US20230285439A1 - Methods for treating triple-negative breast cancer - Google Patents

Methods for treating triple-negative breast cancer Download PDF

Info

Publication number
US20230285439A1
US20230285439A1 US17/823,028 US202217823028A US2023285439A1 US 20230285439 A1 US20230285439 A1 US 20230285439A1 US 202217823028 A US202217823028 A US 202217823028A US 2023285439 A1 US2023285439 A1 US 2023285439A1
Authority
US
United States
Prior art keywords
cdk19
agent
cells
activity
cdk8
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/823,028
Inventor
Michael F. Clarke
Robert W. Hsieh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leland Stanford Junior University
CZ Biohub SF LLC
Original Assignee
Leland Stanford Junior University
CZ Biohub SF LLC
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 Leland Stanford Junior University, CZ Biohub SF LLC filed Critical Leland Stanford Junior University
Priority to US17/823,028 priority Critical patent/US20230285439A1/en
Assigned to THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY reassignment THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CLARK, MICHAEL F., HSIEH, Robert W.
Assigned to THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY reassignment THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY CORRECTIVE ASSIGNMENT TO CORRECT THE LAST NAME OF ASSIGNOR MICHAEL F. CLARKE ON THE COVERSHEET PREVIOUSLY RECORDED AT REEL: 060968 FRAME: 0506. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT. Assignors: CLARKE, MICHAEL F., HSIEH, Robert W.
Assigned to THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY, CHAN ZUCKERBERG BIOHUB, INC. reassignment THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY
Assigned to CZ BIOHUB SF, LLC reassignment CZ BIOHUB SF, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHAN ZUCKERBERG BIOHUB, INC.
Publication of US20230285439A1 publication Critical patent/US20230285439A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • 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
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-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
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11022Cyclin-dependent kinase (2.7.11.22)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/483Physical analysis of biological material
    • G01N33/4833Physical analysis of biological material of solid biological material, e.g. tissue samples, cell cultures
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7023(Hyper)proliferation
    • G01N2800/7028Cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast

Definitions

  • the invention relates to the field of biomedicine, e.g., oncology.
  • TNBC Triple-negative breast cancer
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2-negative invasive breast cancer the so-called triple-negative phenotype: a population-based study from the California cancer Registry” Cancer 109, 1721-1728, doi:10.1002/cncr.22618 (2007).
  • ER estrogen receptor
  • PR progesterone receptor
  • HER2-negative invasive breast cancer the so-called triple-negative phenotype: a population-based study from the California cancer Registry” Cancer 109, 1721-1728, doi:10.1002/cncr.22618 (2007).
  • the invention is directed to methods of treating TNBC in a patient by administering to the patient an agent that inhibits the expression or activity of cyclin-dependent kinase 19 (CDK19).
  • CDK19 cyclin-dependent kinase 19
  • the invention features a method of treating a patient diagnosed with triple-negative breast cancer (TNBC) by administering a therapeutically effective dose of an agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19) and achieves at least one of a reduction in cachexia, increase in survival time, elongation in time to tumor progression, reduction in tumor mass, reduction in tumor burden and/or a prolongation in time to tumor metastasis, time to tumor recurrence, tumor response, complete response, partial response, stable disease, progressive disease, or progression free survival.
  • TNBC triple-negative breast cancer
  • the invention features a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), wherein the cancer is characterized by a tumor comprising EpCAM med/high /CD10 -/low epithelial cells.
  • TNBC triple-negative breast cancer
  • the method includes administering a therapeutically effective dose of an agent that inhibits cyclin-dependent kinase 19 (CDK19) expression or activity, wherein the treatment reduces the number of E P CAM med/high / CD10 -/low cells in the tumor, reduces to number of E P CAM med/high /CD10 -/low cells per unit volume of the tumor, or results in a reduction of the ratio of EpCAMm ed/high /CD1 0 /low epithelial cells to normal (EpCam Hi /CD10 - ) epithelial cells in the tumor.
  • CDK19 cyclin-dependent kinase 19
  • the invention features a method of reducing metastasis of TNBC in a patient by administering a therapeutically effective dose of an agent that inhibits expression or activity of CDK19.
  • the patient is treated with a combination therapy comprising (a) an agent that inhibits expression or activity of CDK19 and (b) radiation therapy and/or chemotherapy.
  • the method comprises detecting EpCAM med/hlgh /CD10 -/low cells in a tissue sample from the patient prior to or after initiating therapy.
  • the agent administered to the patient in the methods described herein does not significantly inhibit expression or activity of CDK8. In some embodiments, the agent inhibits expression or activity of CDK19 to a greater extent than it inhibits expression or activity of CDK8.
  • the agent is a nucleic acid. In some embodiments, the agent is a protein. In some embodiments, the agent is a CRISPR/Cas9 system.
  • the agent is a CDK19 targeting shRNA.
  • the agent is a CDK19 targeting siRNA.
  • the agent is a CDK19 targeting shRNA or siRNA complementary or substantially complementary to the 3′ UTR of CDK19, but not to the 3′UTR CDK8.
  • the agent is a CDK19 targeting shRNA or siRNA complementary or substantially complementary to the coding region of CDK19, but not to the coding region of CDK8.
  • the agent is a CDK19 targeting shRNA or siRNA selected from: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, or SEQ ID NO: 11.
  • the agent binds CDK19 in the cytoplasm of a breast epithelial cell.
  • the invention also features a method of predicting the likely therapeutic responsiveness of a subject with TNBC to a CDK19 targeting agent.
  • the method includes (a) quantitating EpCAM med/high /CD10 -/low cells in a tumor sample obtained from the subject; (b) comparing the quantity of EpCAM med/high /CD10 -/low cells in (a) to a reference value characteristic of tumors responsive to a CDK19 targeting therapy, and treating the patient with the CDK19 targeting agent if the quantity of EpCAM med/high /CD10 -/low cells is equal to or exceeds the reference value.
  • the CDK19 targeting agent is an inhibitor of CDK19 expression or activity
  • FIG. 1 A is a schematic for RNAi dropout viability screens. Two separate screens were performed in a TNBC PDX (PDX-T1). Cells in one experiment were grown in vitro as organoid colonies and in the other in vivo as PDXs in NSG mice.
  • FIGS. 1 B- 1 D are graphs showing that CDK19 knockdown significantly decreased the viability of TNBC cells ( FIG. 1 B : MDA-MB231 cells; FIG. 1 C : MDA-MB468 cells; and FIG. 1 D : HS578T cells) assessed 4 days after transduction with control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2).
  • FIG. 1 E is a graph showing that CDK19 knockdown significantly decreased the formation of organoid colonies in PDX-T1.
  • FIG. 1 F is a graph showing that CDK19 knockdown does not decrease the viability of non-transformed human mammary epithelial cells (HMEC).
  • FIGS. 1 G- 1 J are graphs showing that CDK19 knockdown significantly inhibits the proliferation of PDX tumors ( FIG. 1 G : PDX-T1; FIG. 1 H : PDX-T2; FIG. 11 : PDX-T3; and FIG. 1 J : PDX-T4) grown in NSG mice.
  • FIGS. 1 K and 1 L are bar graphs showing that CDK19 knockdown prevented transduced (RFP positive) TNBC cells ( FIG. 1 K : PDX-T1 and FIG. 1 L PDX-T4) from metastasizing to the lungs in mice.
  • FIG. 1 M shows that in PDX tumors transduced with CDK19 shRNA (images in the second and third rows), very little RFP (images in the last column) is visible. These tumors are composed primarily of un-transduced GFP positive tumor cells (images in the middle column). PDX tumor cells were first labeled with green fluorescent protein (GFP) (middle column) and cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP) (right column).
  • GFP green fluorescent protein
  • RFP red fluorescent protein
  • FIG. 1 N shows representative images of mouse lungs with PDX-T1 metastases. Lungs from mice with PDXs transduced with control shRNA (top row), shCDK19-1 (middle row) or shCDK19-2 (bottom row) are shown. In PDX-T1, which normally metastasizes to the lung, CDK19 knockdown eliminated the detection of any lung metastases by those cells. Bright field images (left column) show gross lung morphology, FITC images (middle column) identify metastatic tumor cells labeled with GFP, and metastatic tumor cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP) (right column).
  • RFP red fluorescent protein
  • FIG. 2 A shows data from representative flow cytometry analyses of a TNBC (PDX-T1) using EpCAM and CD49f (left) or EpCAM and CD10 (right) as cell surface markers.
  • FIG. 2 B is a graph that compares the organoid colony forming capabilities of the EpCAM med/high /CD10 -/low and EPCAM low/med /CD10 low/+ cell sub-populations.
  • FIG. 2 C is a table showing the number of tumors formed and the number of injections performed for six groups of PDX tumor cells. Populations and injections where tumors formed are bolded. PDX tumor cells were isolated by flow cytometry based on the expression of EpCAM and CD10 (as in FIG. 2 A , right)
  • FIGS. 2 D- 2 G are bar graphs showing that CDK19 expression is higher in the EpCAM med/high /CD10 -/low cells compared to the EPCAM low/med /CD10 low/+ cells in PDX-T1, PDX-T2, and PDX-T8.
  • FIG. 3 A includes Venn diagrams showing the number of genes upregulated (upper diagram) and downregulated (lower diagram) by CDK19 knockdown, CDK8 knockdown, or by both CDK19 and CDK8 (overlap region).
  • FIG. 3 B is a Venn diagram of Hallmark gene sets enriched across the genes upregulated (upper diagram) or downregulated (lower diagram) by CDK19 knockdown, CDK8 knockdown, or by both CDK19 and CDK8 knockdowns (overlap region) as determined by GSEA.
  • FIGS. 3 C and 3 D are graphs showing that CHIP-Seq signals across the CDK19KD-H3K27AcUP and CDK19KD-H3K27AcDOWN regions are significantly different in the CDK19 knockdown samples compared to control.
  • FIGS. 3 E and 3 F are graphs showing a gene set enrichment analysis (GSEA) of CDK19KD-EnhancerUP and CDK19KD-EnhancerDOWN genes using averaged CDK19 knockdown versus control expression data.
  • GSEA gene set enrichment analysis
  • FIG. 3 G is a graph showing the hallmark gene sets identified as enriched in Metascape analysis of the CDK19KD-EnhancerUP ‘core’ genes (top and middle bars) and CDK19KD-EnhancerDOWN ‘core’ (bottom bar) genes. The individual genes contributing to the enrichment of each hallmark gene set are shown to the right of each bar.
  • FIGS. 4 A and 4 B are graphs showing that in inducCDK19KD-PDX-Tl cells, induction of CDK19 shRNA by addition of doxycycline significantly decreased the number of organoid colonies in the doxycycline treatment group compared to control. Number of organoid colonies at Day 0 ( FIG. 4 A ) and Day 16 ( FIG. 4 B ) after initiating doxycycline treatment is shown.
  • FIGS. 4 C and 4 D are graphs showing that the induction of CDK19 shRNA in pre-established tumors impaired tumor growth.
  • the growth of pre-established tumors in the doxycycline fed NSG mice and control NSG mice are shown for inducCDK19KD-PDX-Tl ( FIG. 4 C ) and inducCDK19KD-PDX-T3 ( FIG. 4 D ).
  • FIG. 4 E is a graph showing that CDK19 knockdown extends the survival of NSG mice with PDX-T1 tumors.
  • FIG. 4 F shows the chemical structure of CCT251921, an orally bioavailable selective inhibitor of CDK19 and CDK8.
  • FIG. 4 G is a graph showing that the treatment of mice with CCT251921 by daily oral gavage significantly impaired the growth of pre-established PDX-T1 xenograft tumors.
  • FIGS. 5 A and 5 B are graphs showing the shRNA counts in the in vivo growth experimental sample versus the shRNA counts in the baseline sample ( FIG. 5 A ) and the shRNA counts in the in vitro growth experimental sample versus the shRNA counts in the baseline sample ( FIG. 5 B ).
  • FIG. 5 C is a schematic of the criteria used to narrow the initial list of hits from the in vitro and the in vivo screens down to 46 candidate genes.
  • FIG. 5 D is a list of 46 candidate genes determined from the in vitro and the in vivo screens after filtering with the criteria shown in FIG. 5 C .
  • CDK19 is boxed.
  • FIG. 6 A is a bar graph showing that TCGA breast cancer samples from patients with the TNBC subtype are enriched in CDK19 copy number amplifications or CDK19 mRNA upregulation compared to other subtypes.
  • FIG. 6 B includes confocal immunofluorescent images of PDX-T1 stained with cytokeratin 8 (CK8) antibodies (first image from the left), CDK19 antibodies (second image), and DAPI (third image). The composite image composed from all three aforementioned images is shown on the far right (images are representative of three independent experiments).
  • CK8 cytokeratin 8
  • FIGS. 7 A and 7 B are bar graphs showing that CDK19 targeting shRNA effectively silences CDK19 in TNBC cells lines. Expression of CDK19 in MDA-MB231 ( FIG. 7 A ) or MDA-MB468 ( FIG. 7 B ) determined by RT-qPCR for cells transduced with control shRNA, shCDK19-1, and shCDK19-2.
  • FIG. 7 C is a bar graph showing that CDK19 targeting shRNA effectively silences CDK19 in a TNBC PDX. Expression of CDK19 in PDX-T1 as determined by RT-qPCR for cells transduced with control shRNA, shCDK19-1, and shCDK19-2.
  • FIG. 7 D includes images of tissue samples and representative images of mouse lungs bearing PDX-T4 metastases. Lungs from mice with PDXs transduced with control shRNA (top row), shCDK19-1 (middle row), or shCDK19-2 (bottom row) are shown. Bright field images (left column) show gross lung morphology, FITC images (middle column) identify metastatic tumor cells labeled with GFP, and Texas-Red images (right column) identify shRNA-transduced metastatic cells labeled with RFP.
  • FIG. 8 A is a graph showing the flow cytometry analyses of TNBC (PDX-T1) using EpCAM and CD49f and the overlap of the EpCAM med/high /CD10 -/low (1), EPCAM low/med /CD10 low/+ (3) and EpCAM - /CD10 - ( (2)) sub-populations.
  • FIG. 8 B is a bar graph showing that the induction of CDK19 shRNA with doxycycline effectively silences CDK19 in inducCDK19KD-PDX-Tl cells.
  • FIG. 8 C shows that CDK19 knockdown effectively prevents the growth of xenograft tumors in a limiting dilution assay.
  • FIG. 8 D is a graph showing ELDA (Hu et al., Journal of Immunol. Methods 347:70-78, 2009) analysis of the data from FIG. 8 C to determine tumor initiating frequencies in the doxycycline (Group +Dox) and control groups (Group NoDox). P-values as determined by the ELDA software.
  • FIG. 9 shows the amino acid sequence alignment showing 84% sequence homology between CDK19 and CDK8. Amino acid positions are shown above the sequence. Alignment is performed using Clustal W method with MegAlign (DNAStar).
  • FIG. 10 is a table showing hallmark gene sets found enriched by GSEA of the genes upregulated or downregulated by either CDK19 knockdown or CDK8 knockdown.
  • FIGS. 12 A and 12 B show heat map of the expression of CDK19KD-EnhancerUP ‘core’ genes ( FIG. 12 A ) and CDK19KD-EnhancerDOWN ‘core’ genes ( FIG. 12 B ). Normalized expression of each gene in each biological replicate of the CDK19 knockdown and Control samples are shown.
  • FIGS. 13 A- 13 D are graphs showing representative genes where CDK19 knockdown leads to changes in H3K27Ac signals and corresponding changes in gene expression. Representative gene tracks depicting H3K27Ac signals at the loci of select CDK19KD-EnhancerUP ‘core’ ( FIGS. 13 A and 13 B ) and CDK19KD-EnhancerDOWN ‘core’ genes ( FIGS. 13 C and 13 D ).
  • FIG. 13 E is a heat map of the normalized gene expression of ELF3, ETV7, CHI3L2, and CRTAM across each of the three biological replicates in control and CDK19 knockdown samples.
  • FIG. 15 is a table showing the pathological features and patient information for the patient derived xenograft tumors used in the experiments.
  • FIGS. 16 A- 16 D show a nucleic acid alignment of the 3′ UTR of CDK8 and CDK19. The underlined and bolded text indicates the overlapping regions.
  • FIG. 17 shows a nucleic acid alignment of the 5′ UTR of CDK8 and CDK19.
  • the underlined and bolded text indicates the overlapping regions.
  • TNBC Triple Negative Breast Cancer
  • CDK19 has more limited tissue distribution, suggesting reduced toxicity and a broader therapeutic window for CDK19 inhibitors.
  • CDK19 expression was also shown to be enriched in tumor initiating cells, e.g., tumorigenic cells having EpCAM med/high /CD10 -/low expressions, compared to the less tumorigenic cells, e.g., cells having EPCAM low/med /CD10 low/+ expressions (see, e.g., Example 4). Further studies also showed that CDK19 knockdown significantly decreased tumor initiating frequencies ( FIG. 8 D ). This discovery indicates that, compared to other agents, targeting CDK19 will result in a more pronounced and significant effect on highly tumorigenic (e.g., tumor initiating) cells. These discoveries also allow development of theranostic methods for identifying certain TNBC patients likely to respond to CDK19 targeted therapy.
  • TNBC Triple-Negative Breast Cancer
  • TNBC Triple-negative breast cancer
  • ER estrogen receptor
  • PR progesterone receptor
  • Her2 human epidermal growth factor receptor 2
  • Receptor expression can be measured by immunohistochemical staining or other methods.
  • TNBC is generally a diagnosed by exclusion. Widely used breast cancer therapies that target these receptors are not effective against TNBC, making TNBC treatment particularly challenging.
