic DNA was extracted from the TO (reference) and T4 conditions for determination of proviral shRNA abundance.
  • Figure 1G shows the top 10 enriched target genes revealed by RIGER analysis in each condition.
  • TRJJVI33 was among the top 3 ranked genes in all four BETi conditions but not in the DMSO condition.
  • Figure 1H shows multiple individual TRJJVI33 shRNAs were enriched in BETi-treated, but not in DMSO control treated conditions. Log 2 fold change (T4/T0) of each shRNA is plotted from the most depleted to the most enriched. Each red line represents a single shRNA targeting TRJJVI33.
  • Figures 2A-2J are images showing loss of TRJJVI33 conferred resistance to BETi.
  • Figure 2A shows (top) a schematic of TRIM33 domain organization and positions of two pairs of RT-PCR primers, (middle) TRIM33 mRNA levels determined by RT-PCR in shCTRL cell line and cell lines expressing four different TRJJVI33- targeting shRNAs, and (bottom) TRJJVI33 protein levels in these cell lines.
  • Figure 2B shows shCTRL or shTRJJVI33 cells were seeded in a 6-well plate (3 x 10 5 cells per well) in the presence of DMSO, 100 nM JQl or 50 nM GS-626510 and cumulative cell numbers were assessed every 3 days for up to 15 days.
  • Figure 2C shows the growth inhibition assay.
  • shCTRL and shTRJJVI33 cells were cultured with different
  • FIG. 2D shows the IC 50 values (mean ⁇ SEM) calculated from 5 independently performed growth inhibition assays using shCTRL and shTRJJVI33 cells. P values are based on paired t-test.
  • Figure 2E is a set of graphs showing the effect of TRJJVI33 depletion on JQl or GS-626510 sensitivity in a panel of cancer cell lines;
  • Figure 2E shows the IC50 values for each cell line expressing either shCTRL or shTRJJVI33 derived from 3 independent growth inhibition assays and the mean ⁇ SEM of the fold change in IC50 (shTRFM33/shCTRL) calculated (*P ⁇ 0.05, paired t-test).
  • Figure 2F shows 2 x 10 4 shCTRL or shTRJJVI33 cells plated in 6-well plates, treated with DMSO, 100 nM JQl, or 50 nM GS-626510 for two weeks and then stained with crystal violet.
  • FIG. 2G shows the cell proliferation assay of cell lines expressing two independent shRNAs (B5 and A 12) targeting TRJJVI33. Cells were cultured in 100 nM JQl, or 50 nM GS-626510 for two weeks and then stained with crystal violet.
  • Figure 2H shows the shCTRL or shTRJJVI33 cells transduced with either an empty vector control or TRIM33-expressing lentivirus and cell growth was assessed as in Figure 2F.
  • Figure 21 shows TRHVI33 expression levels in cells from Figure 2H assessed by immunoblotting.
  • Figure 2J shows the crystal violet quantification measured at 590 nm absorbance corresponding to Figure 2H.
  • Figures 3 A-3E are graphs showing RT-PCR quantification of mRNA levels of
  • Figures 3F-3J are images showing RNAseq analysis of vehicle or BETi -treated shCTRL or shTRFM33 cells. Waterfall plots show gene expression changes induced by 3 h treatment of shCTRL RKO cells with 1 ⁇ JQl ( Figure 3F) or 0.3 ⁇ GS-626510 (Figure 3G). MYC is down-regulated by both JQl and GS-626510.
  • Figure 3H shows the top 10 sequence motifs enriched in promoter regions of genes down-regulated >2- fold by JQl and GS-626510 in shCTRL cells were determined by Gene Set Enrichment Analysis (Broad Institute).
  • Figure 31 shows the gene expression changes induced by shTRJJVI33 in RKO cells.
  • Figure 3 J shows an immunoblot showing that BRD4 protein level is not changed by TRJJVI33 knockdown.
  • Figures 4A-4F are a panel of images showing TRIM33 modulated MYC sensitivity to BETi.
  • Figure 4 A shows the normalized RNAseq reads of MYC mRNA from two replicate experiments before and after JQl or GS626510 treatment.
  • Figure 4B shows the RT-PCR quantification of MFC mRNA in shCTRL, shTRJJVI33 and shTRJJVI33 rescued (shTRJJVI33 RES ) cells, either untreated or treated with BETi for 3 h.
  • Figure 4C shows cells treated similarly as in panel Figure 4B ⁇ analyzed for MYC protein.
  • Figure 4D shows the MYC protein levels in control or MYC over-expressing cells before and after BETi treatment for 3 h.
  • Figure 4E shows crystal violet staining of control or MYC over-expressing cells growing with DMSO, JQl or GS-626510 for two weeks.
  • Figure 4F shows the cumulative cell growth of control or MYC-over
  • Figures 4G and 4H show the ChIP at MYC gene promoter region. Line threshold indicates IgG control level.
  • Figure 4G shows the TRJJVI33 ChIP using 4 different primer pairs (#2, #14, #15 and #16) in the MYC promoter region.
  • Figure 4H shows the BRD4 ChIP using the same set of primers as in Figure 4G.
  • Figure 5 A shows the shCTRL or shTREVB 3 -transduced RKO and SK-CO-1 cells treated with 1 ⁇ of JQ1 or 0.3 ⁇ of GS626510 for 24 hours. MYC levels were determined by immunoblotting and ERKl/2 was used as a loading control.
  • Figures 5B-5E show the gene set enrichment analysis (GSEA) and down- regulation of TGFP and MYC signatures by JQ1 was significantly decreased in shTREVI33 in comparison to shCTRL cells ( ES: normalized enrichment score).
  • GSEA gene set enrichment analysis
  • Figures 5F-5V are a set of images showing inhibition of TGFP signaling potentiated the anti-proliferative effects of BETi.
  • Figure 5F shows the TGFpi ligand stimulated phosphorylation of SMAD2 potentiated in shTRFM33 cells.
  • shCTRL or shTREVI33 RKO cells were treated with increasing doses of TGFpi for 25 min (left panel) or with 2 ng/ml TGFpi for various times (right panel), cells were lysed and immunoblotted for phospho-SMAD2 (pSMAD2), total SMAD2 and TREVI33.
  • Figure 5G shows the shCTRL or shTREVI33 cells untreated or treated with 100 pM of TGFpi for 25 min and SMALM was immunoprecipitated. Co-precipitating pSMAD2 was assessed by immunoblotting.
  • Figure 5H shows the shCTRL or shTRFM33 cells infected with lentivirus encoding shCTRL or one of two hairpins targeting SMALM (shSMALM- 3 or shSMAD4-4). Cells were untreated or treated with 100 pM of TGFp 1 for 25 min SMALM, pSMAD2 and total SMAD2 levels were assessed by immunoblotting.
  • Figure 51 shows the TGFp receptor II ( ⁇ ) mRNA from RNAseq in shCTRL and shTRFM33 cells.
  • Figures 5J and 5K show the ChIP at TGFBR2 (TpRII) gene promoter region. Line threshold indicates IgG control level.
  • Figure 5 J shows the TRFM33 ChIP using two different pair of primers (#4 and #3) amplifying TGFBR2 gene promoter region.
  • Figure 5K shows the BRD4 ChIP using the same set of primers as in Figure 5 J amplifying TGFBR2 gene promoter region.
  • Figures 5L-5N show the inhibition of TGFP pathway by silencing TpRII increases the magnitude of cell growth inhibition by BETi.
  • Figure 5L shows the RT-PCR quantification of ⁇ ⁇ mRNA levels in shCTRL and shTRFM33 cells expressing control (shCTRL) or two different TpRII-targeting shRNAs (shTpRII-3 and shTpRII-4).
  • Figure 5M shows cells from Figure 5L stimulated with 100 pM of TGFpi for 25 min and pSMAD2 levels assessed by immunoblotting.
  • Figure 5N shows shCTRL cells (left) or shTRFM33 cells (right) expressing control and TpRII-targeting shRNAs cultured for 2 weeks with DMSO or different concentrations of BETi (as indicated) and then stained with crystal violet.
  • Figure 50 shows the crystal violet quantification measured at 590 nm absorbance corresponding to Figure 5N.
  • Figures 5P and 5Q show the TpRI inhibitor LY2157299 potentiated BETi-mediated inhibition of cell proliferation.
  • Figure 5P shows the shTREVI33 cells pre-treated with increasing doses of LY2157299 and then exposed to 100 pM TGFpi for 25 min.
  • FIG. 5Q shows the shCTRL and two shTREVI33 KD cell lines cultured in the presence of JQ 1 or GS-626510, with or without LY2157299 for 2 weeks and stained with crystal violet.
  • Figure 5R shows the crystal violet quantification measured at 590 nm absorbance corresponding to Figure 5Q.
  • Figure 5S shows the shCTRL or shTREVI33 cells treated with 1 ⁇ JQ1 or 0.3 ⁇ GS-626510 with or without 5 ⁇ LY2157299 overnight and MYC protein levels assessed by immunoblotting.
  • Figures 5T, 5U and 5 V show that the over-expression of TpRII was not sufficient to induce resistance to BETi.
  • Figure 5T shows the pLentiCMV-EV or pLentiCMV-TpRII transduced stable cell lines treated with increasing doses of TGFpi for 25 min and pSMAD2 levels assessed by immunoblotting.
  • Figure 5U shows the cells from Figure 10D cultured in the presence of DMSO, 100 nM JQ1 or 50 nM GS-626510 for 2 weeks and stained with crystal violet.
  • Figure 5 V shows the pLentiCMV-EV or pLentiCMV-TpRII stable cell lines treated with 1 ⁇ JQ1 or 0.3 ⁇ GS-626510 for overnight either in the presence or in the absence of 100 pM TGFpi .
  • MYC levels were determined by immunoblotting and actin was used as a loading control.