  • CDK19 Cyclic-Dependent Kinase 19
  • RNAPII RNA polymerase II
  • CDK8 Cyclic-Dependent Kinase 8
  • CDK8 is a paralog of CDK19 with 84% amino acid sequence homology to CDK19. See FIG. 9 . CDK8 is described in Broude et al., Curr. Cancer Drug Targets 15:739, 2015 and Sato et al., Molecular Cell 14:685-691, 2004. See UniProt entry CAA59754.1; Genbank entries X85753 & AL590108. The mRNA sequences for CDK8 are also disclosed herein (e.g., SEQ ID NOs:16-18).
  • agent refers to a biological molecule (e.g., nucleic acids, proteins, peptides, antibodies) or small organic molecule (e.g., having a molecular weight less than 1000, usually less than 500) that can reduce or inhibit the expression or activity of CDK19.
  • biological molecule e.g., nucleic acids, proteins, peptides, antibodies
  • small organic molecule e.g., having a molecular weight less than 1000, usually less than 500
  • inhibitor refers to a compound, composition or system that reduces the expression or activity of CDK19.
  • An agent may also selectively inhibit CDK19 expression or activity over that of CDK8.
  • knock down refers to a reduction in the expression level of the CDK19 gene. Knocking down CDK19 gene expression level may be achieved by reducing the amount of mRNA transcript corresponding to the gene, leading to a reduction in the expression level of CDK19 protein. Knocking down CDK19 gene expression level may also be achieved by reducing the amount of CDK19 protein.
  • An knockdown agent is an example of an inhibitor.
  • knock out refers to deleting all or a portion of the CDK19 gene in a cell, in a way that interferes with the function of the CDK19 gene. For example, a knock out can be achieved by altering the CDK19 sequence.
  • a knock out agent is an example of an inhibitor.
  • the terms “decrease,” “reduced,” “reduction,” and “decreasing” are all used herein to refer to a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 5%, at least about 10%, at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (i.e. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.
  • polynucleotide As used herein, the terms “polynucleotide,” “nucleic acid,” and “oligonucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof.
  • a polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs, or otherwise be modified by art-known methods to render the polynucleotide resistant to nucleases, improve delivery of the polynucleotide to target cells or tissues, improve stability, reduce degradation, improve tissue distribution or to impart other advantageous properties.
  • the DNA or RNA polynucleotide may include one or more modifications on the oligonucleotide backbone (e.g., a phosphorothioate modification), the sugar (e.g., a locked sugar), or the nucleobase. If present, modifications to the nucleotide structure can be imparted before or after assembly of the oligonucleotide.
  • the sequence of nucleotides can be interrupted by non-nucleotide components.
  • An oligonucleotide can be further modified after polymerization, such as by conjugation with a label component, a targeting component, or other component.
  • Polynucleotides may be double-stranded or single-stranded molecules.
  • the DNA or RNA oligonucleotide may be packaged into a lipid molecule (e.g., lipid nanoparticles) or be conjugated to a cell-penetrating peptide.
  • a lipid molecule e.g., lipid nanoparticles
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment can include treatment resulting in inhibiting the disease, i.e., arresting its development; and relieving the disease, i.e., causing regression of the disease.
  • a response to treatment can include a reduction in cachexia, increase in survival time, elongation in time to tumor progression, reduction in tumor mass, reduction in tumor burden and/or a prolongation in time to tumor metastasis, time to tumor recurrence, tumor response, complete response, partial response, stable disease, progressive disease, progression free survival, overall survival, each as measured by standards set by the National Cancer Institute and the U.S. Food and Drug Administration for the approval of new drugs and/or described in Eisenhauer, EA1, et al. “New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).” European journal of cancer 45.2 (2009): 228-247.
  • administering includes any route of introducing or delivering an agent that inhibits the expression or activity of CDK19 to the subject diagnosed with TNBC. Administration can be carried out by any route suitable for the delivery of the agent.
  • delivery routes can include, e.g., intravenous, intramuscular, intraperitoneal, or subcutaneous deliver.
  • the agent is administered directly to the tumor, e.g., by injection into the tumor.
  • the term “therapeutically effective amount” refers to an amount, e.g., pharmaceutical dose, effective in inducing a desired biological effect in a subject or patient or in treating a patient having TNBC described herein.
  • the term “therapeutically effective amount” refers to an amount of an active agent being administered that will treat to some extent a disease, disorder, or condition, e.g., TNBC, relieve one or more of the symptoms of the disease being treated, and/or that amount that will prevent, to some extent, one or more of the symptoms of the disease that the subject being treated has or is at risk of developing.
  • a therapeutically effective amount will show an increase or decrease of therapeutic effect of at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%, or at least 1-fold, 2-fold, or 3-fold.
  • a therapeutically effective dose is usually delivered over a course of therapy that may extend for a period of days, weeks, or months.
  • a therapeutically effective dose of an agent may be taken alone or in combination with other therapeutic agents.
  • a therapeutically effective amount of a CDK19 inhibitor is am amount sufficient to effect a partial response in a patient with TNBC (e.g., a greater than 20% reduction, sometimes a greater than 30% reduction, in the measurable diameter of lesions).
  • a “patient” or “subject,” as used herein, is intended to include either a human or non-human animal, preferably a mammal, e.g., non-human primate. Most preferably, the subject or patient is a human.
  • a “antisense strand” refers to the strand of a double stranded RNAi agent (siRNA or shRNA) which includes a region that is complementary or substantially complementary to a target sequence (e.g., a human CDK8 or CDK19 mRNA including a 5′ UTR, exons of an open reading frame (ORF), or a 3′ UTR).
  • a target sequence e.g., a human CDK8 or CDK19 mRNA including a 5′ UTR, exons of an open reading frame (ORF), or a 3′ UTR.
  • a “sense strand,” as used herein, refers to the strand of a RNAi agent (siRNA or shRNA) that includes a region that is complementary or substantially complementary to a region of the antisense strand.
  • the invention provides a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19).
  • TNBC triple-negative breast cancer
  • CDK19 cyclin-dependent kinase 19
  • the treatment results in an at least 10% reduction in tumor volume within 6 month of initiating therapy.
  • the invention provides a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), wherein the cancer is characterized by a tumor comprising EpCAM med/high /CD10 -/low epithelial cells, the method comprising administering a therapeutically effective dose of an agent that inhibits cyclin-dependent kinase 19 (CDK19) expression or activity, wherein the treatment results in a reduction of the ratio of cells having a medium to high expression level of EpCAM and a low expression level of CD10 to normal cells in the tumor.
  • the method includes the step of detecting EpCAM med/high /CD10 -/low epithelial cells in a tissue sample from the patient prior to or after initiating therapy.
  • a biopsy may be obtained from the patient diagnosed with TNBC.
  • a biopsy may be a needle biopsy, or may be a liquid biopsy be obtained from blood vessels and/or lymph nodes that supply the breast, e.g., internal mammary arteries, lateral thoracic arteries, thoracoacromial arteries, axillary lymph nodes.
  • CD10 and EpCAM biomarkers identify three distinct sub-populations of Tumor Initiating Cells (TICs) in TNBC.
  • EpCAM med/high /CD10 -/low EpCAM low/med /CD10 low/+
  • EpCAM - /CD10 EpCAM - /CD10.
  • the phenotype of cancer cells in a TNBC patient can be determined using art-known methods. In one approach a tissue is obtained from the patient and the cell phenotype determined using immunohistochemistry, mass spectrometry analysis, fluorescence activated cell sorting (FACS) or other methods. The cell phenotype can be assigned relative to standard values characteristic of health or cancerous tissue.
  • FACS fluorescence activated cell sorting
  • the ratio of EpCAM med/high /CD10 -/low cells to normal breast epithelial cells is determined prior to initiation of treatment to assess the likely response of the patient to CDK19 targeted therapy.
  • a change in the ratio of EpCAM med/high /CD10 -/low cells to normal cells, or a change in the quantity of EpCAM med/high /CD10 -/low cells per volume tissue is detected after initiation of treatment.
  • the invention provides a method for reducing metastasis of TNBC in a patient, the method comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of CDK19
  • methods of the invention may be used to treat inflammatory TNBCs or TNBCs that are chemo-resistant. In other embodiments, the methods of the invention may be used to slow down or prevent the metastasis of TNBCs. In further embodiments, the methods described herein that target the CDK19 gene or its corresponding protein may further modulate clinically relevant TNBC pathways regulated by CDK19, such as P53 signaling, KRAS signaling, androgen response, NOTCH signaling, TGF BETA signaling, and IL6-JAK-STAT3 signaling ( FIG. 3 B ), and make them more therapeutically susceptible to cancer treatments.
  • CDK19 such as P53 signaling, KRAS signaling, androgen response, NOTCH signaling, TGF BETA signaling, and IL6-JAK-STAT3 signaling ( FIG. 3 B ), and make them more therapeutically susceptible to cancer treatments.
  • the CDK19 gene is essential for the growth of TNBC.
  • Methods of treating TNBC in a subject as described herein may be accomplished by administering a polynucleotide (e.g., oligonucleotide) to the subject to decrease or inhibit the expression of the CDK19 gene.
  • the polynucleotide may be, for example, a DNA oligonucleotide or an RNA oligonucleotide.
  • the oligonucleotide may be used in a CRISPR/Cas system.
  • An oligonucleotide that inhibits or decreases the expression of the CDK19 gene may knock out or knock down the CDK19 gene (e.g., the CDK19 gene in a TNBC cell) in the subject.
  • the oligonucleotide may be an shRNA or an miRNA. In some embodiments, the oligonucleotide may mediate an RNase H-dependent cleavage of the mRNA transcript of the CDK19 gene. In other embodiments, the oligonucleotide may be used in a CRISPR/Cas system.
  • the mRNA transcript of the CDK19 gene may be targeted for cleavage and degradation. Different portions of the mRNA transcript may be targeted to decrease or inhibit the expression of the CDK19 gene.
  • a DNA oligonucleotide may be used to target the mRNA transcript and form a DNA:RNA duplex with the mRNA transcript. The duplex may then be recognized and the mRNA cleaved by specific proteins in the cell.
  • an RNA oligonucleotide may be used to target the mRNA transcript of the CDK19 gene.
  • a short hairpin RNA or small hairpin RNA is an artificial RNA molecule with a hairpin turn that can be used to silence target gene expression via the small interfering RNA (siRNA) it produced in cells.
  • small interfering RNA siRNA
  • shRNA small interfering RNA
  • Fire et. al. Nature 391:806-811, 1998
  • Elbashir et. Al. Nature 411:494-498, 2001
  • Expression of shRNA in cells is typically accomplished by delivery of plasmids or through viral or bacterial vectors.
  • Suitable bacterial vectors include but not limited to adeno-associated viruses (AAVs), adenoviruses, and lentiviruses.
  • AAVs adeno-associated viruses
  • the shRNA is then transcribed in the nucleus by polymerase II or polymerase III depending on the promoter choice.
  • the resulting pre-shRNA is exported from the nucleus and then processed by Dicer and loaded into the RNA-induced silencing complex (RISC).
  • RISC RNA-induced silencing complex
  • the sense strand is degraded by RISC and the antisense strand directs RISC to an mRNA that has a complementary sequence.
  • shRNA is an advantageous mediator of siRNA in that it has relatively low rate of degradation and tu rnover.
  • the methods described herein include treating TNBC in a subject using an shRNA.
  • the methods may include administering to the subject a therapeutically effective amount of a vector, wherein the vector includes a polynucleotide encoding an shRNA capable of hybridizing to a portion of an mRNA transcript of the CDK19 gene.
  • the vector may also include appropriate expression control elements known in the art, including, e.g., promoters (e.g., tissue specific promoters), enhancers, and transcription terminators.
  • the shRNA may be integrated into the cell’s genome and undergo downstream processing by Dicer and RISC (described in detail further herein) to eventually hybridize to the mRNA transcript of the CDK19 gene, leading to mRNA cleavage and degradation.
  • the shRNA may include a nucleic acid sequence that has at least 85% sequence identity to the sequence of GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1) or the sequence of ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2).
  • the shRNA may target the amino acids at the N-terminus of an mRNA transcript of the CDK19 gene.
  • the shRNA may target the amino acids at an internal region of an mRNA transcript of the CDK19 gene.
  • both shRNAs GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1) and ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2)
  • CDK19 knockdown also inhibited the growth of an aggressive PDX obtained from the brain metastasis of a patient with a chemotherapy-resistant inflammatory breast cancer ( FIG. 1 J ), which was known to be aggressive, difficult to treat, and associated with extremely poor prognoses.
  • shRNAs also inhibited the lung metastases of these tumors in mice ( FIG. 1 L ).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCTTGTAGAGAGATTGTACTT (SEQ ID NO: 3). In some embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GAGGACTGATAGTTCTTCTTT (SEQ ID NO: 4).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GATATTAGAAAGATGCCAGAA (SEQ ID NO: 5). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCCAACAGTAGCCTCATAAAG (SEQ ID NO: 6).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to CGTTCGTATTTATCTAGTTTC (SEQ ID NO: 7). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCATGACTTGTGGCATATTAT (SEQ ID NO: 8).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCTTGTAGAGAGATTGCACTT (SEQ ID NO: 9). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to AGGACTGATAGCTCTTCTTTA (SEQ ID NO: 10).
  • an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GTATGGCTGCTGTTTGATTAT (SEQ ID NO: 11).
  • sequence identity e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity
  • GTATGGCTGCTGTTTGATTAT SEQ ID NO: 11
  • available tools for designing shRNAs include, e.g., Project Insilico, Genomics and Bioinformatics Group, LMP, CCR, NIH.
  • an shRNA may be designed to knockout the CDK19 gene.
  • an shRNA can be designed in consideration of its multiple structural elements.
  • an shRNA should be about 80 nucleotides in length and designed (from 5′ to 3′) to comprise of the following structural elements to make the hairpin structure of the shRNA: (1) a sense strand (e.g., upper stem); (2) followed by a hairpin loop; (3) an antisense strand (e.g., lower stem or guide strand) that has perfect or near perfect complementary to the target mRNA and is antisense to the target mRNA; (4-5) two cleavage motifs such as, “U” or “UH” at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand; and (6) arbitrary spacer nucleotides of about two nucleotides in length between the first nucleotide of guide strand “U” motif and the hairpin loop, and between
  • the sense strand and antisense strand, making up the stem may be designed to consist of a range from about 19 to 29 nucleotides in length, which will form the stem.
  • the loop structure may be designed to consist of a range about 2 to 15 nucleotides in length, and preferably free of any internal secondary structure.
  • sequences that may be used for making the hairpin loop include but are not limited to, a nine nucleotide loop comprising the sequence (TTCAAGAGA), and a seven nucleotide loop comprising the sequence (TCAAGAG).
  • Other design strategies can be found in the relevant disclosure of Ros XB-D, Gu S. Guidelines for the optimal design of miRNA-based shRNAs.
  • target sequence should also be considered, as many mRNAs can share similar sequences. Care should be taken in selecting target sequence that has low sequence homology to other genes in the genome to allow for gene-specific knockdown. Where a gene has multiple forms, to achieve complete knockdown of gene expression, shRNA should target sequences shared among all isoforms of the target mRNA.
  • An alignment of CDK19 and CDK8 mRNA sequences can identify not identical or low percent identity or similarity nucleotide sequence regions which can be used to design shRNAs that have a preference to target to CDK19 mRNA but not CDK8, see for example the 3′ UTR and 5′ UTR alignments in FIG. 16 and FIG. 17 .
  • shRNA that targets a CDK19 mRNA transcript, and not of CDK8 mRNA transcript can be designed.
  • the mRNA sequences for human CDK19 and CDK8 from National Center for Biotechnology Information (NCBI, found at Pubmed.gov) and an alignmenti is performed (e.g., with pairwise alignment program such as, LALIGN).
  • a region of about 19 to 29 contiguous nucleotides e.g., 19-20, 19-21, 19-22, 19-23, 19-24, 19-25, 19-26, 19-27, 19-28, or 19-29
  • low sequence identity e.g., less than 75%, identity, sometimes less than 70% identity, sometimes less than 60% identity.
  • the 19 to 29 nt region has very low (e.g., less than 40%, less than 30% or less than 20% or sequence identity.
  • the contiguous sequence can be in a protein coding region, the 5′-UTR, the 3′-UTR, or span two regions.
  • target-specific knockdown of CDK19 can be accomplished by designing an shRNA with a guide strand that is complementary of the 3′ UTR region of CDK19 (SEQ ID NO:42) and has low or no homology to the 3′UTR of CDK8 (SEQ ID NO:44).
  • the guide strand may be 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, or 29 nucleotides in length.
  • Some exemplary sequence regions that may be used to design a CDK19 shRNA include but are not limited to, CTCCAGCTCCCGTTGGGCCAGGCCAGCCC (SEQ ID NO: 20), AGCCCAGAGCACA GGCTCCAGCAATATGT (SEQ ID NO: 21), CTGCATTGAAAAGAACCAAAAAAATGCAA (SEQ ID NO: 22), ACTATGATGCCATTTCTATCTAAAACTCA (SEQ ID NO: 23), TACACATGGGAG GAAAACCTTATATACTG (SEQ ID NO: 24), AGCATTGTGCAGGACTGATAGCTCTTCTT (SEQ ID NO: 25), TATTGACTTAAAGAAGATTCTTGTGAAGT (SEQ ID NO: 26), TTCCCCTATCTCAGCA CCCCTTCCCTGCA (SEQ ID NO: 27), TGTGTTCCATTGTGACTTCTCTGATAAAG (SEQ ID NO: 28), CGTCTGATCTAATCCCAGCACTTCTGTAA (SEQ ID NO: 29), or CCTTC
  • the shRNA may be designed to be targeted to upstream of CDK19, downstream of CDK19, or in the exons of CDK19.
  • the expression of the CDK19 shRNA results in knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the expression of the CDK19 shRNA can preferentially knockdown CDK19 compared to CDK8.
  • CDK19 a region that does not have significant homology to other CDKs (e.g., CDK8) or other mRNAs in the genome.
  • an appropriate promoter such as a pol II or pol III promotor at the beginning of the cassette, followed by the complementary sense strand (e.g., complementary to the targeting guide strand), which is them followed by the loop structure of about 2 to 15 nucleotides in length.
  • the two Ago cleavage motifs, “U” or “UH” should be included at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand along to 1-2 spacer nucleotides at the end of the loop structure.
  • target-specific knockdown of CDK8 can be performed by using an shRNA with a guide strand that comprises a complementary to the 5′UTR of CDK8 (SEQ ID NO: 43) and has low or no homology to the 5′ UTR of CDK19 (SEQ ID NO:41).
  • the guide strand may be 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29, nucleotides in length.
  • Some exemplary sequences that may be used to design a CDK8 shRNA include but are not limited to, TGGCCGCCCCGCCGCTCCCGCCGCAGCAG (SEQ ID NO: 31), GAGCAGAACGCGCGGCCGGAGA GAGCGGC (SEQ ID NO: 32), GGAGCCGGCCCAGGGAGCCCGCGGGGA (SEQ ID NO: 33), CAAGGGCAGAGACACCGCTCCCCACCCCC (SEQ ID NO: 34),AGCCCTCGTCCCTCGGCTCTCCTTCGCCG (SEQ ID NO: 35), GGGGATCCTCCCCGTTCCTCCACCCCCGG (SEQ ID NO: 36), CCGGCCTCTG CCCCGCCGTCCCCCTGGAT (SEQ ID NO: 37), GTCCCTGGCGCTTTCGCGGGGCCTCCTCC (SEQ ID NO: 38), TGCTCTTGCCGCATCAGTCGGGCTGGTGC (SEQ ID NO: 39), or TGCGGCCGGCGGGCGTAGAGC GGGCGGGT (SEQ ID NO
  • the shRNA may be designed to be targeted to upstream of CDK8, downstream of CDK8, or in the exons of CDK8.
  • the expression of the CDK8 shRNA can result in a knockdown of CDK8 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • CDK8 a region that does not have significant homology to other CDKs (e.g., CDK19) or other mRNAs in the genome.
  • an appropriate promoter such as a pol II or pol III promotor at the beginning of the cassette, followed by the complementary sense strand (e.g., complementary to the targeting guide strand), which is them followed by the loop structure of about 2 to 15 nucleotides in length.
  • the two Ago cleavage motifs, “U” or “UH” should be included at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand along to 1-2 spacer nucleotides at the end of the loop structure.
  • the specificity or knockdown level of an shRNA or siRNA can be confirmed using real-time PCR analysis for mRNA level or ELISA assay for the protein level.
  • Experimental controls may be run in parallel to assess knockdown.
  • Some examples of experimental controls that may be used include but are not limited to, a mock-infected or mock-transfected sample, an empty vector, an shRNA encoding a scrambled target or seed region, an shRNA targeting another gene entirely such as, housekeeping genes GAPDH or Actin, or a GFP positive control.
  • an siRNA or shRNA e.g., RNAi agent
  • RNAi agent e.g., RNAi agent
  • the expression of RNA is measured.
  • the expression of the protein is measured.
  • mRNA may be measured by any PCR-based assay known in the art (e.g., RT-PCR or qRT-PCR or the like).
  • the protein level may be measured by an immunoassay (e.g., ELISA assay or any antibody-based method known in the art).
  • a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 30% and CDK8 greater than about 70% relative to a system without transfection or transduction. In some other embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression at less than about 50% and CDK8 greater than about 95%. In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 5% and CDK8 greater than about 80%. In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 1% and CDK8 greater than about 60%.
  • a targeting CDK19 shRNA or siRNA results in CDK19 expression at less than about 0.5% and CDK8 greater than about 90%. In some embodiments, a targeting CDK19 shRNA results in CDK19 expression at about 0% and CDK8 at about 100% relative to a system without transfection or transduction. In some embodiments, the expression of RNA is measured. In other embodiments, the expression of the protein is measured.
  • the present disclosure also provides siRNA-based therapeutics for inhibiting expression of CDK8 and CDK19 in a patient with triple-negative breast cancer.
  • the double stranded RNAi therapeutic includes a sense strand complementary to an antisense strand.
  • the sense or antisense strands of the siRNA may be about 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.
  • the antisense strand of the siRNA-based therapeutic includes a region complementary to a part of an mRNA encoding CDK8 or CDK19. Additional methods to make therapeutic siRNA can be found in U.S. Pat No. US9399775, which is incorporated by reference in its entirety for all purposes.
  • the expression of CDK19 siRNA may result in a knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the expression of CDK19 siRNA may preferentially knockdown CDK19 compared to CDK8.
  • the expression of CDK8 siRNA may result in a knockdown of CDK8 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • CDK19 siRNA may result in a knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% and CDK8 at least about 10%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, or 30%.
  • delivery vectors that may be used with the present disclosure are viral vectors, plasmids, exosomes, liposomes, bacterial vectors, or nanoparticles.
  • the present disclosure also provides for delivery by any means known in the art.
  • delivery vectors may be genetically modified to target a specific cell type or to tissue type.
  • a targeted delivery vector or plasmid one can identify a unique molecule expressed or associated with a triple-negative breast cancer (e.g., receptor, protein, glycoprotein, or combination thereof) and then create a delivery vector or plasmid that harbors or expresses these markers, preferably on the outside of the delivery vector or plasmid (e.g., cytosol facing).
  • a viral delivery vector can be genetically modified to be continuously replicating, replication-defective, or conditionally replicating as described in, Sliva K, Schnierle BS. Selective gene silencing by viral delivery of short hairpin RNA. Virology Journal . 2010.
  • the CDK8 or CDK19 shRNA or siRNA can be delivered by an adenovirus vector.
  • Adenoviruses non-enveloped viruses with a nucleocapsid and a linear dsDNA genome. While they are able to replicate in the nucleus of mammalian cells, they do not efficiently integrate into the host’s genome and therefore pose only minimal risks of insertional mutagenesis but are inadequate for long-term therapy.
  • the CDK8 or CDK19 shRNA or siRNA can be delivered by an adeno-associated viral vector (AAV).
  • AAV is one of the smallest viruses and belongs to the genus Dependovirus . It has a small, single-stranded DNA genome and can accommodate about eight individual shRNA. AAV permits entry retargeting, allowing delivery of the shRNA to specific cell or tissue types.
  • the present disclosure provides for a modified AAV that is targeted for delivery to a triple-negative breast cancer cell or tissue type.
  • the CDK8 or CDK19 shRNA or siRNA can be delivered by a retrovirus vector.
  • a retrovirus is a single-stranded RNA virus that belongs to the family of Retroviridae and replicate through a double-stranded DNA intermediate. They can integrate into a host’s genome thereby allowing long-term expression of a shRNA.
  • the Env protein plays a central role in targeting retrovirus to a target cell.
  • the present disclosure provides for a retrovirus vector with a modified env gene or its protein product for delivery to a triple-negative breast cancer cell or tissue type.
  • the present disclosure provides for delivery of CDK8 or CDK19 shRNA of siRNA using a retrovirus vector with protease-activated Env proteins.
  • the CDK8 or CDK19 shRNA or siRNA can be delivered by a lentivirus vector.
  • Lentivirus is a subclass of retrovirus in the genus Lentivirinae which can accommodate large amounts of DNA.
  • a lentivirus vector engineered to be “self-inactivating” known as “SIN” vectors.
  • the present disclosure provides for delivery of a CDK8 or CDK19 shRNA by a lentivirus vector with a modified env gene or its protein product for delivery to a triple-negative breast cancer cell or tissue type.
  • the shRNA or siRNA can be delivered by a nanoparticle.
  • nanoparticles that can be use with the present disclosure, include but are not limited to, exosomes, liposomes, organic nanoparticles, or inorganic nanoparticles.
  • Other non-limiting examples of nanoparticles include, but are not limited to, e.g., those provided in Hong, Cheol Am, and Yoon Sung Nam. “Functional Nanostructures for Effective Delivery of Small Interfering RNA Therapeutics.” Theranostics 4.12 (2014): 1211-1232. PMC. Web. 13 Sept. 2018, which is hereby incorporated by reference in its entirety for all purposes.
  • the delivery of the shRNA or siRNA is mediated by receptor, protein, glycoprotein or combination thereof present or specific to triple-negative breast cancer cells.
  • the siRNA CDK19 therapeutic is administered in a solution.
  • the siRNA may be administered in an unbuffered solution.
  • the siRNA is administered in water.
  • the siRNA is administered with a buffer solution, such as an acetate buffer, a citrate buffer, a prolamine buffer, a carbonate buffer, or a phosphate buffer or any combination thereof.
  • the buffer solution is phosphate buffered saline.
  • RNase H-dependent antisense oligonucleotides are single-stranded, chemically modified oligonucleotides that bind to complementary sequences in target mRNAs and reduce gene expression both by RNase H-mediated cleavage of the target RNA and by inhibition of translation by steric blockade of ribosomes.
  • RNase H is an endonuclease enzyme that catalyzes the cleavage of RNA in an RNA:DNA duplex.
  • the most well studied endogenous function for this enzyme is the removal of Okazaki fragments (small RNAs) used to prime the DNA duplication during cell division.
  • a nucleic acid e.g., DNA oligonucleotide
  • a portion of the mRNA may be administered to the subject.
  • the DNA oligonucleotide Once inside the cell (e.g., a TNBC cell), the DNA oligonucleotide base pairs with its targeted mRNA transcript.
  • RNase H may bind to the resulting duplex and cleave the mRNA transcript at one or more places.
  • the DNA oligonucleotide may further bind to other mRNA transcripts to target them for RNase H degradation.
  • the expression of the CDK19 gene may be greatly reduced in a subject with TNBC.
  • the DNA oligonucleotide capable of hybridizing to an mRNA transcript of a CDK19 gene may contain, e.g., between 10 and 30 nucleotides (e.g., 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, or 30 nucleotides). In some embodiments, the DNA oligonucleotide may have 100% complementarity to the portion of the mRNA transcript it binds.
  • the DNA oligonucleotide may have less than 100% complementarity (e.g., 95%, 90%, 85%, 80%, 75%, or 70% complementarity) to the portion of the mRNA transcript it binds, but can still form a stable RNA:DNA duplex for the RNase H to cleave the mRNA transcript.
  • the DNA oligonucleotide may bind to the 5′ UTR or the 3′ UTR of the mRNA transcript of the CDK19 gene.
  • the DNA oligonucleotide capable of hybridizing to an mRNA transcript of a CDK19 gene may contain modified nucleotides at the 5′ end and the 3′ end.
  • the modified nucleotides at the termini may function to protect the internal portion of the DNA oligonucleotide from nuclease degradation and to increase the binding affinity for the target mRNA transcript.
  • the modified nucleotides at the termini may include a modified nucleobase (e.g., 5-methylcytosine) and/or a modified sugar (e.g., a locked sugar).
  • 3-5 nucleotides at each of the 5′ and 3′ ends of the DNA oligonucleotide may be modified.
  • a microRNA is a small non-coding RNA molecule that functions in RNA silencing and post-transcriptional regulation of gene expression. miRNAs base pair with complementary sequences within the mRNA transcript. As a result, the mRNA transcript may be silenced by one or more of the mechanisms such as cleavage of the mRNA strand, destabilization of the mRNA through shortening of its poly(A) tail, and decrease translation efficiency of the mRNA transcript into proteins by ribosomes. In some embodiments, miRNAs resemble the siRNAs of the shRNA pathway, except that miRNAs derive from regions of RNA transcripts that fold back on themselves to form short hairpins, which are also called pri-miRNA.
  • the hairpins are cleaved out of the primary transcript in the nucleus by an enzyme called Drosha.
  • the hairpins, or pre-miRNA are then exported from the nucleus into the cytosol.
  • the loop of the hairpin is cleaved off by an enzyme called Dicer.
  • Dicer an enzyme called Dicer.
  • the resulting product is now a double strand RNA with overhangs at the 3′ end, which is then incorporated into RISC.
  • the second strand is discarded and the miRNA that is now in the RISC is a mature miRNA, which binds to mRNAs that have complementary sequences.
  • miRNAs and siRNAs from the shRNA pathway differ from base pairing with miRNAs comes from the 5′ end of the miRNA, which is also referred to as the seed sequence. Since the seed sequence is short, each miRNA may target many more mRNA transcript. In some embodiments, an miRNA targeting the CDK19 gene may be used in methods described herein.
  • the knocking out or knocking down of the CDK19 gene is performed using a gene editing system such as the CRISPR/Cas system.
  • a gene editing system such as the CRISPR/Cas system. See Sanders and Joung, Nature Biotechnol 32:347-355, 2014, Huang et al., J Cell Physiol 10:1-17, 2017 and Mitsunobu et al., Trends Biotechnol 17:30132-30134, 2017.
  • the CRISPR/Cas system includes a Cas protein and at least one or two ribonucleic acids that are capable of directing the Cas protein to and hybridizing to a target motif in the CDK19 sequence. The Cas protein then cleaves the target motif and results in a double-strand break or a single-strand break.
  • any CRISPR/Cas system that is capable of altering a target polynucleotide sequence in a cell can be used in methods described here.
  • the CRISPR/Cas system is a CRISPR type I system.
  • the CRISPR/Cas system is a CRISPR type II system.
  • the CRISPR/Cas system is a CRISPR type V system.
  • the Cas protein used in the methods described herein can be a naturally occurring Cas protein or a functional derivative thereof.
  • a “functional derivative” includes, but are not limited to, fragments of a native sequence and derivatives of a native sequence polypeptide and its fragments, provided that they have a biological activity in common with the corresponding native sequence polypeptide.
  • a biological activity contemplated herein is the ability of the functional derivative to hydrolyze a DNA substrate (e.g., a CDK19 gene) into fragments.
  • the term “derivative” encompasses both amino acid sequence variants of polypeptide, covalent modifications, and fusions thereof. Suitable derivatives of a Cas protein or a fragment thereof include but are not limited to mutants, fusions, or covalent modifications of Cas protein.
  • the Cas protein used in methods described herein is Cas9 or a functional derivative thereof.
  • the Cas9 protein is from Streptococcus pyogenes.
  • Cas9 contains 2 endonuclease domains, including an RuvC-like domain which cleaves target DNA that is noncomplementary to crRNA, and an HNH nuclease domain which cleaves target DNA complementary to crRNA.
  • the double-stranded endonuclease activity of Cas9 also requires that a short conserved sequence (e.g., 2-5 nucleotides), known as a protospacer-associated motif (PAM), follows immediately after the 3′ end of a target motif in the target sequence.
  • PAM protospacer-associated motif
  • the Cas protein is introduced into TNBC cells in polypeptide form.
  • the Cas protein may be conjugated to a cell-penetrating polypeptide.
  • Non-limiting examples of cell-penetrating peptides include, but are not limited to, e.g., those provided in Milletti et al., Drug Discov. Today 17: 850-860, 2012, the relevant disclosure of which is hereby incorporated by reference in its entirety.
  • a TNBC cell may be genetically engineered to produce the Cas protein.
  • the target motif in the CDK19 gene, to which the Cas protein is directed by the guide RNAs may be between 15 and 25 nucleotides in length (e.g., 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length). In some embodiments, the target motif is at least 20 nucleotides in length. In some embodiments, the target motif in the CDK19 gene immediately precedes a short conserved sequence known as a protospacer-associated motif (PAM), recognized by the Cas protein. In some embodiments, the PAM motif is an NGG motif. In some embodiments, the target motif of the CDK19 gene is within the first exon.
  • PAM protospacer-associated motif
  • the target motifs can be selected to minimize off-target effects of the CRISPR/Cas systems.
  • Those skilled in the art will appreciate that a variety of techniques can be used to select suitable target motifs for minimizing off-target effects (e.g., bioinformatics analyses).
  • the ribonucleic acids that are capable of directing the Cas protein to and hybridizing to a target motif in the CDK19 gene are referred to as single guide RNA (“sgRNA”).
  • the sgRNAs can be selected depending on the particular CRISPR/Cas system employed, and the sequence of the target polynucleotide, as will be appreciated by those skilled in the art.
  • the one or two ribonucleic acids can also be selected to minimize hybridization with nucleic acid sequences other than the target polynucleotide sequence.
  • the one or two ribonucleic acids are designed to hybridize to a target motif immediately adjacent to a deoxyribonucleic acid motif recognized by the Cas protein.
  • Guide RNAs can also be designed using available software, for example, CRISPR Design Tool (Massachusetts Institute of Technology).
  • the one or more sgRNAs can be transfected into TNBC cells, according to methods known in the art.
  • antibodies can be used to make antibody-drug conjugates in which the antibody is linked to a drug and directs that agent to the tumor by specifically binding to the tumor.
  • Ado-Trastuzumab emtansine T-DM1 is an example of an approved antibody-drug conjugate used for the treatment of breast cancer (see, Deng et al., Curr. Med. Chem. , Vol. 24(23), 2505-2527 (2017).
  • Another type of immunotherapy is active immunotherapy, or vaccination, with an antigen present on a specific cancer (e.g., TNBC cells) or a DNA construct that directs the expression of the antigen, which then evokes the immune response in the subject, i.e., to induce the subject to actively produce antibodies against their own cancer.
  • a specific cancer e.g., TNBC cells
  • a DNA construct that directs the expression of the antigen, which then evokes the immune response in the subject, i.e., to induce the subject to actively produce antibodies against their own cancer.
  • Antibodies have been highly effective in targeting cell surface proteins involved in disease. Though it is generally believed that their large size, complex architecture, and structural reliance on disulfide bonds preclude intracellular application, a number of examples of both in situ-expressed (see, e.g, Miersch and Sidhu, F1000Res doi: 10.12688/f1000research.8915.1, 2016) and exogenously supplied whole antibodies shown to maintain functional intracellular activity exist in the literature (see, e.g., Biocca et al., Expression and targeting of intracellular antibodies in mammalian cells. EMBO J .