  • an element means one element or more than one element.
  • antagonist refers to a compound or molecule that inhibits or decreases a biological response.
  • An antagonist has affinity for a target, such as a receptor, and binding to the target disrupts or prevents the interaction between the target and its cognate binding partner to inhibit or decrease activity of the target.
  • An antagonist may also block the action of a stimulating ligand by binding to an allosteric site that may "lock" the target in an inactive state or prevent the dimerization or oligomerizarion of the target protein
  • bromodomain and extraterminal domain refer to epigenetic readers involved in transcriptional control.
  • the small family of BET proteins are characterized by tandem bromodomains, that bind acetylated lysine residues in histones and other proteins, and a C-terminal extraterminal domain responsible for interactions with chromatin regulators.
  • BET proteins include, but are not limited to, BRD2, BRD3, BRD4 and BRDT.
  • bromodomain and extraterminal domain inhibitor refers to a small molecule that interacts with the acetylated lysine binding pocket of the BET family bromodomains and displaces the BET proteins from binding to chromatin. BETi demonstrate anti-tumor activity in a range of malignancies. Some BETi exert antiproliferative effects that disrupt oncogenic pathways.
  • BETi examples include, but are not limited to, JQ1, GS-626510, GS-5829, BMS 986158, RVX2135, CPI203, CPI-0610, ABBV-075, BAY1238097, INCB054329, FT-1101, PFI-1, 1-BET151, ZEN- 3365, 1-BET762, OTX015, and TEN-010. Additional examples of BETi include compounds disclosed in U.S. Patent Nos. 9,255,089 and 9,108,953, and U.S. Patent Publication No. 2014/0336190, all of which are hereby incorporated herein in their entireties.
  • BETi resistant refers to a loss or decrease in anti- tumor efficacy or response of a cellular target to a BETi.
  • BETi sensitivity refers to anti-tumor activity of a BETi in a cellular target.
  • Increasing anti-tumor sensitivity refers to increasing an anti-tumor response in a cellular target to the BETi by increasing a basal level of the anti-tumor response or resensitizing the cellular target to the BETi.
  • cancer as used herein is defined as disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body.
  • cancers include but are not limited to, breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, lung cancer, myeloma and the like.
  • an effective amount is meant the amount required to reduce or improve at least one symptom of a disease relative to an untreated patient.
  • the effective amount of an active compound(s) used for therapeutic treatment of a disease varies depending upon the manner of administration, the age, body weight, and general health of the subject.
  • expression is defined as the transcription and/or translation of a particular nucleotide sequence driven by its promoter.
  • fragment is meant a portion of a polynucleotide or nucleic acid molecule. This portion contains, preferably, at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the entire length of the reference nucleic acids.
  • a fragment may contain 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000 or 2500 (and any integer value in between) nucleotides.
  • the fragment, as applied to a nucleic acid molecule refers to a subsequence of a larger nucleic acid.
  • a "fragment" of a nucleic acid molecule may be at least about 15 nucleotides in length; for example, at least about 50 nucleotides to about 100 nucleotides; at least about 100 to about 500 nucleotides, at least about 500 to about 1000 nucleotides, at least about 1000 nucleotides to about 1500 nucleotides; or about 1500 nucleotides to about 2500 nucleotides; or about 2500 nucleotides (and any integer value in between).
  • the term “functional fragment” refers to a truncated peptide or polypeptide of the parent that retains at least one biological, physiological, and/or pharmacological property of the parent.
  • the term “inhibit” is meant to refer to a decrease in a biological state.
  • the term “inhibit” may be construed to refer to the ability to negatively regulate expression, stability or activity of an expression product, wherein such inhibition may affect expression of a gene, protein mRNA, stability of a protein mRNA, translation of a protein mRNA, stability of a protein, a protein post- translational modifications, and/or a protein activity.
  • inhibitory nucleic acid refers to small RNAs that inhibit gene expression.
  • inhibitory nucleic acids include, but are not limited to, microRNAs (miRNA) and siRNA.
  • the instructional material includes a publication, a recording, a diagram, or any other medium of expression that may be used to communicate the usefulness of the compounds of the invention.
  • the instructional material may be part of a kit useful for effecting alleviating or treating the various diseases or disorders recited herein.
  • the instructional material may describe one or more methods of alleviating the diseases or disorders in a cell or a tissue of a mammal.
  • the instructional material of the kit may, for example, be affixed to a container that contains the compounds of the invention or be shipped together with a container that contains the compounds. Alternatively, the instructional material may be shipped separately from the container with the intention that the recipient uses the instructional material and the compound cooperatively.
  • the instructional material is for use of a kit; instructions for use of the compound; or instructions for use of a formulation of the compound.
  • isolated refers to material that is free to varying degrees from components which normally accompany it as found in its native state.
  • Isolate denotes a degree of separation from original source or surroundings.
  • Purify denotes a degree of separation that is higher than isolation.
  • a “purified” or “biologically pure” protein is sufficiently free of other materials such that any impurities do not materially affect the biological properties of the protein or cause other adverse consequences. That is, a nucleic acid or peptide is purified if it is substantially free of cellular material, viral material, or culture medium when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • Purity and homogeneity are typically determined using analytical chemistry techniques, for example, polyacrylamide gel electrophoresis or high performance liquid chromatography.
  • the term "purified" can denote that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel.
  • modifications for example, phosphorylation or glycosylation, different modifications may give rise to different isolated proteins, which can be separately purified.
  • microRNA or "miRNA” or “miR” is meant a small non-coding RNA, which functions in transcriptional and/or post-transcriptional regulation of gene expression.
  • neutralizing antibody refers to an antibody that binds an antigen and prevents the biological effects of the antigen.
  • the neutralizing antibody binds TGFP or another effector in the TGFP signaling pathway.
  • “Pharmaceutically acceptable” refers to those properties and/or substances that are acceptable to the patient from a pharmacological/toxicological point of view and to the manufacturing pharmaceutical chemist from a physical/chemical point of view regarding composition, formulation, stability, patient acceptance and bioavailability.
  • “Pharmaceutically acceptable carrier” refers to a medium that does not interfere with the effectiveness of the biological activity of the active ingredient(s) and is not toxic to the host to which it is administered.
  • composition or “pharmaceuticaly acceptable composition” refers to a mixture of at least one compound or molecule useful within the invention with a pharmaceutically acceptable carrier.
  • composition facilitates administration of the compound or molecule to a patient.
  • Multiple techniques of administering a compound or molecule exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
  • the term "pharmaceutically acceptable carrier” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound or molecule useful within the invention within or to the patient such that it may perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound or molecule useful within the invention within or to the patient such that it may perform its intended function.
  • Such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound useful within the invention, and not injurious to the patient.
  • materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate;
  • powdered tragacanth malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations.
  • oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil
  • glycols such as
  • pharmaceutically acceptable carrier also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound useful within the invention, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions.
  • the "pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound or molecule useful within the invention.
  • Other additional ingredients that may be included in the pharmaceutical compositions used in the practice of the invention are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • nucleic acid as used herein is defined as a chain of nucleotides.
  • nucleic acids are polymers of nucleotides.
  • nucleic acids and polynucleotides as used herein are interchangeable.
  • nucleic acids are polynucleotides, which may be hydrolyzed into the monomeric “nucleotides.” The monomeric nucleotides may be hydrolyzed into nucleosides.
  • polynucleotides include, but are not limited to, all nucleic acid sequences that are obtained by any means available in the art, including, without limitation, recombinant means, i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCRTM, and the like, and by synthetic means.
  • recombinant means i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCRTM, and the like, and by synthetic means.
  • the following abbreviations for the commonly occurring nucleic acid bases are used. "A” refers to adenosine, "C” refers to cytosine, “G” refers to guanosine, "T” refers to thymidine, and “U” refers to uridine.
  • RNA as used herein is defined as ribon
  • nucleotide sequence encoding an amino acid sequence includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence.
  • the phrase nucleotide sequence that encodes a protein or an RNA may also include introns to the extent that the nucleotide sequence encoding the protein may in some version contain an intron(s).
  • isolated polynucleotide is meant a nucleic acid (e.g., a DNA) that is free of the genes which, in the naturally-occurring genome of the organism from which the nucleic acid molecule of the invention is derived, flank the gene.
  • the term therefore includes, for example, a recombinant DNA that is incorporated into a vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote; or that exists as a separate molecule (for example, a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences.
  • the term includes an RNA molecule that is transcribed from a DNA molecule, as well as a recombinant DNA that is part of a hybrid gene encoding additional polypeptide sequence.
  • the terms "prevent,” “preventing,” “prevention,” and the like refer to reducing the probability of developing a disorder or condition in a subject, who does not have, but is at risk of or susceptible to developing a disorder or condition.
  • reference is meant a standard or control.
  • a “reference” is a defined standard or control used as a basis for comparison.
  • sample refers to anything, which may contain the cells of interest (e.g., cancer or tumor cells thereof) for which the screening method or treatment is desired.
  • the sample may be a biological sample, such as a biological fluid or a biological tissue.
  • a biological sample is a tissue sample including pulmonary arterial endothelial cells.
  • Such a sample may include diverse cells, proteins, and genetic material.
  • biological tissues also include organs, tumors, lymph nodes, arteries and individual cell(s).
  • biological fluids include urine, blood, plasma, serum, saliva, semen, stool, sputum, cerebral spinal fluid, tears, mucus, amniotic fluid or the like.
  • sensitivity refers to the ability of a target to respond in a qualitative fashion to a pharmacologic action of a compound or agent.
  • small molecule inhibitor refers to a compound or agent that inhibits a target.
  • the small molecule inhibitor inhibits a target in the TGFP signaling pathway.
  • Examples of the small molecule inhibitor include, but are not limited to, SB431542, A83-01, RepSox, SB208, SB505124, LY364947, LY2157299 R268712, D4476, SB525334, GW788388 and TEW-7197.
  • small interfering RNA or “siRNA” is meant a short RNA molecule that may be double stranded, which interferes with the expression of a specific gene that includes a nucleotide sequence complementary to the RNA molecule.
  • specifically binds an antibody or antibody fragment which recognizes and binds with a specific antigen, but does not substantially recognize or bind other molecules in a sample.
  • an antibody that specifically binds to an antigen from one species may also bind to that antigen from one or more species. But, such cross-species reactivity does not itself alter the classification of an antibody as specific.
  • an antibody that specifically binds to an antigen may also bind to different allelic forms of the antigen. However, such cross reactivity does not itself alter the classification of an antibody as specific.
  • the terms “specific binding” or “specifically binding,” can be used in reference to the interaction of an antibody, a protein, or a peptide with a second chemical species, to mean that the interaction is dependent upon the presence of a particular structure (e.g., an antigenic determinant or epitope) on the chemical species; for example, an antibody recognizes and binds to a specific protein structure rather than to proteins generally. If an antibody is specific for epitope "A”, the presence of a molecule containing epitope A (or free, unlabeled A), in a reaction containing labeled "A” and the antibody, will reduce the amount of labeled A bound to the antibody.
  • a particular structure e.g., an antigenic determinant or epitope
  • a “subject” or “patient,” as used therein, may be a human or non-human mammal.
  • Non-human mammals include, for example, livestock and pets, such as ovine, bovine, porcine, canine, feline and murine mammals.
  • the subject is human.
  • substantially identical is meant a polypeptide or nucleic acid molecule exhibiting at least 50% identity to a reference amino acid sequence (for example, any one of the amino acid sequences described herein) or nucleic acid sequence (for example, any one of the nucleic acid sequences described herein).
  • a reference amino acid sequence for example, any one of the amino acid sequences described herein
  • nucleic acid sequence for example, any one of the nucleic acid sequences described herein.
  • such a sequence is at least 60%, more preferably 80%> or 85%>, and more preferably 90%, 95% or even 99% identical at the amino acid level or nucleic acid to the sequence used for comparison.
  • TGFP pathway inhibitor refers to an agent that inhibits the TGFP signaling pathway.
  • a TGFP pathway inhibitor include, but are not limted to, a small molecule, an inhibitory nucleic acid, neutralizing antibody, and an antagonist.
  • treat refers to reducing or improving a disorder and/or symptom associated therewith. It will be appreciated that, although not precluded, treating a disorder or condition does not require that the disorder, condition or symptoms associated therewith be completely ameliorated or eliminated.
  • tripartitie motif-containing 33 or “TRHVI33” refer to a transcriptional corepressor protein and tumor suppressor with three zinc binding domains, a RING, a B-box type 1, a B-box type 2 and a coiled-coil region.
  • TREVI33 is also known as PTC7, RFG7, TFIG, TIFIG, FLJ32925, TIFGAMMA, ECTODERMIN or TIF 1 GAMMA.
  • Three alternatively spliced transcript variants for this gene have been described; however, the full-length nature of one variant has not been determined.
  • TRFM33 regulates TGF -beta/BMP signaling cascade and promotes physiological differentiation of hematopoietic stem cells by associating with SMAD2 and SMAD3.
  • TRFM33 also acts as an E3 ubiquitin-protein ligase to promote SMAD4 ubiquitination, nuclear exclusion and degradation via the ubiquitin proteasome pathway.
  • An exemplary embodiment includes TRFM33 nucleic acid sequence comprising GenBank Accession No. NM_015906 or NM_033020 for human TRFM33 or NM_001079830 or NM 053170 for mouse TRFM33.
  • TRFM33 polypeptide sequence comprising GenBank Accession No. NP 056990.3 or
  • a “vector” is a composition of matter that comprises an isolated nucleic acid and that may be used to deliver the isolated nucleic acid to the interior of a cell.
  • vector includes an autonomously replicating plasmid or a virus.
  • the term should also be construed to include non-plasmid and non- viral compounds which facilitate transfer of nucleic acid into cells, such as, for example, polylysine compounds, liposomes, and the like.