  • antibody encompasses, but is not limited to, whole immunoglobulin (i.e., an intact antibody) of any class.
  • Native antibodies are usually heterotetrameric glycoproteins, composed of two identical light (L) chains and two identical heavy (H) chains.
  • L light
  • H heavy
  • each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes.
  • Each heavy and light chain also has regularly spaced intrachain disulfide bridges.
  • Each heavy chain has at one end a variable domain (V(H)) followed by a number of constant domains.
  • V(H) variable domain
  • Each light chain has a variable domain at one end (V(L)) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
  • Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains.
  • the light chains of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda (A), based on the amino acid sequences of their constant domains.
  • immunoglobulins can be assigned to different classes.
  • immunoglobulins There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG-1, IgG-2, IgG-3, and IgG-4; IgA-1 and IgA-2.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • epitope is meant to include any determinant capable of specific interaction with the provided antibodies.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • Identification of the epitope that the antibody recognizes is performed as follows. First, various partial structures of the target molecule that the monoclonal antibody recognizes are prepared. The partial structures are prepared by preparing partial peptides of the molecule. Such peptides are prepared by, for example, known oligopeptide synthesis technique or by incorporating DNA encoding the desired partial polypeptide in a suitable expression plasmid. The expression plasmid is delivered to a suitable host, such as E.
  • a series of polypeptides having appropriately reduced lengths, working from the C- or N-terminus of the target molecule, can be prepared by established genetic engineering techniques. By establishing which fragments react with the antibody, the epitope region is identified. The epitope is more closely identified by synthesizing a variety of smaller peptides or mutants of the peptides using established oligopeptide synthesis techniques. The smaller peptides are used, for example, in a competitive inhibition assay to determine whether a specific peptide interferes with binding of the antibody to the target molecule. If so, the peptide is the epitope to which the antibody binds.
  • kits such as the SPOTs Kit (Genosys Biotechnologies, Inc., The Woodlands, TX) and a series of multipin peptide synthesis kits based on the multipin synthesis method (Chiron Corporation, Emeryvile, CA) may be used to obtain a large variety of oligopeptides.
  • antibody or fragments thereof can also encompass chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab′)2, Fab′, Fab and the like, including hybrid fragments.
  • fragments of the antibodies that retain the ability to bind their specific antigens are provided.
  • fragments of antibodies which maintain CDK19 binding activity are included within the meaning of the term antibody or fragment thereof.
  • Such antibodies and fragments can be made by techniques known in the art and can be screened for specificity and activity according to general methods for producing antibodies and screening antibodies for specificity and activity (See Harlow and Lane. Antibodies, A Laboratory Manual. Cold Spring Harbor Publications, New York (1988)).
  • antibody or fragments thereof conjugates of antibody fragments and antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference in their entirety.
  • a therapeutic antibody (or antibody fragment) can be prepared using methods known in the art, having specificity for an antigen present in breast cancer, and in particular TNBC cells, that is absent or present only at low levels in any normal (non-cancerous) tissue.
  • the therapeutic antibody would therefore have biological activity against TNBC cells and be able to recruit the immune system’s response to treat the disease.
  • the therapeutic antibody can be administered as a therapeutic alone or in combination with current treatments (such as chemotherapy, radiation, or platinum-based therapies) or used to prepare immunoconjugates linked to toxic agents, such as drugs.
  • Monoclonal antibodies to CDK19 can be used to identify the presence or absence of cancerous cells in breast tissue, for purposes of diagnosis or treatment.
  • Anti-CKD19 antibodies can also be used to identify the presence or absence of cancerous cells, or the level thereof, which are circulating in the blood after their release from a solid tumor.
  • Such circulating antigen can include an intact CDK19 antigen, or a fragment thereof that retains the ability to be detected according to the methods taught herein. Such detection may be effected for example, by FACS analysis using standard methods commonly used in the art.
  • methods of targeting CDK19 can include administering to a subject in need thereof, a therapeutically effective amount of an antibody (e.g., an anti-CKD19 antibody) that is immunoreactive to CDK19 for the treatment of breast cancer, in particular treatment of TNBC.
  • an antibody e.g., an anti-CKD19 antibody
  • the antibody having immunoreactivity to CDK19 targets intracellular signaling molecules, such as kinases, as opposed to cell surface molecules, whereby the specificity of the antibody is provided by neutralizing epitope(s) present on CDK19 that are not present on CDK8.
  • the anti-CDK19 antibody can target the Pl3K/mTOR/AKT pathway or ERK5 (see, Ocana and Pandiella, Oncotarget , 8(13), 22218-22234 (2017)).
  • the anti-CDK19 antibody can target multiple intracellular signaling molecules, for example, the Pl3K/mTOR and JAK/STAT pathway.
  • the anti-CDK19 antibody can comprise an engineered protein that binds to a neutralizing epitope present on CDK19 that is not present on CDK8.
  • methods of targeting CDK19 can include administering to a subject in need thereof, a therapeutically effective amount of a tumor antigen (TA)-specific monoclonal antibody for the treatment of TNBC.
  • TA tumor antigen
  • the TA-specific mAB can be directed to an intracellular antigen associated with TNBC (See for example, Wang et al., Molecular Oncology , Vol. 9(10), (2015) 1982-1993 and Just, FEBS letters , 2:21 (2014), 350-355).
  • a method of treating a subject with breast cancer including the step of administering to the subject a pharmaceutically effective amount of a composition comprising a CDK19 targeting agent.
  • the CDK19 targeting agent may be a CDK19 targeted antibody, a CDK19 targeted peptide, a CDK19 targeted small molecule, a CDK19 targeted RNA molecule, or a combination thereof.
  • the CDK19 targeted agent may be conjugated to a therapeutic agent.
  • the method further includes administering a second form of cancer therapy (e.g., chemotherapy or radiation therapy) to the subject.
  • the breast cancer is TNBC.
  • a method of inhibiting expression of the CDK19 gene in a breast cancer cell the method including the steps of contacting a breast cancer cell expressing the CDK19 gene with a synthetic CDK19 targeted RNA molecule.
  • a method of assessing responsiveness of a subject with cancer to a CDK19 targeted agent including the steps of: (a) measuring in a tumor sample from a subject the amount of CDK19; (b) determining if a subject has a cancer characterized as having a high level of CDK19 expression; and (c) indicating that the subject is more likely to respond to the CDK19 targeted agent if the subject’s cancer is characterized as having a high level of CDK19 expression or that the subject is less likely to respond to the CDK19 targeted agent if the subject’s cancer is characterized as having a low level of CDK19 expression.
  • a method of treating a subject with cancer comprising administering to the patient a pharmaceutically effective amount of a composition comprising a CDK19 targeted agent.
  • the CDK19 targeted agent is an agent that specifically binds to CDK19 protein or to CDK19 mRNA.
  • CDK19 targeted agents include antibodies, or fragments thereof, peptides, small molecules, and polynucleotides (such as RNA molecules) that specifically bind to CDK19 protein or to CDK19 mRNA.
  • the composition may further comprise a pharmaceutically acceptable carrier.
  • CDK19 targeted agents that bind to the CDK19 protein may directly inhibit CDK19 activity.
  • CDK19 targeted agents that bind to CDK19 mRNA may inhibit CDK19 expression and thereby inhibit CDK19 activity.
  • the CDK19 targeted agent may comprise a CDK19 targeted antibody.
  • the CDK19 targeted antibody may be a monoclonal antibody.
  • the CDK19 targeted antibody may be a humanized antibody.
  • the CDK19 targeted agent may be a CDK19 targeted peptide.
  • the CDK19 targeted agent may be a CDK19 targeted small molecule.
  • the CDK19 targeted peptides and small molecules may be derived in a variety of manners as discussed further below. In some instances, the peptides are derived from the sequence of a CDK19 targeted antibody.
  • treating a subject with the methods described herein inhibits at least one of: formation of a tumor, the proliferation of tumor cells, the growth of tumor cells, or metastasis of tumor cells in the subject.
  • treating a subject with the methods described herein may result in reduction of tumor size and, in some instances, elimination of one or more tumors in the subject.
  • methods for treating TNBC include targeting the CDK19 protein using a small molecule inhibitor of CDK19 activity.
  • small molecule inhibitors of CDK19 are described in U.S. Pat. No. 9,321,737, U.S. Pat. Publication No. US 20170071942, Mallinger et al., J. Med. Chem. 59:1078, 2016, and Czodrowski et al., J. Med. Chem . 59:9337, 2016.
  • the small molecule inhibitors bind to the ATP binding site of CDK19 to inhibit its activity.
  • the small molecule inhibitor of CDK19 may bind to the ATP binding site of CDK19 covalently or non-covalently to inhibit its activity. In other embodiments, the small molecule inhibitor may bind to other parts of CDK19 outside of the ATP binding site. For example, the small molecule inhibitor may form a covalent interaction with an amino acid (e.g., methionine, tyrosine, or serine) outside of the ATP binding site to inhibit CDK19 activity. In addition to occupying the ATP binding to inhibit kinase activity, a small molecule inhibitor may also bind to CDK19 to cause a conformational change in CDK19 that prevents CDK19 from functioning.
  • an amino acid e.g., methionine, tyrosine, or serine
  • the small molecule inhibitor may bind to CDK19 with a higher affinity than to CDK8. As shown in FIG. 9 , the vast majority of amino acid differences between CDK19 and CDK8 are in the C-terminal domain. In some embodiments, without being bound by any theory, a small molecule inhibitor may bind to an amino acid or a portion in the C-terminal domain of CDK19, that is different from the corresponding amino acid or portion of CDK8, to achieve selective inhibition of CDK19 over CDK8.
  • the small molecule inhibitor is other than a compound described in U.S. Pat. No. 9,321,737. In some embodiments the small molecule inhibitor is other than a compound described in U.S. Pat. Publication No. US 20170071942. In some embodiments the small molecule inhibitor is other than a compound described in, Mallinger et al., J. Med. Chem . 59:1078, 2016. In some embodiments the small molecule inhibitor is other than a compound described in Czodrowski et al., J. Med. Chem . 59:9337, 2016.
  • the small molecule inhibitor is other than one or more compounds selected from the group consisting of Cortistatin A, Sorafenib, Linifanib, Ponatinib, Senexin B, CCT251545, and CCT251921
  • Agents that inhibitors expression or activity of CDK19 but do not inhibit expression or activity of CDK8, or agents that inhibit expression or activity of CDK19 to a greater extent than expression or activity of CDK8 is inhibited can be designed based on differences in sequence and structure of the CDK19 and CDK8 proteins and their corresponding genes and mRNAs. For example, an alignment of CDK19 and CDK8 mRNA sequences can identify non-identical or low identity nucleotide sequences that can be used to design shRNAs or other nucleic acid agents that associate with CDK19 mRNA but not CDK8 sequences. (see, FIGS. 16 and 17 ).
  • aligning CDK19 and CDK8 amino acid sequences can identify divergent regions and antibodies or other binding agents can be produced to specifically bind the CDK19 protein.
  • small molecule agents can be identified (by rational drug design or screening) that specifically inhibit CDK19 activity or inhibit CDK19 activity to a greater degree that CDK8 activity.
  • an agent that inhibits CDK19 activity but does not significantly inhibit activity of CDK8 refers to an agent that is capable of specifically binding and inhibiting the activity of CDK19 such that minimal CDK19 activity is detected in vivo or in vitro; while the agent causes no significant decrease in CDK8 activity under the same conditions.
  • an agent that inhibits activity of CDK19 can specifically bind to CDK19 and fully or significantly inhibit CDK19 activity in vivo or in vitro.
  • a CDK19 inhibitor can be identified by its ability to preferentially bind to the CDK19 gene or a CDK19 gene product, and fully inhibit expression or activity of CDK19.
  • CDK19 activity when exposed to an agent, is at least about 70% less, for example, at least about 75%, 80%, 90%, or 95% less than CDK19 activity in the presence of a control or in the absence of the agent.
  • No significant decrease in CDK8 activity occurs when CDK8 activity, upon exposure to the agent, is at least about 90%, for example, at least 95%, 96%, 97%, 98%, 99%, or 100%, in comparison to CDK8 activity in the absence of the agent.
  • the agent can include small molecules (i.e., a molecule having a formula weight of 1000 Daltons or less), such as small molecule chemical inhibitors or large molecules, such as siRNA, shRNA, antisense oligonucleotides, or proteins.
  • small molecules i.e., a molecule having a formula weight of 1000 Daltons or less
  • small molecule chemical inhibitors or large molecules such as siRNA, shRNA, antisense oligonucleotides, or proteins.
  • Determining the effect of the agent on CDK19 and/or CDK8 activity can be measured using one or more methods known in the art, including but not limited to, half maximal inhibitory concentration (IC 50 ), dissociation constant (K D ), and inhibitor constant (K l ).
  • IC 50 is a measure of the effectiveness of a substance in inhibiting a specific biological or biochemical function. This value indicates the concentration of the substance needed to inhibit a given biological process (or component of the biological process) by half.
  • the IC 50 values are typically expressed as molar concentration. According to the Food and Drug Administration (FDA), IC 50 represents the concentration of a drug required for 50% inhibition in vitro.
  • FDA Food and Drug Administration
  • an agent that inhibits CDK19 activity but does not significantly inhibit activity of CDK8 has an IC 50 that is at least about 2-fold, 5-fold, 10- fold, 50-fold, 75-fold, or 100-fold, lower than the concentration of the agent required to effect CDK8 activity under the same conditions.
  • the IC 50 for the agent to inhibit CDK19 activity is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the IC 50 for the agent to inhibit the activity of CDK8.
  • the effect of the agent on CDK19 and CDK8 activity can be determined by calculating the equilibrium dissociation constant (K D ) of the agent to each CDK.
  • K D equilibrium dissociation constant
  • an agent that inhibits the activity of CDK19 but does not significantly inhibit activity of CDK8 has a K D that is at least about 2-fold, 5-fold, 10- fold, 50-fold, or 100-fold lower than the K D of the agent to CDK8 under the same conditions.
  • the K D for the agent (to CDK19) is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the Ko for the agent (to CDK8).
  • the K D is lower for the agent to CDK19 as compared to the K D of the agent to CDK8.
  • the equilibrium dissociation constant of the agent (to CDK8) is greater than the equilibrium dissociation constant of the agent (to CDK19).
  • the agent can include an antibody having a K D value in the micromolar (10 -6 ) to nanomolar (10 -7 to 10 -9 ) range.
  • the agent can include an antibody having a K D in the nanomolar range (10 -9 ) to the picomolar (10 -12 ) range.
  • the agent can have a nanomolar (nM) equilibrium dissociation constant to CDK19 and a micromolar ( ⁇ M) equilibrium dissociation constant to CDK8.
  • nM nanomolar
  • ⁇ M micromolar
  • the effect of the agent on CDK19 and CDK8 activity can be determined by calculating the inhibitor constant (K l ) of the agent to each CDK.
  • K l is an indication of how potent an inhibitor is; it is the concentration required to produce half maximum inhibition.
  • the lower the Ki the greater the binding affinity between the agent and the CDK gene.
  • an agent that inhibits the activity of CDK19 but does not significantly inhibit activity of CDK8 has a K l that is at least about 2-fold, 5-fold, 10- fold, 50-fold, 75-fold, or 100-fold lower than the K l of the agent (to CDK8) under the same conditions.
  • the K l for the agent to CDK19 is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the K l for the agent to CDK8.
  • the K l is lower for the agent to CDK19 as compared to the K l of the agent to CDK8.
  • the inhibitor constant of the agent to CDK8 is greater than the inhibitor constant of the agent to CDK19.
  • an agent that inhibits activity of CDK19 can bind to CDK19 and significantly inhibit CDK19 activity in vivo or in vitro.
  • a CDK19 inhibitor can be identified by its ability to preferentially bind to CDK19 and fully inhibit activity of CDK19. Inhibition of CDK19 occurs when CDK19 activity, when exposed to an agent of the invention, is at least about 70% less, for example, at least about 75%, 80%, 90%, 95%, 96%, 97%, 98%, 99% less, or totally inhibited, in comparison to CDK19 activity in the presence of a control or in the absence of the agent. No significant decrease in CDK8 activity occurs when, CDK8 activity upon exposure to the agent, is at least about 90%, for example, at least 95%, 96%, 97%, 98%, 99%, or 100%, in comparison to CDK8 activity in the absence of the agent.
  • an agent that inhibits activity of CDK19 to a greater extent than it inhibits activity of CDK8 refers to an agent that is capable of binding and inhibiting the activity of CDK19 significantly more than the agent’s effect on inhibiting the activity of CDK8 under the same conditions.
  • an agent that inhibits activity of CDK19 to a greater extent than inhibiting the activity of CDK8 occurs when CDK19 activity, when exposed to an agent of the invention, is at least about 10% less, for example, at least about 15%, 20%, 30%, 40%, or 50% less, than the activity of CDK8 under the same conditions in vitro or in vivo.
  • an agent inhibits the activity of CDK19 to a greater extent than the activity of CDK8, when the activity of CDK19 observed is at least 10% less than the activity of CDK8 under the same conditions. In another embodiment, an agent inhibits the activity of CDK19 to a greater extent than CDK8 activity, when at least 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold less CDK19 activity is observed as compared to CDK8 activity under the same conditions.
  • the extent of inhibition can be determined using one or more methods known in the art, including but not limited to assays described herein in the Examples section of the specification and for example, “Percent Of Control (POC)” or “Normalized Percent Inhibition (NPI)”.
  • POC and NPI are methods that normalize data and are often used when comparing multiple agents (e.g., various antibodies or small molecules) against multiple targets (e.g., CDK19 and CDK8).
  • POC is a method that corrects for plate-to-plate variability (for example in high-throughput drug screening) by normalizing an agent’s measurement relative to one or more controls present in the plate.
  • Raw measurements for each agent can be divided by the “average” of within-plate controls.