  • viral vectors include, but are not limited to, adenoviral vectors, adeno-associated virus vectors, retroviral vectors, and the like.
  • “Expression vector” refers to a vector comprising a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed.
  • An expression vector comprises sufficient cis-acting elements for expression; other elements for expression may be supplied by the host cell or in an in vitro expression system.
  • Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses) that incorporate the recombinant polynucleotide.
  • Ranges provided herein are understood to be shorthand for all of the values within the range.
  • a range of 1 to 50 is understood to include any number, combination of numbers, or sub-range from the group consisting 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50.
  • compositions or methods provided herein can be combined with one or more of any of the other compositions and methods provided herein.
  • extraterminal domain protein inhibitors limits long-term responsiveness to BETi treatment. It has also been discovered that loss of TRTM33 or over-activation of a TGFP signaling pathway decreases sensitivity to BET inhibitors. As described herein, increased expression of TREVI33 or inhibition of a TGFP signaling pathway increases anti-tumor sensitivity.
  • the invention therefore includes compositions and methods to increase anti-tumor sensitivity in a cell or tumor.
  • BETi are small molecules that interact with BET family bromodomains and displace the BET proteins, such as BRD2, BRD3, BRD4 and BRDT, from binding to chromatin. BETi demonstrate anti-tumor activity, such as antiproliferative effects, to disrupt oncogenic pathways.
  • BETi examples include, but are not limited to, JQl, GS-626510, GS-5829, BMS 986158, RVX2135, CPI203, CPI-0610, ABBV-075, BAY1238097, INCB054329, FT-1101, PFI-1, 1-BET151, ZEN-3365, 1-BET762, OTX015, and TEN-010.
  • the BETi is selected from the group consisting of JQl, GS-626510, GS-5829, BMS 986158, RVX2135, CPI203, CPI-0610, ABBV-075, BAY1238097, INCB054329, FT-1101, PFI-1, 1-BET151, ZEN-3365, 1- BET762, OTX015, TEN-010, and any combination thereof.
  • the present invention includes a composition comprising a TGFP pathway inhibitor and a bromodomain and extraterminal domain protein inhibitor (BETi), wherein the TGFP pathway inhibitor sensitizes cells to the BETi.
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the TGFP pathway inhibitor inhibits at least one TGFP signaling pathway, such as TGFP receptor signaling.
  • the TGFP pathway inhibitor is selected from the group consisting of a small molecule inhibitor, an inhibitory nucleic acid, a neutralizing antibody, and an antagonist.
  • the TGFP pathway inhibitor is a small molecule inhibitor.
  • the small molecule inhibitor is a compound or agent that inhibits TGFP signaling pathway, such as inhibiting TGFP receptor signaling.
  • the small molecule inhibitor is selected from the group consisting of SB431542, A83-01, RepSox, SB208, SB505124, LY364947, LY2157299 R268712, D4476, SB525334, GW788388, TEW-7197, and any combination thereof.
  • the TGFP pathway inhibitor is an inhibitory nucleic acid.
  • the inhibitory nucleic acid includes small RNAs that inhibit gene expression.
  • the inhibitory nucleic acid inhibits expression of at least one gene in a TGFP signaling pathway, such as TGFP or a TGFP receptor.
  • the inhibitory nucleic acid is selected from the group consisting of a miRNA and a siRNA.
  • the siRNA inhibits expression of TGFP or a TGFP receptor.
  • the TGFP pathway inhibitor is a neutralizing antibody.
  • the neutralizing antibody specifically binds TGFP or another effector in the TGFP signaling pathway. In some embodiments, the neutralizing antibody specifically binds TGFP or a TGFP receptor. In one embodiment, the neutralizing antibody specifically binds one selected from the group consisting of TGFP and a TGFP receptor.
  • the neutralizing antibody is selected from the group consisting of ab66043, ab61213, ab31013, 341-BR, 241-R2, AF-241-NA, AF1003, 1600-R2, AF532, MAB1835, MAB240, MAB2411, MAB532, 3C11, V, D-12, 2E5, 500-M66, TB21, H- 100, T-19, V-22, G-16, R-20, RM— 10-3A11, C-4, D-2, E-6, 1-20, L-21, S-20, T-20, H- 567, C-16, A-4, C-20, H-280, F-20, ABF17, H-112, 3711, 56E4, LY3022859, and LY238770.
  • the TGFP pathway inhibitor is a TGFP pathway antagonist.
  • the TGFP pathway antagonist has affinity for a TGFP pathway target, such as a TGFp receptor, and binding to the TGFp pathway target disrupts or prevents the interaction between the TGFP pathway target and its cognate binding partner to inhibit or decrease activity of the TGFp pathway target.
  • the antagonist includes agents that bind TGFP and prevent TGFP from binding to a TGFP receptor, such as blocking (neutralizing) antibodies specific for a TGFP (NAbs) or TGFP receptor (Types I, II or III) such as those described by Dasch et al. (J. Immunol. (1989) 142: 1536) and Lucas et al. (J.
  • TGFp receptors soluble TGFp receptors
  • protease inhibitors that inactivate a protease responsible for activating a precursor TGFP into mature TGFP
  • combinations thereof include monoclonal and polyclonal antibodies directed against one or more isoforms of TGFp (U.S. Pat. No. 5,571,714 and PCT patent application WO 97/13844), TGFp receptors, fragments thereof, derivatives thereof and antibodies directed against TGFp receptors (U.S. Pat. Nos.
  • WO 91/04748, WO 91/10727, WO 93/09800 and WO 94/10187 Further examples of such antagonists include follistatin, somatostatin (PCT patent application WO 98/08529), mannose-6-phosphate or mannose-1 -phosphate (U.S. Pat. No. 5,520,926), prolactin (PCT patent application WO 97/40848), insulin-like growth factor II (PCT patent application WO 98/17304), IP-10 (PCT patent application WO97/00691), arg-gly-asp containing peptides (U.S. Pat. No.
  • the antagonist is selected from the group consisting of HtrAl, decorin, biglycan, fibromodulin, lumican, endoglin, somatostatin, follistatin, RAP-1332, pirfenidone (5-methyl-l-phenyl-2(lH)- pyridone), soluble ectodomains of TGFp receptor type II (RII) or betaglycan (BG), and any combination thereof.
  • the present invention includes a composition comprising a BETi and at least one selected from the group consisting of TRFM33 or a fragment thereof, and a nucleic acid encoding TRFM33, wherein the TGFP pathway inhibitor sensitizes cells to the BETi.
  • the composition of the present invention comprises TRFM33.
  • TRFM33 includes a polypeptide that is recombinantly or synthetically produced.
  • TRFM33 comprises an isolated TRFM33 polypeptide.
  • TRFM33 comprises a polypeptide having a GenBank
  • TRFM33 includes at least one post-translational modification.
  • the composition of the present invention comprises a functional fragment of TRFM33.
  • the functional fragment of TRFM33 includes fragments that retain at least one biological, physiological, and/or pharmacological property of TRFM33.
  • the functional fragment of TRFM33 comprises about 10%, 25%, 30%, 50%, 68%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or any percent therebetween of TRFM33.
  • the composition of the present invention comprises a nucleic acid encoding TRFM33.
  • the nucleic acid encoding TRFM33 comprises a nucleic acid sequence having a GenBank Accession No selected from the group consisting M_015906, M_033020, NM_001079830, and
  • the nucleic acid encoding TRXM33 is an expression vector comprising a TRIM33 gene.
  • the vector is a viral vector, such as an adenoviral vector, an adeno-associated virus vector, a retroviral vector, a lentiviral vector, and the like.
  • the present invention also includes a method for increasing expression of TRIM33 or inhibiting a TGFP signaling pathway to increase anti -turn or sensitivity.
  • loss of TREVI33 or over-activation of a TGFP signaling pathway decreases sensitivity to BET inhibitors.
  • Administering a composition that includes a TREVI33 or a TGFP pathway inhibitor with a BETi to a subject in need thereof increases anti -tumor sensitivity in a cell or tumor in the subject.
  • the invention includes a method of increasing anti -tumor sensitivity to a bromodomain and extraterminal domain protein inhibitor (BETi) in a cell in a subject.
  • the method comprising administering a composition comprising a TGFP pathway inhibitor and a BETi to a subject in need thereof, wherein the TGFP pathway inhibitor sensitizes the cell to the BETi.
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the invention includes a method of treating a tumor comprising administering to a subject in need thereof a composition comprising a TGFP pathway inhibitor and a bromodomain and extraterminal domain protein inhibitor (BETi), wherein the TGFP pathway inhibitor sensitizes the tumor to the BETi.
  • a composition comprising a TGFP pathway inhibitor and a bromodomain and extraterminal domain protein inhibitor (BETi), wherein the TGFP pathway inhibitor sensitizes the tumor to the BETi.
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the invention includes a method of increasing anti-tumor sensitivity of a cell in a subject to a bromodomain and extraterminal domain protein inhibitor (BETi).
  • BETi bromodomain and extraterminal domain protein inhibitor
  • the method comprising administering a composition comprising a BETi and one selected from the group consisting of TREVI33 or a fragment thereof, and a nucleic acid encoding TREVI33 to a subject in need thereof, wherein the TREVI33 sensitizes the cell to the BETi.
  • the invention includes a method of treating a tumor comprising administering to a subject in need thereof a composition comprising a BETi and at least one selected from the group consisting of TREVI33 or a fragment thereof, and a nucleic acid encoding TREVI33, wherein TREVI33 sensitizes the tumor to the BETi.
  • the tumor is a cancer selected from the group consisting of breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, and lung cancer.
  • the subject has a cancer, such as a cancer selected from the group consisting of breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, and lung cancer.
  • a cancer such as a cancer selected from the group consisting of breast cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, pancreatic cancer, colorectal cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia, and lung cancer.
  • compositions of the present invention are useful to increase an anti-tumor response in a cellular target to a BETi by increasing a basal level of the anti- tumor response or resensitizing the cellular target to the BETi.
  • the cell or tumor is BETi resistant.
  • cell or tumor has acquired decreased BETi sensitivity.
  • administration of the composition to the subject increases an anti-tumor response to the BETi.
  • the present invention includes, in some embodiments, a composition comprising a TGFP pathway inhibitory nucleic acid, and in some embodiments, a composition comprising a nucleic acid encoding TREVI33.
  • Such nucleic acids are introduced into a cell for the benefit of a subject.
  • Methods of introducing nucleic acids into a cell include physical, biological and chemical methods.
  • Physical methods for introducing a polynucleotide, such as DNA like cDNA, into a host cell include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like.
  • DNA, like cDNA can be introduced into target cells using commercially available methods which include electroporation (Amaxa Nucleofector-II (Amaxa Biosystems, Cologne, Germany)),
  • DNA can also be introduced into cells using cationic liposome mediated transfection using lipofection, using polymer encapsulation, using peptide mediated transfection, or using biolistic particle delivery systems such as "gene guns” (see, for example, Nishikawa, et al. Hum Gene Ther., 12(8):861-70 (2001)).
  • Biological methods for introducing a polynucleotide of interest into a host cell include the use of DNA and RNA vectors.
  • Viral vectors, and especially retroviral vectors have become the most widely used method for inserting genes into mammalian, e.g., human cells.
  • Other viral vectors can be derived from lentivirus, poxviruses, herpes simplex virus I, adenoviruses and adeno-associated viruses, and the like. See, for example, U.S. Pat. Nos. 5,350,674 and 5,585,362.
  • Chemical means for introducing a polynucleotide into a host cell include colloidal dispersion systems, such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • colloidal dispersion systems such as macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • An exemplary colloidal system for use as a delivery vehicle in vitro and in vivo is a liposome (e.g., an artificial membrane vesicle).
  • Lipids suitable for use can be obtained from commercial sources.
  • DMPC dimyristyl phosphatidylcholine
  • DCP dicetyl phosphate
  • Choi cholesterol
  • DMPG phosphatidylglycerol
  • Avanti Polar Lipids, Inc. (Birmingham, AL).
  • Liposome is a generic term encompassing a variety of single and multilamellar lipid vehicles formed by the generation of enclosed lipid bilayers or aggregates. Liposomes can be characterized as having vesicular structures with a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution.
  • the lipids may assume a micellar structure or merely exist as nonuniform aggregates of lipid molecules.
  • lipofectamine- nucleic acid complexes are also contemplated.
  • assays include, for example, "molecular biological” assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR; "biochemical” assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention.
  • molecular biological assays well known to those of skill in the art, such as Southern and Northern blotting, RT-PCR and PCR
  • biochemical assays, such as detecting the presence or absence of a particular peptide, e.g., by immunological means (ELISAs and Western blots) or by assays described herein to identify agents falling within the scope of the invention.
  • the invention also encompasses the use of a pharmaceutical composition of the invention to practice the methods of the invention.
  • the invention includes a pharmaceutical composition comprising the composition as described herein and a pharmaceutically acceptable carrier.
  • the composition described herein is used in the manufacture of a medicament for the treatment of a tumor or cancer in a subject in need thereof.
  • the invention includes a pharmaceutical composition comprising the composition as described herein in combination with another therapeutic agent used in the treatment of a tumor or cancer.
  • Such pharmaceutical compositions may be provided in a form suitable for
  • composition described herein may comprise a physiologically acceptable salt, such as a compound contemplated within the invention in combination with a physiologically acceptable cation or anion, as is well known in the art.
  • compositions that are useful in the methods of the invention may be suitably developed for inhalational, oral, rectal, vaginal, parenteral, topical, transdermal, pulmonary, intranasal, buccal, ophthalmic, intrathecal, intravenous or another route of administration.
  • Other contemplated formulations include projected nanoparticles, liposomal preparations, resealed erythrocytes containing the active ingredient, and immunologically-based formulations.
  • the route(s) of administration will be readily apparent to the skilled artisan and will depend upon any number of factors including the type and severity of the disease being treated, the type and age of the veterinary or human patient being treated, and the like.
  • compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology.
  • preparatory methods include the step of bringing the active ingredient into association with a carrier or one or more other accessory ingredients, and then, if necessary or desirable, shaping or packaging the product into a desired single- or multi- dose unit.
  • compositions of the invention are formulated using one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions of the invention comprise a therapeutically effective amount of at least one compound of the invention and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include, but are not limited to, glycerol, water, saline, ethanol and other pharmaceutically acceptable salt solutions such as phosphates and salts of organic acids. Examples of these and other pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1991, Mack Publication Co., New Jersey).
  • BETi Bromodomain and extraterminal domain protein inhibitors
  • TREVI33 silencing attenuates downregulation of MYC in response to BETi. Moreover, loss of TREVI33 enhances TGFP receptor expression and signaling, and blocking TGFp receptor activity potentiates the anti-proliferative effect of BETi. Described herein is a mechanism for BETi resistance and combining inhibition of TGFP signaling with BET bromodomain inhibition may offer new therapeutic benefits.
  • Cell lines, antibodies and drugs Cell lines 293 T, RKO, HCT 15, HCT 116,
  • Lentiviral expression vectors for shRNAs in the pLKO.1 puro vector were used to stably knockdown TRFM33, TpRII or SMAD4.
  • the shTRFM33-B5 sequence was cloned into pLKO. l blast (Addgene #26655) to silence TRFM33 expression.
  • the shRNA target sequences used are listed in Table 1.
  • cDNAs from Addgene #19147 and Addgene #15734 respectively were cloned into pLentiCMV-hygro(DEST) (Addgene #17454)) through Gateway cloning (Invitrogen). Seven silent mutations were made to TRIM33 cDNA to render resistance to shTREVI33-B5.