  • NPI is a control-based method in which the difference between the agent measurement and the mean of the positive controls is divided by the difference between the means of the measurements on the positive and the negative controls (Malo et al., Nature Biotechnology , Vol. 24, 167-175 (2006)). By normalizing the extent of inhibition observed, accurate conclusions can be made regarding which agent(s) are effective at inhibiting the activity of each target under investigation.
  • the patient is treated with a combination therapy comprising an agent that inhibits expression or activity of CDK19 and (a) radiation therapy and/or chemotherapy.
  • radiation or chemotherapy eliminates the bulk of the tumor mass and the CDK19 inhibitor reduces the number of viable cancer stem cells (e.g., EpCAM med/high /CD10 -/low ) cells.
  • the chemotherapy comprises administration of an anthracycline (e.g., Doxorubicin or Epirubicin), a taxane (e.g., Paclitaxel or Docetaxel), an anti-metabolite (e.g., Capecitabine or Gemcitabine), a platinum agent (e.g., Carboplatin or Cisplatin), Vinorelbine, or Eribulin.
  • anthracycline e.g., Doxorubicin or Epirubicin
  • a taxane e.g., Paclitaxel or Docetaxel
  • an anti-metabolite e.g., Capecitabine or Gemcitabine
  • a platinum agent e.g., Carboplatin or Cisplatin
  • Vinorelbine e.g., or Eribulin.
  • a course of therapy with the CDK19 inhibitor will have a beneficial outcome for the patient, including, for example, a reduction in tumor volume, a reduction in metastases, and a reduction in tumor cells having the phenotype EpCAM med/high and CD10 -/low .
  • Tumor volume may be measured using art-known methods. See, e.g., Wapnir et al., Breast Cancer Res Treat 41:15-19, 1996; Sapi et al., PLoS One 10:e0142190, 2015. Tumor volume may be reduced by at least 10%, optionally at least 20% and sometimes by at least 50% after a course of treatment with a CDK19 inhibiting agent as monotherapy or in combination with other agent(s) or treatments. In some embodiments, the reduction in tumor volume (e.g., at least 10%, 20%, or 30% reduction in tumor volume) may be observed as soon as within 1 month of initiating therapy.
  • the reduction in tumor volume (e.g., at least 10%, 20%, 30%, 40%, 50%, or 60% reduction in tumor volume) may be observed within 2, 3, 4, 5, or 6 months of initiating therapy.
  • the methods described herein to treat TNBC may also slow down or inhibit the further growth of a tumor.
  • a patient receives combination therapy and a therapeutic benefit is observed that exceeds that of monotherapy with the second agent.
  • a reduction in metastases in an individual may be determined as described in Makela et al., Sci Rep. 7:42109, 2017 and may be observed in a population according to standard methodology.
  • the presence or amount of cancer cells having the expression profile EpCAM med/high and CD10 -/low in a TNBC tumor tissue obtained from a subject may be used to predict or assess the therapeutic responsiveness of the subject to treatments that target the CDK19 gene or its corresponding protein.
  • cells having the expression profile EpCAM med/high /CD10 -/low have a high tumor initiating capacity and are also enriched in CDK19.
  • subjects having a high percentage of EpCAM med/high and CD10 -/low TNBC cells may be especially responsive.
  • the likely therapeutic responsiveness of a subject with TNBC to a CDK19 targeting agent is determined by (a) quantitating EpCAM med/high /CD10 -/low cells in a tumor sample obtained from the subject; (b) comparing the quantity of EpCAM med/high /CD10 - /low cells in (a) to a reference value characteristic of tumors responsive to a CDK19 targeting therapy, and treating the patient with an inhibitor of CDK19 expression or activity if the quantity of EpCAM med/high /CD10 -/low cells is equal to or exceeds the reference value.
  • the reference value can be determined by quantitating EpCAM med/high /CD10 -/low cells in healthy and TNBC populations and calculating statistically significant ranges characteristic of healthy and tumor tissues.
  • tumor tissue and healthy tissue from the same subject can be tested, and subjects with elevated EpCAM med/high /CD10 -low cells in tumor relative to healthy tissues can be identified as likely to respond to CDK19 targeted therapy.
  • compositions used in methods described herein may include an active ingredient and one or more pharmaceutically acceptable carriers or excipients, which can be formulated by methods known to those skilled in the art.
  • a pharmaceutical composition of the present invention includes, in a therapeutically effective amount, a DNA or RNA oligonucleotide that decreases the expression level of the CDK19 gene.
  • a pharmaceutical composition of the present invention includes, a pharmaceutical composition of the present invention includes a DNA or RNA oligonucleotide in a therapeutically effective amount, a small molecule that inhibits the activity of CDK19.
  • the therapeutically effective amount of the active ingredient in a pharmaceutical composition is sufficient to prevent, alleviate, or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is within the capability of those skilled in the art.
  • a pharmaceutical composition of the present invention is formulated as a depot preparation.
  • depot preparations are typically longer acting than non-depot preparations.
  • such preparations are administered by implantation (for example subcutaneously) or by intramuscular injection.
  • depot preparations are prepared using suitable polymeric or hydrophobic materials (for example an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical composition may include a delivery system.
  • delivery systems include, but are not limited to, exosomes, liposomes, and emulsions.
  • an active ingredient may be loaded or packaged in exosomes that specifically target a cell type, tissue, or organ to be treated.
  • Exosomes are small membrane-bound vesicles of endocytic origin that are released into the extracellular environment following fusion of mutivesicular bodies with the plasma membrane. Exosome production has been described for many immune cells including B cells, T cells, and dendritic cells. Techniques used to load a therapeutic compound into exosomes are known in the art and described in, e.g., U.S. Pat. Publication Nos.
  • therapeutic compounds may be loaded into exosomes by electroporation or the use of a transfection reagent (i.e., cationic liposomes).
  • a transfection reagent i.e., cationic liposomes
  • an exosome-producing cell can be engineered to produce the exosome and load it with the therapeutic compound.
  • exosomes may be loaded by transforming or transfecting an exosome-producing host cell with a genetic construct that expresses the active ingredient (i.e., a DNA or RNA oligonucleotide), such that the active ingredient is taken up into the exosomes as the exosomes are produced by the host cell.
  • a targeting moieties may be introduced into exosomes, so that the exosomes can be targeted to a selected cell type, tissue, or organ.
  • Targeting moieties may bind to cell-surface receptors or other cell-surface proteins or peptides that are specific to the targeted cell type, tissue, or organ.
  • exosomes have a targeting moiety expressed on their surface.
  • the targeting moiety expressed on the surface of exosomes is fused to an exosomal transmembrane protein.
  • Techniques of introducing targeting moieties to exosomes are known in the art and described in, e.g., U.S. Pat. Publication Nos. US 20130053426 and US 20140348904, and International Patent Publication No. WO 2015002956, which are incorporated herein by reference.
  • CCT152921 is4-[(2-Phenylethyl)amino]-6-quinazolinecarbonitrile (NIH NCAT).
  • NIH NCAT n-butyl-N-butyl-N-butyl-N-butyl-N-butyl-N-butyl-N
  • Pairs of complementary ssDNA oligonucleotides containing the sense target sequence, a 15-mer loop sequence (5′-GTTAATATTCATAGC-3′ SEQ ID NO: 19), and the reverse complement of the sense sequence were synthesized (Elim Biopharmaceuticals).
  • the oligonucleotides were annealed in 50 ⁇ M annealing buffer (10 mM Tris-HCl pH 8.0, 50 mM NaCl, 1 mM EDTA).
  • the double-stranded DNA oligo templates were subsequently cloned into the pRSI12-U6-(sh)-HTS4-UbiC-TagRFP-2A-Puro shRNA expression vector (Cellecta) digested with Bbsl for constitutively active shRNA vector constructs and pRSITUR-U6Tet-(sh)-UbiC-TetRep-2A-TagRFP digested with Bbsl for inducible shRNA vector constructs.
  • the sense strands in the shRNA vectors used in this study were: 5′-GCG AGA ATT GAA GTA CCT TAA-3′ (shCDK19-1 (SEQ ID NO: 1)), 5′-ACC AGC AAA TAT CCT AGT AAT-3′ (shCDK19-2 (SEQ ID NO:2)), and 5′-GCA GGG TAATAA CCA CATTAA-3′ (shCDK8-2 (SEQ ID NO: 3)).
  • the unmodified pRSI12-U6-(sh)-HTS4-UbiC-TagRFP-2A-Puro shRNA expression vector above was used as the ‘empty’ control shRNA.
  • the pHIV-ZsGreen expression vector (Addgene) was used to produce GFP positive tumor cells.
  • the DECIPHER 27 K Pooled shRNA lentivirus library - Human Module 1 (Cellecta) used for the RNAi screen contains 27,500 unique shRNA constructs targeting 5,043 human genes (approximately five or six redundant shRNAs per gene) in the same pRSI12 shRNA expression vector.
  • MDA-MB231, MDA-MB468, HS578T, and 293T cells were obtained from ATCC.
  • HMEC cells were obtained from ThermoFisher Scientific. These cells were certified by the vendors to be mycoplasma free. None of the cell lines used are listed in the database of commonly misidentified cell lines maintained by ICLAC. All cell lines used were passaged less than 10 times from when the original cells from the vendors were thawed. All MDA-MB231, MDA-MB468, 293T, and HS578T cells were grown in DMEM (Invitrogen) supplemented with PSA (Life Technologies), 10% FBS (Hyclone), Glutamax (ThermoFisher Scientific), and sodium pyruvate (Life Technologies). HMEC cells were grown in HuMEC Ready Medium (ThermoFisher Scientific).
  • mice Nod scid gamma (NSG) mice (NOD.Cg-Prkdc scid IL2Rg tm1Wjl /SzJ) were purchased from the Jackson Laboratory.
  • Mice used for PDX experiments were adult female mice between 8 and 10 weeks old. All the mice used in this study were maintained at the Stanford Animal Facility in accordance with a protocol approved by the Stanford University APLAC committee. Mice were maintained in-house under aseptic sterile conditions. Mice were administered autoclaved food and water.
  • mice were provided rodent feed containing 625 mg Doxycycline hyclate/kg diet (Envigo) in place of their normal rodent diet.
  • Xenografts were mechanically chopped with a razor blade to approximately 1 mm pieces and then incubated at 37°-C for 3 to 4 hours with collagenase and hyaluronidase (Stem Cell Technologies) in Advanced DMEM/F12 (Invitrogen) with 120 ⁇ g/mL penicillin, 100 ⁇ g/mL streptomycin, 0.25 ⁇ g/mL amphotericin-B (PSA) (Life Technologies). Cells were then treated with ACK lysis buffer (Gibco) to lyse red blood cells, followed by 5 mins of treatment with pre-warmed dispase (Stem Cell Technologies) plus DNAsel (Sigma) and filtered through a 40 ⁇ m nylon mesh filter. Cells were finally washed with flow cytometry buffer (HBBS, 2% FCS, PSA).
  • flow cytometry buffer HBBS, 2% FCS, PSA
  • PDX tumors were dissociated into single cells, the number of live cells were determined with Trypan blue staining and manually counted with a hemocytometer.
  • Cells were resuspended with flow cytometry buffer to a concentration of 10 6 live cells/mL and incubated 1:50 (v/v) with Biotin anti-human CD326 (EpCAM) antibody (Biolegend) for 20 mins at 4°-C.
  • EpCAM Biotin anti-human CD326
  • Cells were washed with flow cytometry buffer and then resuspended to 80 ⁇ L and incubated with 20 ⁇ L anti-biotin microbeads (Miltenyi Biotec) for 20 mins at 4°-C.
  • Lentivirus was produced with Packaging Plasmid Mix (Cellecta) and subcloned pRSl12 shRNA expression plasmids using Lipofectamine 2000 (Thermofisher Scientific) in 293T cells per manufacturer’s instructions. Supernatants were collected at 48 h and 72 h, filtered with a 0.45 ⁇ m filter and precipitated with Lentivirus Precipitation Solution (Alstem LLC) per manufacturer’s instructions. Virus was resuspended in 1/100 original volume. Viral titers were determined by flow cytometry analyses of 293T cells infected with serial dilutions of concentrated virus.
  • Organoid media consisted of: Advanced DMEM/F12 (Invitrogen), 10% FBS (Hyclone), 2.5% growth factor-reduced Matrigel (BD), 10 ng/mL mouse EGF (R&D), 100 ng/mL Noggin (R&D), 250 ng/mL RSPO-I (R&D), 1X B27 (Invitrogen), 1X N2 (Invitrogen), and PSA (Life Technologies). Cells were then spinoculated by centrifuging at 15 oC for 2 hours at 1200xg. Cells were resuspended by pipetting and left overnight in 48-well ultra-low attachment cell culture plates (Corning).
  • cells were transferred the next day to matrigel.
  • in vivo PDX assays approximately 75% of the cells were injected into NSG mice as described in the PDX tumor engraftment section. The remainder 25% of cells were plated on matrigel and grown in organoid media for 72 hours until the cells became RFP positive. At that point media was removed and exchanged for dispase and incubated for 2-3 h until the matrigel dissolved. Dissociated cells were resuspended in flow cytometry buffer and analyzed by flow cytometry to determine the ‘baseline’ RFP percentage for cells that were injected into the mice.
  • Irradiated L1-Wnt3a feeder cells (generous gift of Dr. Roel Nusse) were mixed with growth factor reduced matrigel (BD Biosciences) and allowed to solidify at 37 oC.
  • Single cell suspensions of PDX tumor cells were transferred onto the solidified matrigel/feeder cell mix substrate and grown in organoid media. Cells were grown for approximately 2 weeks in a 37 oC incubator with 5% CO 2 . 50% of media was exchanged with fresh media every 3-4 days. Colonies were counted under fluorescence microscopy. Only RFP positive colonies (which represent transduced cells) were counted.
  • doxycycline hyclate was added to a final concentration of 100 ng/mL into the media.
  • WST-1 Cell Proliferation Reagent (Roche) was added at 1:10 (v/v) final dilution to each well per manufacturer’s instructions. Cells were subsequently incubated at 37 oC and 5% CO 2 . Between 1 and 4 hours after addition of reagent, plates were analyzed on a SpectraMax M3 Bioanalyzer (Molecular Devices). Absorbance for each well was measured at 450 nm (signal wavelength) and 650 nm (reference wavelength). Thus, the signal for each experimental sample was Absorbance experimental (A 450nm -A 650nm ).
  • Absorbance background (A 450nm -A 650nm ) was obtained by measuring absorbance in a blank well.
  • a corrected Absorbance experimental -Absorbance background .
  • All A corrected values for the knockdowns were normalized to the A corrected value for the control sample to obtain a ‘Relative Viability’.
  • TaqMan Gene Expression Master Mix (Applied Biosystems) and the following TaqMan Gene Expression Assays (Applied Biosystems) were used following manufacturer’s instructions: ACTB, Hs00357333_g1; CDK19, Hs01039931_m1; CDK8, Hs00993274_m1. Data was collected on a 7900HT Fast Real-Time PCR System (Applied Biosystems) and data analyzed with SDS 2.4 software (Applied Biosystems). Gene expression data in each sample was normalized against the expression of beta-actin.
  • Single cell suspensions of PDX cells were resuspended in 50% (v/v) mixtures of normal matrigel (BD Biosciences) and flow cytometry buffer in a total volume of 50-100 ⁇ L.
  • BD Biosciences normal matrigel
  • flow cytometry buffer in a total volume of 50-100 ⁇ L.
  • cells were injected subcutaneously into the nipple of female NSG mice at the fourth abdominal fat pad.
  • the specific number of cells injected into the mice were determined by flow cytometry and secondarily by manual counting with a hemocytometer.
  • PDX tumors were detected by palpation.
  • Tumor volumes were determined by measuring the length (l) and width (w) and calculating volumes using the ellipsoid formula 1 ⁇ 6 x l x w 2 x ⁇ . Tumors volumes and mice weights were determined twice per week.
  • mice Xenograft tumors and mice lungs were surgically resected after the mice were euthanized. A 3 to 4 mm section is cut from each tumor and saved in ice cold PBS for imaging. The mice lungs and tumors were imaged on a M205FA Fluorescence Stereo Microscope (Leica) and images were captured with a DFC310FX camera (Leica).
  • Flow cytometry was performed with a 100 ⁇ m nozzle on a Flow Cytometry Aria II (BD Biosciences) with Diva software (BD Biosciences). Data analysis was performed using Flowjo software (Flowjo). For all experiments, side scatter and forward scatter profiles (area and width) were used to eliminate debris and cell doublets. Dead cells were eliminated by excluding 4′,6-diamidino-2-phenylindole (DAPI)-positive cells (Molecular Probes). For PDX tumor cells, they were gated for GFP positivity and then for RFP positivity. RFP percentage is the percentage of GFP positive cells that are also RFP positive.
  • DAPI 4′,6-diamidino-2-phenylindole
  • RFP fraction that is: the RFP % in the tumor divided by the baseline RFP % (see ‘Lentivirus infection’ section).
  • RFP fraction for each sample is then normalized to the RFP fraction for the shRNA control sample which is set at 100% to obtain the ‘Normalized % RFP’.
  • GFP positive PDX-T1 tumors grown in NSG mice were dissected, processed to single cells, and enriched with EpCAM as described previously. Analysis of cells at this point showed that they were approximately 98%-100% GFP positive.
  • RNAi dropout viability screen 60 million dissociated PDX-T1 cells were transduced with the DECIPHER 27 K Pooled shRNA lentivirus library-Human Module 1 (Cellecta) at an MOI of 1 in the presence of polybrene and then spinoculated for 2 hours as described previously. The next day, half the cells were spun down and frozen as the in vitro baseline reference sample. A small number of cells were plated separately in organoid colony formation conditions to determine lentiviral infection percentage after 72 hours (cells were found to be approximately 80% RFP positive).