  • MYC lentiviral expression vector is from Addgene (#46970).
  • Table 1 shRNA target sequences.
  • shRNA Target sequence shCTRL C AAC AAGATGAAGAGC AC C AA SEQ ID NO: l shTRIM33-B5 GTACTAGTTGTGAAGACAATG SEQ ID NO:2 shTRIM33-A12 GCTCCTGGTTATACTCCTAAT SEQ ID NO: 3 shT RII-3 GCTCCCTAAACACTACCAAAT SEQ ID N0:4 shT RII-4 AATGACGAGAACATAACACTC SEQ ID NO: 5 shSMAD4-3 CAGATTGTCTTGCAACTTCAG SEQ ID NO:6 shSMAD4-4 TACCATACAGAGAACATTGGA SEQ ID NO:7
  • TGFBR2 GTAGCTCTGATGAGTGCAATGAC CAGATATGGCAACTCCCAGTG
  • HBE1 ATGGTGCATTTTACTGCTGAGG GGGAGACGACAGGTTTCCAAA
  • BCL2A1 TACAGGCTGGCTCAGGACTAT CGCAACATTTTGTAGCACTCTG
  • MAP1LC3C CCCAAGCGTCAGACCCTTC GGGGAACTTTGCCCGGATT
  • SEQ ID NO:46 SEQ ID NO:47 Cumulative cell growth assay.
  • RKO cells (3 x 10 5 ) transduced with the indicated virus were plated in a single well of a 6-well plate at day 0 in the presence or absence of inhibitors. Three days later cells were detached, counted, and 3 x 10 5 cells were transferred to a new well. The process was repeated until day 15. The cumulative cell number was then calculated from fold changes and the individual cell counts at each passage.
  • Crystal violet cell proliferation assay Cells (5-20 x 10 3 ) were plated in each well of a 6-well plate with 3 mL of media with or without inhibitors and cultured for 14 days undisturbed. Medium was aspirated, and cells were stained with crystal violet staining solution (0.05% w/v crystal violet, 1% formaldehyde, 1% methanol in PBS) for 30 minutes and washed with water several times. Stained plates were then air-dried and imaged with CHEMIDOC® using Image Lab software (Bio-Rad). To quantify the crystal violet staining, 1 mL of 10% acetic acid was added to each well to solubilize the stain for 20 min and the stain was diluted 1 :4 in water and absorbance was measured at 590 nm.
  • crystal violet staining solution 0.05% w/v crystal violet, 1% formaldehyde, 1% methanol in PBS
  • GRAPHPAD PRISM 6® by fitting the data to the "3 -parameter log (inhibitor) vs response" equation. At least three independent growth inhibition assays were performed for each pair of cell lines to derive mean IC 50 values.
  • shRNA screening The Mission human shRNA library (Luo et al., 2008, Proc Natl Acad Sci USA 105(51):20380-20385) generated in pLKO lentiviral delivery vectors by The RNAi Consortium (TRC) was obtained as arrayed bacterial stocks (Sigma). All shRNAs targeting 517 genes annotated as protein kinases (5634 shRNAs in total) and 85 non-targeting control shRNA vectors were picked from the library, cultured on LB-agar plates and plasmid DNA was prepared from a mixture of these cultures using GeneElute HP Endotoxin Free Plasmid Maxi-prep kit (Sigma).
  • Lentiviral particles were generated by co-transfecting 293T cells with the pLKO plasmid mixture, pCMV dR8.91 packaging vector, and pCMV-VSV-G envelope vector in a 10: 10: 1 ratio. Viral supernatant was collected 48 and 72 h after transfection and stored at -80°C. RKO cells were transduced by incubating for 24 h with the pool of shRNA-expressing viruses diluted to give a MOI (multiplicity of infection) of -0.3 to ensure that most of the cells received a single viral integration.
  • MOI multiplicity of infection
  • Infected cells were selected with 1 ⁇ g/mL
  • RNAseq data analysis and gene set enrichment analysis RKO cells expressing shCTRL or shTREVI33 were treated with 0.1% DMSO vehicle, 1 ⁇ JQ1 or 0.3 ⁇ GS-626510 for 3 h, and mRNA was extracted using RNEASY® mini kit with on column DNase I digestion option (Qiagen) and submitted to the Yale Center for Genome Analysis for RNAseq analysis. Low quality reads and bases were trimmed, and filtered reads were then mapped to the human reference genome (hgl9) using TophatTM v2.0.13. Only reads that mapped to a single unique location within the genome were reported. Tophat alignments from duplicate RNAseq experiments were then processed through DESeq to produce one differential expression data set.
  • edgeR was used to normalize the gene level read counts across samples. Genes with less than one shortread count per million (CPM) in at least one sample were filtered out to remove genes with low levels of expression. Generalized linear regression in edgeR was then used to estimate log 2 fold changes and p values. To identify the genes that respond differently to BETi in the shTRXM33 cells relative to the shCTRL cells, the following contrast was specified in the edgeR analysis: (BETi in shTRIM33 - DMSO in shTRIM33) - (BETi in shCTRL - DMSO in shCTRL).
  • Example 1 Pooled shRNA library screening identified TRIM33 as a negative regulator of BETi resistance
  • a pooled shRNA screen was performed in a BETi- sensitive colorectal cancer cell line (RKO). Screening was carried out in the presence of one of two structurally unrelated inhibitors: the widely used compound JQl and a novel BETi GS-626510 (Figure 1A). GS-626510 binds with high affinity and specificity to BET family bromodomains ( Figure IB, Table 3). Both JQl and GS-626510 potently inhibited growth of RKO cells with IC 50 values of 81 nM and 33 nM respectively ( Figure 1C).
  • RNAseq analysis showed a strong correlation between genes up- and down-regulated following 3 h treatment of RKO cells with 1 ⁇ of JQl or 0.3 ⁇ of GS-626510 ( Figure IE), suggesting that growth suppression by these compounds is attributable to a common mechanism of action.
  • Table 3 Kd values of bromodomains to BETi GS-626510
  • a custom lentiviral shRNA library was generated containing 5634 shRNA constructs targeting 517 genes annotated as protein kinases and 85 non-targeting control shRNAs.
  • RKO cells were infected with the pooled shRNA virus, and following puromycin selection for infected cells, 6 x 10 6 cells were removed for genomic DNA extraction to serve as a reference (TO) population. The remaining cells were placed into each of 5 different inhibitor conditions: DMSO vehicle control and low and high doses of either JQ1 or GS-626510 (Figure IF). Cells were allowed to proliferate and were passaged when they approached confluence. This treatment was maintained until cells reached passage 4 (T4).
  • RIGER analysis was used to identify and rank genes preferentially targeted by hairpins enriched upon drug treatment but not in the DMSO-treated control cells. These genes presumably encode proteins that promote susceptibility to BETi. Silencing expression of these genes thus causes drug resistance, resulting in cells harboring their respective hairpins being enriched at the end of the screen. Strikingly, TRIM33 was the top ranked enriched target gene in all four BETi- treated conditions, but was not enriched in the absence of inhibitor (Figure 1G).
  • TRJJVI33 hairpins appear to be preferentially depleted in the DMSO vehicle control sample.
  • TRJJVI24 the most closely related TRJJVI33 family member, was also highly enriched in all four inhibitor treated conditions but not in the DMSO control ( Figure 1G), supporting the potential functional relevance of TRJJVI33 to modulate BETi sensitivity.
  • TRJJVI33 knockdown conferred a selective growth advantage in BETi-treated RKO cells.
  • TRJJVI33 and TRJJVI24 were included in the shRNA library on the basis of early reports identifying TRJJVI24 and TREVI28 as protein kinases, but the absence of a recognizable kinase catalytic domain and lack of subsequent verification suggests that these proteins are unlikely to have such activity.
  • Example 2 BETi resistance in shTRIM33 cells was due to the specific loss of TRIM33 protein
  • TRJJVI33 knockdown produced a rightward shift in the growth inhibition curves for both JQl and GS-626510 (Figure 2C).
  • Multiple replicates revealed that the IC 50 value of JQl and GS-626510 was increased by approximately 3-fold in shTRJJVI33 cells, suggesting the shTRJJVI33 cells were more resistant to BETi ( Figure 2D).
  • This effect was not limited to RKO cells as similar experiments performed in a panel of colorectal, breast and prostate cancer cell lines revealed that TRJJVI33 knockdown also decreased sensitivity to JQl and GS-626510 in a subset of the cell lines tested ( Figures 2E and Table 4).
  • TRJJVI33 knockdown facilitated outgrowth of BETi-treated RKO cells ( Figure 2F). Similar effects were observed with a different shRNA targeting TRJJVI33 (A 12) ( Figures 2 A and 2G), suggesting that the results were not due to off target effects.
  • MDA-MB-231 0.114 ⁇ 0.019 0.267 ⁇ 0.025 0.085 ⁇ 0.022 0.216 ⁇ 0.024