  • the remainder of the cells were plated into twelve 150 mm dishes prepared with 12 mL matrigel containing irradiated L1-Wnt3a feeder cells at 250,000 cells/mL of matrigel.
  • the cells were grown for 19 days with an exchange for fresh media every 3-4 days. On the final day, all the media was exchanged with dispase in order to dissolve the matrigel and to recover the cells.
  • the cells from all the plates were pooled, washed, and frozen as the in vitro organoid growth experimental sample.
  • RNAi dropout viability screen 30 million dissociated PDX-T1 cells were transduced with the DECIPHER 27 K Pooled shRNA lentivirus library-Human Module 1 (Cellecta) at an MOI of 1.25 in the presence of polybrene and then spinoculated for 2 hours as described previously. The next day, half the cells were spun down and frozen as the in vivo baseline reference sample. A small number of cells were plated separately in organoid colony formation conditions to determine lentiviral infection percentage after 72 hours (cells were found to be approximately 70% RFP positive).
  • mice The remainder of the cells were resuspended in 50% (v/v) mixtures of normal matrigel (BD Biosciences) and flow cytometry buffer in a total volume of 1.8 mL. These cells were injected evenly into the right and left mammary fat pads of seventeen NSG mice. When tumors reached approximately 10 mm in diameter, the mice were euthanized and the tumors dissected as previously described. These tumors were then processed into single cells, pooled, washed, and frozen as the in vivo growth experimental sample.
  • BD Biosciences normal matrigel
  • Sections of the PDX tumors were fixed in formalin overnight and then transferred to 70% ethanol. Samples were then embedded in paraffin and sectioned for histology. Formalin fixed paraffin embedded sections were de-parafinized in xylene and rehydrated in an ethanol gradient. Antigen retrieval was performed in a Tris-EDTA buffer by heating in a microwave for 20 min. The primary antibodies, polyclonal Rabbit anti-CDK19 (Sigma) and polyclonal chicken anti-CDK8 (Novus Biologicals), were diluted 1:50 and 1:100, respectively, in TBS + 1% BSA before applying to samples overnight.
  • EpCAM enriched PDX-T1 cells were infected with shCDK19-2, shCDK8-2 or control shRNA and grown in organoid culture conditions for 72 hours. They were subsequently recovered from matrigel with dispase, resuspended in flow cytometry buffer and sorted by flow cytometry to obtain cells that were both GFP and RFP positive. RNA was extracted from these cells by RNeasy plus micro kit (Qiagen) according to manufacturer’s instructions and quantified on an Agilent 2100 Bioanalyzer. 50 ng of total RNA from each sample was used. In vitro transcription, fragmentation, labeling, hybridization to the microarray and scanning was performed by the Stanford Protein and Nucleic acid facility (PAN facility).
  • PAN facility Stanford Protein and Nucleic acid facility
  • ChIP assays were performed as described in, e.g., Zarnegar et al., Nucleic Acids Research, gkx648, July, 2017. Approximately 250,000 to 500,000 MDA-MB231 cells were used per ChIP. 1 ⁇ g of anti-H3K27ac (Active Motif #39133) were used per ChIP.
  • ChIP enriched DNA was quantified using a Qubit 3.0 and dsDNA HS assay. Up to 1 ng of DNA was used for library construction using transposition based NEXTERA XT (followed manufacturer’s protocol with ⁇ 14 PCR cycles for indexing). Indexed samples were pooled and submitted for sequencing on a NextSeq500 to obtain 75 bp single end reads with read depths of ⁇ 60 million reads.
  • DiffBind The DiffBind output peak list was annotated by fetching the nearest nonoverlapping feature of the human RefSeq table from UCSC. Data for aggregation plots of ChIP signal across various peaks sets were generated using DeepTools′ computeMatrix (scale-regions: 1000; 50 bp bins) and plotProfile. Data was then plotted with GraphPad Prism software.
  • GSEA Gene set enrichment analysis
  • Results are shown as mean ⁇ s.d. Statistical calculations were performed with GraphPad Prism software (GraphPad Software Inc). Variance was analyzed using the F-test. To determine P-values, t-test was performed on homoscedastic populations, and t-test with Welch correction was applied on samples with different variances. For animal studies, sample size was not predetermined to ensure adequate power to detect a pre-specified effect size, no animals were excluded from analyses, experiments were not randomized and investigators were not blinded to group allocation during experiments.
  • RNAi dropout viability screens were performed using a 27,500 shRNA library targeting 5000 genes in PDX-T1, a TNBC PDX ( FIG. 15 ).
  • the screens were performed in two different formats, in vitro as organoid cultures and in vivo as PDXs in nod scid gamma (NSG) mice ( FIG. 1 A ).
  • the abundance of individual shRNA in each experimental sample and the baseline reference samples were determined by high throughput sequencing of the shRNA barcodes. The goal was to identify genes whose knockdown by shRNA inhibited the growth of PDX tumor cells across different experimental conditions.
  • FIGS. 5 A and 5 B are graphs showing the shRNA counts in the in vivo growth experimental sample ( FIG. 5 A ) and in the in vitro growth experimental sample ( FIG. 5 B ) versus the shRNA counts in the baseline sample. Control shRNA targeting luciferase (light gray dots) and shRNA targeting CDK19 (dark gray dots) are highlighted. All other shRNA are shown as black dots (each experiment performed once).
  • the final candidate list was restricted to genes with the lowest 5% of shRNA ratios in each screen that were targeted by more than two shRNAs and were also identified both in vitro and in vivo ( FIG. 5 C ). This resulted in the identification of 46 candidate genes ( FIG. 5 D ).
  • CDK19 was chosen because data from the Cancer Genome Atlas (TCGA) showed that CDK19 copy number amplifications and mRNA upregulation were more prevalent in TNBC patient samples (23%) compared to samples from other breast cancer subtypes (see, e.g., Cancer Genome Atlas Research, N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45:1113-1120, 2013; FIG. 6 A ). Additionally, high CDK19 expression has been reported to correlate with poor relapse free survival in breast cancer patients (see, e.g., Broude et al., Current cancer drug targets 15, 739-749, 2015 and Porter et al., Proc Natl Acad Sci U S A 109: 13799-13804, 2012).
  • CDK19 belongs to a subset of the CDK family that is reportedly more associated with regulation of RNA polymerase II (RNAPII) transcription than cell cycle progression.
  • CDK19 and its paralog, CDK8, can both form the CDK module (CKM) by binding with three other proteins: MED12, MED13, and Cyclin C.
  • the presence and nuclear localization of CDK19 in our PDX cells were confirmed by immunofluorescence ( FIG. 6 B ).
  • FIGS. 6 A and 6 B the percentage shows the percentage of samples with CDK19 copy number amplifications or CDK19 mRNA upregulation in triple-negative, HER2 positive, estrogen receptor positive, and all breast cancers. The fractions show the number of positive samples and total samples in each group. Data obtained from cBioPortal (see, e.g., Gao et al., SciSignal 6, pl1, 2013).
  • FIGS. 7 A and 7 B demonstrate that CDK19 knockdown significantly decreased the viability of TNBC cells (viability of MDA-MB231 cells, ****P ⁇ 0.0001 ( FIG. 1 B ), MDA-MB468 cells, ***P ⁇ 0.001; ****P ⁇ 0.0001 ( FIG.
  • CDK19 knockdown ( FIG. 7 C ) also inhibited the formation of organoid colonies ( FIG. 1 E ).
  • HMEC human mammary epithelial cells
  • PDX-T1, PDX-T2, and PDX-T3 were derived from chemotherapy naive patients ( FIG. 15 ).
  • all PDX tumor cells were first labeled with green fluorescent protein (GFP) and cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP).
  • GFP green fluorescent protein
  • RFP red fluorescent protein
  • CDK19 knockdown led to a significant reduction in the percentage of RFP positive cells in tumors from all three TNBC PDXs ( FIGS. 1 G- 1 I and FIG. 1 M ). Tumor growth was monitored and tumors were analyzed when they exceeded 17 mm.
  • FIG. 1 M shows representative images of PDX-T1 tumors transduced with control shRNA (top row), shCDK19-1 (middle row), or shCDK19-2 (bottom row).
  • Bright field images show gross tumor morphology
  • FITC images (middle column) identify tumor cells labeled with GFP
  • Texas-Red images (right column) identify shRNA-transduced cells labeled with RFP.
  • CDK19 is critical for tumor growth in vivo.
  • FIG. 1 I , n 3, experiment performed once).
  • CDK19 knockdown eliminated the detection of any lung metastases by those cells ( FIG. 1 K and FIG. 1 N ).
  • FIG. 1 N bright field images (left column) show gross lung morphology
  • FITC images (middle column) identify metastatic tumor cells labeled with GFP
  • Texas-Red images (right column) identify shRNA-transduced metastatic cells labeled with RFP.
  • CDK19 knockdown inhibited both the growth of the PDX ( FIG. 1 J ) and the lung metastases in these mice ( FIG. 1 L and FIG. 7 D ). These data show that in vivo, CDK19 knockdown not only affected primary tumor growth, but also inhibited tumor metastasis.
  • TICs tumor initiating cells
  • CD10 with EpCAM to FACS-sort breast cancer PDXs.
  • EpCAM can separate PDX cells into three distinct sub-populations, EpCAM med/high /CD10 -/low , EPCAM low/med /CD10 low/+ , and EpCAM - /CD10 - ( FIG. 2 A , right).
  • EpCAM med/high /CD10 -/low EpCAM low/med /CD10 low/+
  • EpCAM - /CD10 - FIG. 2 A , right.
  • EpCAM med/high /CD10 -/low gate (1)
  • EPCAM low/med /CD10 low/+ gate (2)
  • EpCAM - /CD10 - gate (3)
  • organoid colony formation assays in vitro and transplantation limiting dilution assays (LDA) in vivo.
  • LDA transplantation limiting dilution assays
  • injection of EpCAM med/high /CD10 -/low cells from all six PDXs consistently formed tumors ( FIG. 2 C ), sometimes with the transplant of as little as 100 cells (PDX-T1 and PDX-T2).
  • transplant of EPCAM low/med /CD10 low/+ cells only formed tumors in two PDXs (PDX-T1 and PDX-T2), and only when transplanting high cell numbers (i.e. 2500 cells) ( FIG. 2 C ). Furthermore, no tumors formed from the transplant of EpCAM - /CD10 - cells from any PDX. Hence, TIC’s are enriched in the EpCAM med/high /CD10 -/low sub-population of all PDX breast tumors we examined.
  • CDK19 expression was enriched in the more tumorigenic EpCAM med/high /CD10 -/low cells compared to the less tumorigenic EPCAM low/med /CD10 low/+ cells.
  • CDK19 expression was higher in the more tumorigenic EpCAM med/high /CD10 -/low cells compared to the less tumorigenic EPCAM low/med /CD10 low/+ cells ( FIGS. 2 D- 2 G ). To generate the data in FIGS.
  • FIG. 8 B the relative expression of CDK19 in doxycycline treated inducCDK19KD-PDX-T1 cells is normalized to the mean expression of CDK19 in control inducCDK19KD-PDX-T1 cells.
  • inducCDK19KD-PDX-T1 cells were injected into the mammary fat pads of NSG mice at 50, 250 and 1250 cells. Mice in the doxycycline group were fed a doxycycline containing rodent feed to induce CDK19 shRNA, while mice in the control group were fed a normal rodent diet. Tumors were detected by palpation of tumors.
  • the tumor initiating frequencies significantly decreased from 1 in 342 cells (95%Cl: 1 in 828 to 1 in 142) in the control (No Dox) group to 1 in ⁇ cells (95%Cl: 1 in ⁇ to 1 in 2587) in the CDK19 knockdown (+Dox) group ( FIG. 8 D ).
  • Both the significant decrease in tumor initiating frequency caused by CDK19 knockdown and CDK19′s higher expression in the TIC sub-population suggests that TIC inhibition is likely responsible for the impaired tumor growth observed with CDK19 knockdown.
  • CDK8 has been shown to play a role in a variety of malignancies including colon cancer, acute myeloid leukemia, and melanoma. Higher expression of CDK8 has been associated with worse prognosis in colon cancer (Firestein et al., Nature 455:547-551, 2008). CDK8 knockout in embryonic stem cells was shown to prevent embryonic development (Porter et al., Proc Natl Acad Sci USA, 109:13799-13804, 2012) due to its essential role in the pluripotent stem cell phenotype.
  • CDK8 may include positive regulation of Wnt/ ⁇ - catenin pathway, growth factor-induced transcription, and TGFP signaling.
  • CDK8 has also been shown to either negatively or positively regulate transcription.
  • CDK19 may function differently from CDK8.
  • In vitro studies showed that CDK19 and CDK8 participate mutually exclusively of each other in binding to other CKM components, while gene knockdown studies in cell lines of cervical cancer and colon cancer showed that CDK19 and CDK8 regulate different genes.
  • Our goal was to investigate in TNBC whether CDK19 and CDK8 have distinct biological functions by examining global gene expression changes resulting from targeted knockdown of CDK19 or CDK8.
  • CDK19 knockdown affected 3909 genes and CDK8 knockdown affected 4233 genes ( FIG. 3 A ). However, only 12% of upregulated and 5% of downregulated genes in the CDK19 knockdown experiment were also affected by CDK8 knockdown. This suggested that CDK19 and CDK8 largely regulate distinct genes ( FIG. 3 A ).
  • GSEA Gene set enrichment analysis of the CDK19 and CDK8 knockdown genes allowed us to identify enriched Hallmark gene sets amongst the most upregulated or downregulated genes ( FIG. 3 B and FIG. 10 ).
  • the Hallmark gene sets uniquely enriched in the knockdown of CDK19 or CDK8 are shown in black, enriched in both the knockdown of CDK19 and CDK8 are marked by “*” and enriched by genes expressed in opposite directions between the knockdown of CDK19 and CDK8 are marked by “**”.
  • Normalized enrichment scores and FDR q-value are determined by the GSEA software. An FDR cutoff of ⁇ 0.25 was used to select significant Hallmarks.
  • genes associated with early estrogen response, epithelial to mesenchymal transition (EMT), cholesterol homeostasis, MYC pathways (Myc Targets v2), interferon alpha response, and fatty acid metabolism changed in the opposite direction in response to knockdown by CDK19 compared to CDK8 ( FIG. 3 B , boxes), which suggests a counter-regulatory relationship exists between CDK19 and CDK8.
  • Hallmark gene sets enriched by the expression of genes in opposite directions by CDK19 knockdown compared to CDK8 knockdown are boxed. A number of the Hallmark gene sets were only enriched in the genes that uniquely changed due to CDK19 knockdown ( FIG. 3 B , left region).
  • Hallmarks reflected by these gene sets included P53 signaling, KRAS signaling, androgen response, NOTCH signaling, TGF BETA signaling, and IL6-JAK-STAT3 signaling, which may be potential biological pathways for targeted therapies for TNBC. All of these biological pathways represent active areas of clinical investigation in the evaluation of targeted therapies for TNBC. Consistent with our findings, a number of the pathways found enriched in our CDK19 knockdown experiments, such as cholesterol homeostasis, P53 signaling, mitosis, and NF K B pathways have been shown previously in other cell types to also be regulated by CDK19.
  • CDK19 and CDK8 have the potential to co-regulate certain pathways, while counter-regulating others.
  • CDK19 like CDK8, is capable of positively or negatively regulating biological pathways.
  • the multitude of clinically relevant TNBC pathways regulated by CDK19 suggests that targeting CDK19 can provide the opportunity to modulate multiple pathways simultaneously and at the same time, avoid potential toxicity because of the advantageous limited tissue distribution of CDK19.
  • This approach could overcome the resistance to single agent therapy commonly seen in TNBC and also potentially enable the targeting of ‘undruggable’ processes such as those involving P53 or MYC.
  • CDK19 and CDK8 have highlighted the role of CDK19 and CDK8, as well as other transcriptional CDKs (CDK7, CDK12/CDK13), in regulating the transcription of critical oncogenic genes by acting at large clusters of enhancers (also called ‘super-enhancers’) that are marked by histone 3 lysine 27 acetylation (H3K27Ac).
  • enhancers also called ‘super-enhancers’
  • H3K27Ac histone 3 lysine 27 acetylation
  • CDK19 and CDK8 can also regulate the epigenetic modifications at enhancer sites as a mechanism to control gene expression. While enhancer modification through other signaling pathways have been identified, this mechanism of gene control has not yet been reported for the CDKs.
  • chromatin immunoprecipitation and sequencing (CHIP-Seq) for the H3K27Ac modification was performed on MDA-MB231 cells under three different conditions: Control (empty vector transduction), CDK19 knockdown, and CDK8 knockdown.
  • Control empty vector transduction
  • CDK19 knockdown CDK19 knockdown
  • CDK8 knockdown Genome-wide analysis of all H3K27Ac modified regions showed that both CDK19 knockdown and CDK8 knockdown had similar global H3K27Ac levels compared to control ( FIG. 11 ).
  • H3K27Ac CHIP-Seq signals across all identified H3K27Ac peak regions are normalized to 1-Kb and centered on the middle of those regions. Signals of the flanking 2-Kb regions are also shown.
  • the total signal of each biological replicate was determined by summing the signals of each 50-base window 1-Kb around the center of each region. P-values between total CHIP-Seq signals of each sample were determined by unpaired t-test.
  • H3K27Ac CHIP-Seq signals of the CDK19KD-H3K27AcUP or CDK19KD-H3K27AcDOWN regions are normalized to 1-Kb and centered on the middle of those regions. Signals of the flanking 2-Kb regions are also shown. To compare relative signal changes, the total signal of each biological replicate was determined by summing the signals of each 50-base window 1-Kb around the center of each region.
  • CDK19KD-H3K27UP and CDK19KD-H3K27DOWN define peak regions where the H3K27Ac signal is more specific for, and most sensitive to, knockdown of CDK19 compared to knockdown of CDK8.