  • RNAseq was used to investigate changes in gene expression resulting from treatment with BETi and with loss of TREVI33. RNAseq was performed in shCTRL and shTRIM33 cells after 3h treatment with JQ1 (1 ⁇ ), GS- 626510 (0.3 ⁇ ) or vehicle control (DMSO). Results from two independent replicate experiments were analyzed by DESeq. Results consistent with RNAseq data were obtained by measuring mRNA levels for 15 genes by qRT-PCR ( Figures 3A-3E).
  • TRDVI33 KD In contrast to BET bromodomain inhibition, TRDVI33 KD influenced the expression of a relatively small fraction of genes (Figure 31). Following TRPM33 knockdown, 272 transcripts were up-regulated by at least 2-fold, while only 84 were down-regulated by at least 2-fold, arguing that TRDVI33 works preferentially as a transcriptional repressor rather than an activator. Notably, loss of TRPM33 had no effect on expression of BET genes (BRD2, BRD3 and BRD4) themselves and did not affect BRD4 protein levels (Figure 3 J).
  • MYC Repression of MYC is believed to be a major mechanism by which BETi suppress growth of some cell types (10, 12). It was therefore examined whether there was a potential role for MYC in mediating the effect of TRFM33 knockdown.
  • MYC was stably over-expressed in RKO cells. Ectopically expressed MYC was resistant to BETi-mediated down regulation (Figure 4D). While RKO cells overexpressing MYC proliferated at the same rate as control cells, possibly reflecting the high basal levels of MYC expression in this cell line, MYC over- expressing cells had a growth advantage in long-term culture in the presence of JQl or GS-626510 ( Figures 4E-4F). Thus, protection of MYC levels from downregulation is likely to contribute to BETi resistance in shTRFM33 RKO cells.
  • chromatin immunoprecipitation revealed that TRFM33 associated with the MYC promoter in BETi-treated RKO cells ( Figure 4G).
  • BRD4 ChIP showed that BRD4 associated with similar sites in the MYC promoter and that BRD4 was displaced following BETi treatment ( Figure 4H).
  • Example 4 TRIM33 knockdown potentiates TGFp signaling and inhibition of TGFp pathway increased BETi sensitivity While the efficacy of BETi has been linked to down-regulation of MYC expression in hematopoietic cancers and a subset of solid tumors, in other tumor cells BETi-mediated growth suppression is independent of MYC. Notably, in contrast to what was observed in RKO cells, MYC levels in another colorectal cancer cell line, SK-CO-1, were much less sensitive to either BETi treatment or TRFM33 knockdown (Figure 5A). Nonetheless, in this cell line TRFM33 knockdown conferred resistance to BETi ( Figure 2E and Table 4). This observation suggests that other pathways in addition to MYC signaling can contribute to shTRFM33 cell resistance to BETi.
  • GSEA Gene set enrichment analysis
  • TRFM33 has been implicated as a regulator of TGFp signaling. Furthermore, as TGFp signaling can promote resistance to other targeted therapies, it was investigated how the pathway was altered in shTRFM33 RKO cells.
  • Canonical TGFP signaling involves TGFP ligand-induced formation of heterotetramers containing dimers of the TGFP receptor I (TpRI) and TGFP receptor II (TpRII) serine-threonine kinases.
  • Receptor clustering promotes TpRII phosphorylation of TpRI, leading to recruitment and phosphorylation of regulatory SMADs (SMAD2/3) by TpRI.
  • Phosphorylated SMAD2/3 then binds to SMAD4 to form a complex that enters the nucleus to drive transcription of target genes.
  • TGFpl-induced phosphorylated SMAD2 (pSMAD2) seen in shTRFM33 cells co-immunoprecipitated with SMAD4, suggesting that the pSMAD2 enters functional complexes with SMAD4 (Figure 5G).
  • TRFM33 antagonizes TGFP signaling by negatively regulating SMAD4 through either mono- ubiquitinating SMAD4 or competing with SMAD4 for phosphorylated SMAD2/3.
  • knockdown of SMAD4 in shTRFM33 cells had no impact on the TGFpi- mediated induction of pSMAD2 (Figure 5H).
  • RNAseq data showed that the TpRII mRNA was upregulated ⁇ 2 fold in shTRIM33 cells (Figure 51). Furthermore, ChIP experiments revealed that TREVI33 association with the TPRII promoter was increased by BETi, while BRD4 association was decreased ( Figures 5J-5K), similar to the manner that MYC is regulated by TREVI33 and BRD4.
  • TGFp pathway inhibitors and BET bromodomain inhibitors may provide a more potent inhibition of cell growth and may provide a means to overcome resistance to BET bromodomain inhibitors.
  • the small molecule TpRI inhibitor LY2157299 (galunisertib) was used.
  • the top hit from the screen was TRFM33, with its close family member,
  • TRFM24 also being identified. These data suggest that loss of TRFM33 confers resistance to BETi, and this was confirmed in both short and long-term growth assays. Mechanistically, loss of TRFM33 reduces BETi-mediated down-regulation of MYC and sensitizes cells to TGFP signaling. Notably, inhibition of TGFP signaling re-sensitizes TRFM33 knockdown cells to BETi, suggesting that combining TGFP inhibitors with BETi may have therapeutic benefit.
  • MYC oncogenic transcription factor
  • JQ1 suppressed growth by downregulating the transcription factor FOSL1 rather than MYC, suggesting that alternative mechanisms may underlie the activity of BETi in solid tumors.
  • a pooled shRNA screen was performed with a library targeting genes annotated as protein kinases. It was found that loss of TRTM33 conferred resistance to either JQ1 or GS-626510 treatment, indicating that TRIM33 is required, in at least some cell types, for cells to be fully sensitive to BETi. In such cells, TREVI33 appears to promote downregulation of MYC by BETi.
  • TREVI33 Classically TREVI33, TREVI24 and TREVI28 act as potent transcriptional co- repressors when recruited to the promoters of target genes, and consistent with this mechanism, it was found that TREVI33 associates with the MYC promoter. Notably this association is enhanced by BETi, possibly due to direct competition between BRD4 and TREVI33 for binding at these sites.
  • Transcriptional modulation of MYC by TREVI33 could involve its E3 ligase activity, for example by triggering ubiquitin-mediated degradation of factors co- associated with promoter or enhancer regions. Attempts were made to test this model using TREVI33 mutants with impaired E3 ligase activity. Mutant TREVI33, while unable to restore JQ1 sensitivity in shTRFM33 cells, was also very poorly expressed, making it unclear whether its ligase activity was essential.
  • up-regulation of the transcription factor GLI2 contributes to acquired BETi-resistance in pancreatic cancer cells (Kumar K, et al. (2015). Sci Rep 5:9489) by driving MYC expression, and in models of acute myeloid leukemia (AML), increased WNT signaling apparently bypasses BET bromodomain-mediated transcription to maintain MYC expression through utilization of a cryptic enhancer region.
  • TRFM33 partially protects MYC levels after BETi treatment, but loss of TRFM33 was not found to affect ⁇ -catenin levels or localization in RKO cells. Furthermore, as judged by RNAseq analysis, TRIM33 knockdown did not induce GLI2 in RKO cells.
  • TRIM33 knockdown apparently confers BETi-resistance at least in part by preventing MYC downregulation
  • the pathways involved are distinct from those previously characterized.
  • BETi function independently of MYC reported mechanisms of resistance likewise appear to involve maintaining expression of BRD4-target genes.
  • triple negative breast cancer cells can acquire BETi- resistance through BRD4 hyperphosphorylation, which drives expression of target genes through interactions with the mediator complex in a manner independent of the acetylated lysine binding pocket of its bromodomains.
  • sparing of critical target genes appears to be an important component of BETi resistance caused by loss of TRFM33.
  • BRD4 A short isoform of BRD4 was recently shown to be an inhibitor of DNA damage response signaling by influencing chromatin structure independently of its role as a transcriptional activator. Resistance to BETi could theoretically arise by reduction of DNA damage signaling, bypassing growth arrest. However, it was found that TRFM33 knockdown did not alter DNA damage signaling as assessed by ⁇ 2 ⁇ staining, suggesting that an alternative resistance pathway must be involved.
  • TRFM33 may therefore act as a direct modulator of TpRII gene transcription.
  • TGFP signaling either by silencing TpRII expression or with a small molecule inhibitor of TpRI, sensitized TRFM33 knockdown cells to BETi.
  • overexpressing TpRII was sufficient to sensitize cells to TGFpi, it did not prevent BETi-mediated suppression of MYC levels or cell growth.
  • inhibition of TGFP signaling was sufficient to sensitize cells to BETi.
  • TGFP-induced resistance to targeted therapies is associated with enhanced signaling through the ERK MAP kinase pathway.