  • the aforementioned GSEA also enabled us to identify the leading edge ‘core’ genes that contribute the most to each enrichment ( FIGS. 12 A and 12 B ).
  • FIGS. 13 A- 13 D differences in H3K27Ac enhancer signals due to CDK19 knockdown ( FIGS. 13 A- 13 D ) result in large corresponding changes in gene expression ( FIG. 13 E ).
  • the gene tracks at the ELF3 ( FIG. 13 A ) and ETV7 ( FIG. 13 B ) loci show enrichment of H3K27Ac signals in the CDK19 knockdown samples, whereas the gene tracks at the CHI3L2 ( FIG. 13 C ) and CRTAM ( FIG. 13 D ) loci show enrichment of H3K27Ac signals in the Control samples.
  • FIGS. 4 A and 4 B show the effect of CDK19 knockdown on the growth of pre-established organoids in vitro and in pre-established PDX tumors in vivo. This aimed to model the treatment of patients’ pre-existing tumors.
  • adding doxycycline to the treatment group significantly reduced the number of pre-established organoids compared to the control (no doxycycline) ( FIGS. 4 A and 4 B ).
  • FIGS. 4 A and 4 B number of organoid colonies at Day 0 ( FIG. 4 A ) and Day 16 ( FIG.
  • CDK19 shRNA induced tumors were ultimately 82% smaller in inducCDK19KD-PDX-T1 tumors and 38% smaller in inducCDK19KD-PDX-T3 tumors when compared to control tumors ( FIGS. 4 C and 4 D ).
  • mouse total body weights were not significantly different between the treatment and control groups ( FIGS. 14 A and 14 B ).
  • survival studies showed that overall survival was significantly longer in mice whose PDX-T1 tumors were transduced with CDK19 shRNA compared to mice transduced with control shRNA ( FIG. 4 E ). Shown in FIG.
  • mice with pre-established PDX tumors with CCT251921 ( FIG. 4 F ), an orally bioavailable inhibitor of both CDK19 and the closely related paralog, CDK8.
  • PDX-T1 tumors were pre-established in mice before starting daily oral administration (30 mg/kg) of CCT251921 or vehicle.
  • Treatment with CCT251921 resulted in a significant reduction in tumor growth by day 14 ( FIG. 4 G ).
  • CDK19 regulates multiple cancer relevant pathways and that it is a potential therapeutic target in TICs.
  • CDK19 inhibition is useful both to therapeutic strategies targeting transcriptional co-factors such as CDK8, CDK9, and BRD4, and to those targeting TICs and their self-renewal pathways such as Hedgehog, Wnt/ ⁇ -catenin, and Notch.
  • transcriptional co-factors such as CDK8, CDK9, and BRD4
  • BRD4 inhibition for example, resulted in a disruption of tissue homeostasis in multiple organs in mice.
  • CDK19 has more limited tissue distribution (see, e.g., Tsutsui et al., Genes to cells : devoted to molecular & cellular mechanisms 16:1208-1218, 2011), potentially limiting the toxicity from CDK19 inhibition, while CDK8, CDK9, and BRD4 knockouts are lethal (see, e.g., Brown et al., Mamm Genome 23:632-640, 2012; Westerling, Molecular and Cellular Biology 27:6177-6182, 2007; and Houzelstein et al., Molecular and Cellular Biology 22, 3794-3802, 2002).
  • CDK19 in tissues could broaden the therapeutic window to enable the otherwise toxic inhibition of stem cell pathways such as NOTCH, or critical processes, such as G2/M checkpoint.
  • stem cell pathways such as NOTCH
  • G2/M checkpoint critical processes, such as G2/M checkpoint.
  • CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity. Nature 455, 547-551, doi:10.1038/nature07179 (2008).
  • HIF1A employs CDK8-mediator to stimulate RNAPII elongation in response to hypoxia.
  • MSigDB Molecular Signatures Database
  • N-myc downstream regulated gene (NDRG) family diverse functions, multiple applications. FASEB J 24, 4153-4166, doi:10.1096/fj.09-151464 (2010).
  • CDK19 Transcript Variant 1 (NM_015076.4) (SEQ ID NO: 12)
  • CDK19 Transcript Variant 2 (NM_001300960.1) (SEQ ID NO: 13)
  • CDK19 Transcript Variant 3 (NM_001300963.1) (SEQ ID NO: 14)
  • CDK19 Transcript Variant 4 (NM_001300964.1) (SEQ ID NO: 15)
  • CDK8 Cyclin dependent kinase 8
  • NM_001260.2 transcript variant 1
  • CDK8 Cyclin dependent kinase 8
  • NM_001318368.1 transcript variant 2
  • CDK8 Cyclin dependent kinase 8
  • NM_001346501.1 transcript variant 3

Abstract

The invention is directed to methods of treating TNBC in a patient by administering to the patient an agent that inhibits the expression or activity of cyclin-dependent kinase 19 (CDK19). In some embodiments, the agent may be a small molecule inhibitor, a polynucleotide (e.g., shRNA. siRNA), or a protein (e.g., an antibody). In some embodiments, the agent does not inhibit the activity or expression of CDK8.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims benefit and is a continuation of Application No. 16/648,088 filed Mar. 17, 2020, which is a national phase application of PCT Application No. PCT/US2018/051489, filed Sep. 18, 2018, which claims benefit of U.S. Provisional Pat. Application No.: 62/560,140, filed Sep. 18, 2017, which applications are incorporated by reference in their entirety for all purposes.
  • FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
  • This invention was made with Government support under contract W81XWH-11-1-0287 awarded by the Department of Defense; under contract W81XWH-13-1-0281 awarded by the Department of Defense; and under contract CA100225 awarded by the National Institutes of Health. The Government has certain rights in the invention.
  • REFERENCE TO A SEQUENCE LISTING
  • [0002A] The Sequence Listing written in file 103182-1342710-000120US_Seq_Listing.xml created on Aug. 18, 2022, 81 KB, is hereby incorporated by reference in its entirety for all purposes.
  • FIELD OF THE INVENTION
  • The invention relates to the field of biomedicine, e.g., oncology.
  • BACKGROUND
  • Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype disproportionately affecting younger women and associated with poor prognoses. See Bauer et al. “Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry” Cancer 109, 1721-1728, doi:10.1002/cncr.22618 (2007). Despite affecting 20% of all breast cancer patients, there are currently no clinically approved targeted therapies for these patients. There exists a need in the art for effective methods of treating TNBC.
  • SUMMARY
  • The invention is directed to methods of treating TNBC in a patient by administering to the patient an agent that inhibits the expression or activity of cyclin-dependent kinase 19 (CDK19).
  • In one aspect, the invention features a method of treating a patient diagnosed with triple-negative breast cancer (TNBC) by administering a therapeutically effective dose of an agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19) and achieves at least one of a reduction in cachexia, increase in survival time, elongation in time to tumor progression, reduction in tumor mass, reduction in tumor burden and/or a prolongation in time to tumor metastasis, time to tumor recurrence, tumor response, complete response, partial response, stable disease, progressive disease, or progression free survival.
  • In another aspect, the invention features a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), wherein the cancer is characterized by a tumor comprising EpCAMmed/high/CD10-/low epithelial cells. The method includes administering a therapeutically effective dose of an agent that inhibits cyclin-dependent kinase 19 (CDK19) expression or activity, wherein the treatment reduces the number of EPCAMmed/high/ CD10-/low cells in the tumor, reduces to number of EPCAM med/high /CD10-/low cells per unit volume of the tumor, or results in a reduction of the ratio of EpCAMmed/high/CD10 /low epithelial cells to normal (EpCamHi/CD10-) epithelial cells in the tumor.
  • In yet another aspect, the invention features a method of reducing metastasis of TNBC in a patient by administering a therapeutically effective dose of an agent that inhibits expression or activity of CDK19.
  • In some embodiments of all aspects of the invention described herein, the patient is treated with a combination therapy comprising (a) an agent that inhibits expression or activity of CDK19 and (b) radiation therapy and/or chemotherapy.
  • In some embodiments, the method comprises detecting EpCAMmed/hlgh/CD10-/low cells in a tissue sample from the patient prior to or after initiating therapy.
  • In some embodiments, the agent administered to the patient in the methods described herein does not significantly inhibit expression or activity of CDK8. In some embodiments, the agent inhibits expression or activity of CDK19 to a greater extent than it inhibits expression or activity of CDK8.
  • In some embodiments of the methods describe herein, the agent is a nucleic acid. In some embodiments, the agent is a protein. In some embodiments, the agent is a CRISPR/Cas9 system.
  • In some embodiments of the methods describe herein, the agent is a CDK19 targeting shRNA.
  • In some embodiments of the methods describe herein, the agent is a CDK19 targeting siRNA.
  • In some embodiments of the methods describe herein, the agent is a CDK19 targeting shRNA or siRNA complementary or substantially complementary to the 3′ UTR of CDK19, but not to the 3′UTR CDK8.
  • In some embodiments of the methods describe herein, the agent is a CDK19 targeting shRNA or siRNA complementary or substantially complementary to the coding region of CDK19, but not to the coding region of CDK8.
  • In some embodiments of the methods describe herein, the agent is a CDK19 targeting shRNA or siRNA selected from: SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, or SEQ ID NO: 11.
  • In some embodiments, the agent binds CDK19 in the cytoplasm of a breast epithelial cell.
  • In another aspect, the invention also features a method of predicting the likely therapeutic responsiveness of a subject with TNBC to a CDK19 targeting agent. The method includes (a) quantitating EpCAMmed/high/CD10-/low cells in a tumor sample obtained from the subject; (b) comparing the quantity of EpCAMmed/high/CD10-/low cells in (a) to a reference value characteristic of tumors responsive to a CDK19 targeting therapy, and treating the patient with the CDK19 targeting agent if the quantity of EpCAMmed/high/CD10-/low cells is equal to or exceeds the reference value. In some embodiments, the CDK19 targeting agent is an inhibitor of CDK19 expression or activity
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a schematic for RNAi dropout viability screens. Two separate screens were performed in a TNBC PDX (PDX-T1). Cells in one experiment were grown in vitro as organoid colonies and in the other in vivo as PDXs in NSG mice.
  • FIGS. 1B-1D are graphs showing that CDK19 knockdown significantly decreased the viability of TNBC cells (FIG. 1B: MDA-MB231 cells; FIG. 1C: MDA-MB468 cells; and FIG. 1D: HS578T cells) assessed 4 days after transduction with control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2).
  • FIG. 1E is a graph showing that CDK19 knockdown significantly decreased the formation of organoid colonies in PDX-T1.
  • FIG. 1F is a graph showing that CDK19 knockdown does not decrease the viability of non-transformed human mammary epithelial cells (HMEC).
  • FIGS. 1G-1J are graphs showing that CDK19 knockdown significantly inhibits the proliferation of PDX tumors (FIG. 1G: PDX-T1; FIG. 1H: PDX-T2; FIG. 11 : PDX-T3; and FIG. 1J: PDX-T4) grown in NSG mice.
  • FIGS. 1K and 1L are bar graphs showing that CDK19 knockdown prevented transduced (RFP positive) TNBC cells (FIG. 1K: PDX-T1 and FIG. 1L PDX-T4) from metastasizing to the lungs in mice.
  • FIG. 1M shows that in PDX tumors transduced with CDK19 shRNA (images in the second and third rows), very little RFP (images in the last column) is visible. These tumors are composed primarily of un-transduced GFP positive tumor cells (images in the middle column). PDX tumor cells were first labeled with green fluorescent protein (GFP) (middle column) and cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP) (right column).
  • FIG. 1N shows representative images of mouse lungs with PDX-T1 metastases. Lungs from mice with PDXs transduced with control shRNA (top row), shCDK19-1 (middle row) or shCDK19-2 (bottom row) are shown. In PDX-T1, which normally metastasizes to the lung, CDK19 knockdown eliminated the detection of any lung metastases by those cells. Bright field images (left column) show gross lung morphology, FITC images (middle column) identify metastatic tumor cells labeled with GFP, and metastatic tumor cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP) (right column).
  • FIG. 2A shows data from representative flow cytometry analyses of a TNBC (PDX-T1) using EpCAM and CD49f (left) or EpCAM and CD10 (right) as cell surface markers.
  • FIG. 2B is a graph that compares the organoid colony forming capabilities of the EpCAMmed/high/CD10-/low and EPCAMlow/med/CD10low/+ cell sub-populations.
  • FIG. 2C is a table showing the number of tumors formed and the number of injections performed for six groups of PDX tumor cells. Populations and injections where tumors formed are bolded. PDX tumor cells were isolated by flow cytometry based on the expression of EpCAM and CD10 (as in FIG. 2A, right)
  • FIGS. 2D-2G are bar graphs showing that CDK19 expression is higher in the EpCAMmed/high/CD10-/low cells compared to the EPCAMlow/med/CD10low/+ cells in PDX-T1, PDX-T2, and PDX-T8.
  • FIG. 3A includes Venn diagrams showing the number of genes upregulated (upper diagram) and downregulated (lower diagram) by CDK19 knockdown, CDK8 knockdown, or by both CDK19 and CDK8 (overlap region).
  • FIG. 3B is a Venn diagram of Hallmark gene sets enriched across the genes upregulated (upper diagram) or downregulated (lower diagram) by CDK19 knockdown, CDK8 knockdown, or by both CDK19 and CDK8 knockdowns (overlap region) as determined by GSEA.
  • FIGS. 3C and 3D are graphs showing that CHIP-Seq signals across the CDK19KD-H3K27AcUP and CDK19KD-H3K27AcDOWN regions are significantly different in the CDK19 knockdown samples compared to control.
  • FIGS. 3E and 3F are graphs showing a gene set enrichment analysis (GSEA) of CDK19KD-EnhancerUP and CDK19KD-EnhancerDOWN genes using averaged CDK19 knockdown versus control expression data.
  • FIG. 3G is a graph showing the hallmark gene sets identified as enriched in Metascape analysis of the CDK19KD-EnhancerUP ‘core’ genes (top and middle bars) and CDK19KD-EnhancerDOWN ‘core’ (bottom bar) genes. The individual genes contributing to the enrichment of each hallmark gene set are shown to the right of each bar.
  • FIGS. 4A and 4B are graphs showing that in inducCDK19KD-PDX-Tl cells, induction of CDK19 shRNA by addition of doxycycline significantly decreased the number of organoid colonies in the doxycycline treatment group compared to control. Number of organoid colonies at Day 0 (FIG. 4A) and Day 16 (FIG. 4B) after initiating doxycycline treatment is shown.
  • FIGS. 4C and 4D are graphs showing that the induction of CDK19 shRNA in pre-established tumors impaired tumor growth. The growth of pre-established tumors in the doxycycline fed NSG mice and control NSG mice are shown for inducCDK19KD-PDX-Tl (FIG. 4C) and inducCDK19KD-PDX-T3 (FIG. 4D).
  • FIG. 4E is a graph showing that CDK19 knockdown extends the survival of NSG mice with PDX-T1 tumors.
  • FIG. 4F shows the chemical structure of CCT251921, an orally bioavailable selective inhibitor of CDK19 and CDK8.
  • FIG. 4G is a graph showing that the treatment of mice with CCT251921 by daily oral gavage significantly impaired the growth of pre-established PDX-T1 xenograft tumors.
  • FIGS. 5A and 5B are graphs showing the shRNA counts in the in vivo growth experimental sample versus the shRNA counts in the baseline sample (FIG. 5A) and the shRNA counts in the in vitro growth experimental sample versus the shRNA counts in the baseline sample (FIG. 5B).
  • FIG. 5C is a schematic of the criteria used to narrow the initial list of hits from the in vitro and the in vivo screens down to 46 candidate genes.
  • FIG. 5D is a list of 46 candidate genes determined from the in vitro and the in vivo screens after filtering with the criteria shown in FIG. 5C. CDK19 is boxed.
  • FIG. 6A is a bar graph showing that TCGA breast cancer samples from patients with the TNBC subtype are enriched in CDK19 copy number amplifications or CDK19 mRNA upregulation compared to other subtypes.
  • FIG. 6B includes confocal immunofluorescent images of PDX-T1 stained with cytokeratin 8 (CK8) antibodies (first image from the left), CDK19 antibodies (second image), and DAPI (third image). The composite image composed from all three aforementioned images is shown on the far right (images are representative of three independent experiments).
  • FIGS. 7A and 7B are bar graphs showing that CDK19 targeting shRNA effectively silences CDK19 in TNBC cells lines. Expression of CDK19 in MDA-MB231 (FIG. 7A) or MDA-MB468 (FIG. 7B) determined by RT-qPCR for cells transduced with control shRNA, shCDK19-1, and shCDK19-2.
  • FIG. 7C is a bar graph showing that CDK19 targeting shRNA effectively silences CDK19 in a TNBC PDX. Expression of CDK19 in PDX-T1 as determined by RT-qPCR for cells transduced with control shRNA, shCDK19-1, and shCDK19-2.
  • FIG. 7D includes images of tissue samples and representative images of mouse lungs bearing PDX-T4 metastases. Lungs from mice with PDXs transduced with control shRNA (top row), shCDK19-1 (middle row), or shCDK19-2 (bottom row) are shown. Bright field images (left column) show gross lung morphology, FITC images (middle column) identify metastatic tumor cells labeled with GFP, and Texas-Red images (right column) identify shRNA-transduced metastatic cells labeled with RFP.
  • FIG. 8A is a graph showing the flow cytometry analyses of TNBC (PDX-T1) using EpCAM and CD49f and the overlap of the EpCAMmed/high/CD10-/low (1), EPCAMlow/med/CD10low/+ (3) and EpCAM-/CD10- ( (2)) sub-populations.
  • FIG. 8B is a bar graph showing that the induction of CDK19 shRNA with doxycycline effectively silences CDK19 in inducCDK19KD-PDX-Tl cells. Expression of CDK19 in control inducCDK19KD-PDX-Tl cells (black bar) and doxycycline treated inducCDK19KD-PDX-Tl cells (gray bar) as determined by RT-qPCR.