Abstract

La présente invention concerne des compositions et des procédés pour renforcer la sensibilité anti-tumorale d'une cellule ou d'une tumeur à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (BETi). Selon un aspect, une composition comprend un BETi et un inhibiteur de la voie TGFβ, TRIM33 ou un fragment de celui-ci, ou un acide nucléique codant pour TRIM33. Selon un autre aspect, l'invention concerne un procédé permettant de renforcer la sensibilité anti-tumorale aux BETi. L'invention porte également sur des procédés concernant une tumeur et impliquant un BETi.
PCT/US2017/038669 2016-06-22 2017-06-22 Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti) WO2017223268A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662353424P 2016-06-22 2016-06-22
US62/353,424 2016-06-22

Publications (1)

Publication Number Publication Date
WO2017223268A1 true WO2017223268A1 (fr) 2017-12-28

Family

ID=60784636

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/038669 WO2017223268A1 (fr) 2016-06-22 2017-06-22 Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti)

Country Status (1)

Country Link
WO (1) WO2017223268A1 (fr)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019113462A1 (fr) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine et dérivés et leurs utilisations anti-virales
US10633379B2 (en) 2016-04-15 2020-04-28 Abbvie Inc. Bromodomain inhibitors
WO2020169698A1 (fr) * 2019-02-21 2020-08-27 F. Hoffmann-La Roche Ag Sensibilisation de cellules cancéreuses au tnf par inhibition de bet
WO2020192637A1 (fr) * 2019-03-22 2020-10-01 石药集团中奇制药技术(石家庄)有限公司 Composé inhibiteur de brd4 sous forme solide, son procédé de préparation et son application
CN113521074A (zh) * 2020-04-17 2021-10-22 南京圣和药业股份有限公司 一种包含喹啉类TGF-β1抑制剂的组合物及其用途
RU2793346C1 (ru) * 2019-03-22 2023-03-31 СиЭсПиСи ЧЖУНЦИ ФАРМАСЬЮТИКАЛ ТЕКНОЛОДЖИ (ШИЦЗЯЧЖУАН) КО., ЛТД. Соединение ингибитора brd4 в твердой форме, способ его получения и применение
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010808A1 (fr) * 1991-12-04 1993-06-10 La Jolla Cancer Research Foundation INHIBITION DU FACTEUR DE CROISSANCE TRANSFORMATEUR β AFIN DE PREVENIR L'ACCUMULATION DE LA MATRICE EXTRACELLULAIRE
US20150174138A1 (en) * 2012-06-19 2015-06-25 The Broad Institute, Inc. Diagnostic and treatment methods in subjects having or at risk of developing resistance to cancer therapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993010808A1 (fr) * 1991-12-04 1993-06-10 La Jolla Cancer Research Foundation INHIBITION DU FACTEUR DE CROISSANCE TRANSFORMATEUR β AFIN DE PREVENIR L'ACCUMULATION DE LA MATRICE EXTRACELLULAIRE
US20150174138A1 (en) * 2012-06-19 2015-06-25 The Broad Institute, Inc. Diagnostic and treatment methods in subjects having or at risk of developing resistance to cancer therapy

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
FONG ET AL.: "BET Inhibitor Resistance Emerges From Leukaemia Stem Cells", NATURE, vol. 525, 24 September 2015 (2015-09-24), pages 538 - 542, XP055449874 *
HE ET AL.: "Hematopoiesis Controlled by Distinct TIF1g and Smad4 Branches of the TGFb . Pathway", DEVELOPMENTAL CELL, vol. 125, no. 5, 2 June 2006 (2006-06-02), pages 929 - 941, XP055449884 *
LU ET AL.: "Drug Delivery Using Nanoparticles for Cancer Stem-Like Cell Targeting", FRONTIERS IN PHARMACOLOGY, vol. 7, 12 April 2016 (2016-04-12), pages 1 - 12, XP055449872 *
SHI ET AL.: "Loss of TRIM33 Causes Resistance to BET Bromodomain Inhibitors Through MYC *and TGF-beta-Dependent Mechanisms", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 113, no. 113, 18 July 2016 (2016-07-18), pages E4558 - E4566, XP055449887 *
XUE ET AL.: "Tumour Suppressor TRIM33 Targets Nuclear beta-catenin Degradation", NAT. COMMUN, vol. 6, 2 February 2015 (2015-02-02), pages 1 - 33, XP055449878 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11628181B2 (en) 2014-12-26 2023-04-18 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US10633379B2 (en) 2016-04-15 2020-04-28 Abbvie Inc. Bromodomain inhibitors
WO2019113462A1 (fr) 2017-12-07 2019-06-13 Emory University N4-hydroxycytidine et dérivés et leurs utilisations anti-virales
US11331331B2 (en) 2017-12-07 2022-05-17 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
US11903959B2 (en) 2017-12-07 2024-02-20 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2020169698A1 (fr) * 2019-02-21 2020-08-27 F. Hoffmann-La Roche Ag Sensibilisation de cellules cancéreuses au tnf par inhibition de bet
WO2020192637A1 (fr) * 2019-03-22 2020-10-01 石药集团中奇制药技术(石家庄)有限公司 Composé inhibiteur de brd4 sous forme solide, son procédé de préparation et son application
CN113646312A (zh) * 2019-03-22 2021-11-12 石药集团中奇制药技术(石家庄)有限公司 固体形式的brd4抑制剂化合物及其制备方法与应用
CN113646312B (zh) * 2019-03-22 2022-12-06 石药集团中奇制药技术(石家庄)有限公司 固体形式的brd4抑制剂化合物及其制备方法与应用
RU2793346C1 (ru) * 2019-03-22 2023-03-31 СиЭсПиСи ЧЖУНЦИ ФАРМАСЬЮТИКАЛ ТЕКНОЛОДЖИ (ШИЦЗЯЧЖУАН) КО., ЛТД. Соединение ингибитора brd4 в твердой форме, способ его получения и применение
CN113521074A (zh) * 2020-04-17 2021-10-22 南京圣和药业股份有限公司 一种包含喹啉类TGF-β1抑制剂的组合物及其用途

Similar Documents

Publication Publication Date Title
WO2017223268A1 (fr) Compositions et procédés de resensibilisation de cellules à des inhibiteurs des protéines de bromodomaine et de domaine extraterminal (beti)
Kim et al. Polycomb-and methylation-independent roles of EZH2 as a transcription activator
Iniguez et al. EWS/FLI confers tumor cell synthetic lethality to CDK12 inhibition in Ewing sarcoma
Webster et al. W nt5 A promotes an adaptive, senescent‐like stress response, while continuing to drive invasion in melanoma cells
JP2018517660A (ja) 周皮細胞長鎖非コーディングrna
US10617709B2 (en) miRNAs as novel therapeutic adjuvants and biomarkers for the prognosis and treatment of drug resistant breast cancers
Sobral et al. KDM3A/Ets1/MCAM axis promotes growth and metastatic properties in Rhabdomyosarcoma
CA2907184C (fr) Antigene falz destine a etre utilise comme cible pour therapies destinees a traiter le cancer
JP5887413B2 (ja) 非小細胞肺癌におけるtm4sf4の発現または活性を調節することによって癌細胞の放射線耐性ならびに増殖、転移および浸潤を低減させる方法
US11246868B2 (en) Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment
US8686127B2 (en) Apoptosis inducer
US20200123544A1 (en) Gene therapy targeting the neonatal form of nav1.5 for treating cancer
WO2016179222A1 (fr) Znf532 pour le diagnostic et le traitement du cancer
US20200222444A1 (en) Methods of inhibiting cell proliferation and mettl8 activity
JP6839707B2 (ja) Gpr160を過剰発現する癌の予防、診断および治療
WO2012113779A1 (fr) Moyens et procédés pour traiter une maladie ou un trouble relatif à la lymphangiogenèse ou prévenir la métastase
Zhang et al. EZH2/G9a interact to mediate drug resistance in non-small-cell lung cancer by regulating the SMAD4/ERK/c-Myc signaling axis
RU2811918C2 (ru) Способы лечения раковых заболеваний и опухолей с использованием ингибиторов pde1
WO2024003350A1 (fr) Polythérapie pour mélanome
CN116496349A (zh) 治疗肿瘤放疗敏感的多肽化合物
WO2021216415A1 (fr) Compositions et procédés pour augmenter le flux de sodium dans les cellules cardiaques
Sun et al. shRNA drop-out screen identifies BRD4 targeting transcription from RNA polymerase II system to activate β-catenin to promote soft-tissue tumor proliferations
Stevens The Role Of Cpeb2 Alternative Splicing In TNBC Metastasis
Sethuraman Role of SMARCE1 and BHLHE40 in Breast Cancer Metastasis
KR101497930B1 (ko) Hs2st1 유전자 또는 상기 유전자가 암호화하는 단백질에 대한 억제제를 포함하는 세포 노화 유도용 조성물 및 이를 이용한 세포 노화 유도 방법

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: 17816185

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17816185

Country of ref document: EP

Kind code of ref document: A1