  • FIG. 8C shows that CDK19 knockdown effectively prevents the growth of xenograft tumors in a limiting dilution assay.
  • FIG. 8D is a graph showing ELDA (Hu et al., Journal of Immunol. Methods 347:70-78, 2009) analysis of the data from FIG. 8C to determine tumor initiating frequencies in the doxycycline (Group +Dox) and control groups (Group NoDox). P-values as determined by the ELDA software.
  • FIG. 9 shows the amino acid sequence alignment showing 84% sequence homology between CDK19 and CDK8. Amino acid positions are shown above the sequence. Alignment is performed using Clustal W method with MegAlign (DNAStar).
  • FIG. 10 is a table showing hallmark gene sets found enriched by GSEA of the genes upregulated or downregulated by either CDK19 knockdown or CDK8 knockdown.
  • FIG. 11 is a graph showing that genome-wide H3K27Ac CHIP-Seq signals across all identified H3K27Ac peak regions are not significantly different between the CDK19 knockdown, CDK8 knockdown, and control samples. Aggregate plots of normalized H3K27Ac CHIP-Seq signals across all H3K27Ac peak regions in the CDK19 knockdown (1), CDK8 knockdown (2) and control (3) samples (ns is P > 0.05, all samples n = 3, experiments performed three times).
  • FIGS. 12A and 12B show heat map of the expression of CDK19KD-EnhancerUP ‘core’ genes (FIG. 12A) and CDK19KD-EnhancerDOWN ‘core’ genes (FIG. 12B). Normalized expression of each gene in each biological replicate of the CDK19 knockdown and Control samples are shown.
  • FIGS. 13A-13D are graphs showing representative genes where CDK19 knockdown leads to changes in H3K27Ac signals and corresponding changes in gene expression. Representative gene tracks depicting H3K27Ac signals at the loci of select CDK19KD-EnhancerUP ‘core’ (FIGS. 13A and 13B) and CDK19KD-EnhancerDOWN ‘core’ genes (FIGS. 13C and 13D).
  • FIG. 13E is a heat map of the normalized gene expression of ELF3, ETV7, CHI3L2, and CRTAM across each of the three biological replicates in control and CDK19 knockdown samples.
  • FIGS. 14A and 14B are graphs showing that total body weights of mice were not significantly different between the mice fed doxycycline rodent feed (doxycycline group) compared to the mice fed standard rodent feed (control group) in the inducCDK19KD-PDX-T1 (mean ± s.d., n = 5, experiments performed twice) (FIG. 14A) and inducCDK19KD-PDX-T3 (mean ± s.d., n = 5, experiment performed once) (FIG. 14B) tumor experiments.
  • FIG. 14C is a graph showing that total body weights of mice were not significantly different between the mice receiving oral gavage with CCT251921 compared to Vehicle (mean ± s.d., n = 5, experiment performed once).
  • FIG. 15 is a table showing the pathological features and patient information for the patient derived xenograft tumors used in the experiments.
  • FIGS. 16A-16D show a nucleic acid alignment of the 3′ UTR of CDK8 and CDK19. The underlined and bolded text indicates the overlapping regions.
  • FIG. 17 shows a nucleic acid alignment of the 5′ UTR of CDK8 and CDK19. The underlined and bolded text indicates the overlapping regions.
  • DETAILED DESCRIPTION OF THE INVENTION 1. Introduction - Cdk19 Is Required for Triple-Negative Breast Cancer (TNBC) Growth
  • We have discovered that reducing expression or activity of CDK19 in TNBC cell lines or breast cancer patient derived xenografts in mice inhibits growth and metastases of Triple Negative Breast Cancer (TNBC) tumors. See §4 below (Examples). We have also shown that the biological functions of CDK19 are distinct from those of its paralog, CDK8, and that the CDK19-mediated effect on TNBC tumors is independent of CDK8 activity. These data demonstrate that TNBC can be treated by agents that inhibit CDK19 but do not inhibit CDK8, or agents that preferentially inhibit CDK19 compared to CDK8. The discovery that inhibition of CDK19 is necessary and sufficient for inhibition of TNBC growth and metastases is significant, in part, because of the potential advantages of CDK19 as a therapeutic target. Compared to other ubiquitous transcriptional co-factors, such as CDK8, CDK9, and BRD4, CDK19 has more limited tissue distribution, suggesting reduced toxicity and a broader therapeutic window for CDK19 inhibitors.
  • In addition to demonstrating that CDK19 knockdown had tumor growth inhibitory effects, CDK19 expression was also shown to be enriched in tumor initiating cells, e.g., tumorigenic cells having EpCAMmed/high/CD10-/low expressions, compared to the less tumorigenic cells, e.g., cells having EPCAMlow/med/CD10low/+ expressions (see, e.g., Example 4). Further studies also showed that CDK19 knockdown significantly decreased tumor initiating frequencies (FIG. 8D). This discovery indicates that, compared to other agents, targeting CDK19 will result in a more pronounced and significant effect on highly tumorigenic (e.g., tumor initiating) cells. These discoveries also allow development of theranostic methods for identifying certain TNBC patients likely to respond to CDK19 targeted therapy.
  • 2. Definitions 2.1 Triple-Negative Breast Cancer (TNBC)
  • Triple-negative breast cancer (TNBC) is a breast cancer subtype characterized by lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (Her2). Receptor expression can be measured by immunohistochemical staining or other methods. TNBC is generally a diagnosed by exclusion. Widely used breast cancer therapies that target these receptors are not effective against TNBC, making TNBC treatment particularly challenging.
  • 2.2 Cyclic-Dependent Kinase 19 (CDK19)
  • Cyclic-Dependent Kinase 19 (CDK19) is described in Broude et al., Curr. Cancer Drug Targets 15:739, 2015 and Sato et al., Molecular Cell 14:685-691, 2004. CDK19 belongs to a subset of the CDK family that is reportedly more associated with regulation of RNA polymerase II (RNAPII) transcription (see, e.g., Galbraith et al., Transcription 1: 4-12, 2010) than cell cycle progression. See UniProt entry NP_055891.1; Genbank entries AY028424 & AL603914. The mRNA sequences for CDK19 are also disclosed herein (e.g., SEQ ID NOs:12 -15).
  • 2.3 Cyclic-Dependent Kinase 8 (CDK8)
  • CDK8 is a paralog of CDK19 with 84% amino acid sequence homology to CDK19. See FIG. 9 . CDK8 is described in Broude et al., Curr. Cancer Drug Targets 15:739, 2015 and Sato et al., Molecular Cell 14:685-691, 2004. See UniProt entry CAA59754.1; Genbank entries X85753 & AL590108. The mRNA sequences for CDK8 are also disclosed herein (e.g., SEQ ID NOs:16-18).
  • 2.4 Agent
  • As used here, the term “agent” refers to a biological molecule (e.g., nucleic acids, proteins, peptides, antibodies) or small organic molecule (e.g., having a molecular weight less than 1000, usually less than 500) that can reduce or inhibit the expression or activity of CDK19.
  • 2.5 Inhibitors
  • As used herein, the term “inhibitor” as used in the context of CDK19, refers to a compound, composition or system that reduces the expression or activity of CDK19. An agent may also selectively inhibit CDK19 expression or activity over that of CDK8.
  • 2.6 Knockdown
  • As used herein, the term “knock down” refers to a reduction in the expression level of the CDK19 gene. Knocking down CDK19 gene expression level may be achieved by reducing the amount of mRNA transcript corresponding to the gene, leading to a reduction in the expression level of CDK19 protein. Knocking down CDK19 gene expression level may also be achieved by reducing the amount of CDK19 protein. An knockdown agent is an example of an inhibitor.
  • 2.7 Knockout
  • As used herein, the term “knock out” refers to deleting all or a portion of the CDK19 gene in a cell, in a way that interferes with the function of the CDK19 gene. For example, a knock out can be achieved by altering the CDK19 sequence. Those skilled in the art will readily appreciate how to use various genetic approaches, e.g., CRISPR/Cas systems, to knockout the CDK19 gene or a portion thereof. An knockout agent is an example of an inhibitor.
  • 2.8 Reduction Relative to a Reference Level
  • As used here, the terms “decrease,” “reduced,” “reduction,” and “decreasing” are all used herein to refer to a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 5%, at least about 10%, at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (i.e. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.
  • 2.9 Nucleic Acids
  • As used herein, the terms “polynucleotide,” “nucleic acid,” and “oligonucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides or analogs thereof. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs, or otherwise be modified by art-known methods to render the polynucleotide resistant to nucleases, improve delivery of the polynucleotide to target cells or tissues, improve stability, reduce degradation, improve tissue distribution or to impart other advantageous properties. For example, the DNA or RNA polynucleotide may include one or more modifications on the oligonucleotide backbone (e.g., a phosphorothioate modification), the sugar (e.g., a locked sugar), or the nucleobase. If present, modifications to the nucleotide structure can be imparted before or after assembly of the oligonucleotide. The sequence of nucleotides can be interrupted by non-nucleotide components. An oligonucleotide can be further modified after polymerization, such as by conjugation with a label component, a targeting component, or other component. Polynucleotides may be double-stranded or single-stranded molecules. Furthermore, in order to improve the oligonucleotide delivery, the DNA or RNA oligonucleotide may be packaged into a lipid molecule (e.g., lipid nanoparticles) or be conjugated to a cell-penetrating peptide.
  • 2.10 Treatment
  • As used herein, the terms “treatment,” “treating,” and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. “Treatment,” as used herein, can include treatment resulting in inhibiting the disease, i.e., arresting its development; and relieving the disease, i.e., causing regression of the disease. For example, in the case of cancer, a response to treatment can include a reduction in cachexia, increase in survival time, elongation in time to tumor progression, reduction in tumor mass, reduction in tumor burden and/or a prolongation in time to tumor metastasis, time to tumor recurrence, tumor response, complete response, partial response, stable disease, progressive disease, progression free survival, overall survival, each as measured by standards set by the National Cancer Institute and the U.S. Food and Drug Administration for the approval of new drugs and/or described in Eisenhauer, EA1, et al. “New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).” European journal of cancer 45.2 (2009): 228-247.
  • 2.11 Administration
  • As used herein, the term “administering” or “administration” includes any route of introducing or delivering an agent that inhibits the expression or activity of CDK19 to the subject diagnosed with TNBC. Administration can be carried out by any route suitable for the delivery of the agent. Thus, delivery routes can include, e.g., intravenous, intramuscular, intraperitoneal, or subcutaneous deliver. In some embodiments, the agent is administered directly to the tumor, e.g., by injection into the tumor.
  • 2.12 Therapeutically Effective Dose
  • As used here, the term “therapeutically effective amount” refers to an amount, e.g., pharmaceutical dose, effective in inducing a desired biological effect in a subject or patient or in treating a patient having TNBC described herein. The term “therapeutically effective amount” refers to an amount of an active agent being administered that will treat to some extent a disease, disorder, or condition, e.g., TNBC, relieve one or more of the symptoms of the disease being treated, and/or that amount that will prevent, to some extent, one or more of the symptoms of the disease that the subject being treated has or is at risk of developing. For example, for a given parameter (e.g., tumor volume, tumor diameter, metastases, etc.), a therapeutically effective amount will show an increase or decrease of therapeutic effect of at least 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%, or at least 1-fold, 2-fold, or 3-fold. A therapeutically effective dose is usually delivered over a course of therapy that may extend for a period of days, weeks, or months. A therapeutically effective dose of an agent may be taken alone or in combination with other therapeutic agents. In some cases, a therapeutically effective amount of a CDK19 inhibitor is am amount sufficient to effect a partial response in a patient with TNBC (e.g., a greater than 20% reduction, sometimes a greater than 30% reduction, in the measurable diameter of lesions).
  • 2.13 Patient or Subject
  • A “patient” or “subject,” as used herein, is intended to include either a human or non-human animal, preferably a mammal, e.g., non-human primate. Most preferably, the subject or patient is a human.
  • 2.14 Antisense Strand
  • A “antisense strand” refers to the strand of a double stranded RNAi agent (siRNA or shRNA) which includes a region that is complementary or substantially complementary to a target sequence (e.g., a human CDK8 or CDK19 mRNA including a 5′ UTR, exons of an open reading frame (ORF), or a 3′ UTR). Where the region of “complementarity” or “substantially complementary” need not be fully complementary to the target sequence and may have sequence % identity or % similarity of least 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
  • 2.15 Sense Strand
  • A “sense strand,” as used herein, refers to the strand of a RNAi agent (siRNA or shRNA) that includes a region that is complementary or substantially complementary to a region of the antisense strand.
  • 3. Methods of Treatment
  • In one approach the invention provides a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19). In some embodiments, the treatment results in an at least 10% reduction in tumor volume within 6 month of initiating therapy.
  • In one approach the invention provides a method of treating a patient diagnosed with triple-negative breast cancer (TNBC), wherein the cancer is characterized by a tumor comprising EpCAMmed/high/CD10-/low epithelial cells, the method comprising administering a therapeutically effective dose of an agent that inhibits cyclin-dependent kinase 19 (CDK19) expression or activity, wherein the treatment results in a reduction of the ratio of cells having a medium to high expression level of EpCAM and a low expression level of CD10 to normal cells in the tumor. In some embodiments, the method includes the step of detecting EpCAMmed/high/CD10-/low epithelial cells in a tissue sample from the patient prior to or after initiating therapy.
  • To determine the phenotype of a tumor or to assess treatment prognosis, a biopsy may be obtained from the patient diagnosed with TNBC. A biopsy may be a needle biopsy, or may be a liquid biopsy be obtained from blood vessels and/or lymph nodes that supply the breast, e.g., internal mammary arteries, lateral thoracic arteries, thoracoacromial arteries, axillary lymph nodes.
  • As described in §4, below, CD10 and EpCAM biomarkers identify three distinct sub-populations of Tumor Initiating Cells (TICs) in TNBC. EpCAMmed/high/CD10-/low, EPCAMlow/med/CD10low/+, and EpCAM-/CD10. The phenotype of cancer cells in a TNBC patient can be determined using art-known methods. In one approach a tissue is obtained from the patient and the cell phenotype determined using immunohistochemistry, mass spectrometry analysis, fluorescence activated cell sorting (FACS) or other methods. The cell phenotype can be assigned relative to standard values characteristic of health or cancerous tissue. In one approach the ratio of EpCAMmed/high/CD10-/low cells to normal breast epithelial cells is determined prior to initiation of treatment to assess the likely response of the patient to CDK19 targeted therapy. In one approach a change in the ratio of EpCAMmed/high /CD10-/low cells to normal cells, or a change in the quantity of EpCAMmed/high/CD10-/low cells per volume tissue is detected after initiation of treatment.
  • In one approach the invention provides a method for reducing metastasis of TNBC in a patient, the method comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of CDK19
  • In some embodiments, methods of the invention may be used to treat inflammatory TNBCs or TNBCs that are chemo-resistant. In other embodiments, the methods of the invention may be used to slow down or prevent the metastasis of TNBCs. In further embodiments, the methods described herein that target the CDK19 gene or its corresponding protein may further modulate clinically relevant TNBC pathways regulated by CDK19, such as P53 signaling, KRAS signaling, androgen response, NOTCH signaling, TGF BETA signaling, and IL6-JAK-STAT3 signaling (FIG. 3B), and make them more therapeutically susceptible to cancer treatments.
  • 3.1 Therapeutic Agents (Inhibitors) 3.1.1. Polynucleotides
  • As demonstrated in the examples, the CDK19 gene is essential for the growth of TNBC. Methods of treating TNBC in a subject as described herein may be accomplished by administering a polynucleotide (e.g., oligonucleotide) to the subject to decrease or inhibit the expression of the CDK19 gene. In some embodiments, the polynucleotide may be, for example, a DNA oligonucleotide or an RNA oligonucleotide. In other embodiments, the oligonucleotide may be used in a CRISPR/Cas system. An oligonucleotide that inhibits or decreases the expression of the CDK19 gene may knock out or knock down the CDK19 gene (e.g., the CDK19 gene in a TNBC cell) in the subject.
  • In some embodiments, the oligonucleotide may be an shRNA or an miRNA. In some embodiments, the oligonucleotide may mediate an RNase H-dependent cleavage of the mRNA transcript of the CDK19 gene. In other embodiments, the oligonucleotide may be used in a CRISPR/Cas system.
  • In some embodiments, the mRNA transcript of the CDK19 gene may be targeted for cleavage and degradation. Different portions of the mRNA transcript may be targeted to decrease or inhibit the expression of the CDK19 gene. In some embodiments, a DNA oligonucleotide may be used to target the mRNA transcript and form a DNA:RNA duplex with the mRNA transcript. The duplex may then be recognized and the mRNA cleaved by specific proteins in the cell. In other embodiments, an RNA oligonucleotide may be used to target the mRNA transcript of the CDK19 gene.
  • 3.1.1.1. shRNA
  • A short hairpin RNA or small hairpin RNA (shRNA) is an artificial RNA molecule with a hairpin turn that can be used to silence target gene expression via the small interfering RNA (siRNA) it produced in cells. See, e.g., Fire et. al., Nature 391:806-811, 1998; Elbashir et. Al., Nature 411:494-498, 2001; Chakraborty et al. Mol Ther Nucleic Acids 8:132-143, 2017;, Bouard et al., Br. J. Pharmacol. 157:153-165, 2009. Expression of shRNA in cells is typically accomplished by delivery of plasmids or through viral or bacterial vectors. Suitable bacterial vectors include but not limited to adeno-associated viruses (AAVs), adenoviruses, and lentiviruses. Once the vector has integrated into the host genome, the shRNA is then transcribed in the nucleus by polymerase II or polymerase III depending on the promoter choice. The resulting pre-shRNA is exported from the nucleus and then processed by Dicer and loaded into the RNA-induced silencing complex (RISC). The sense strand is degraded by RISC and the antisense strand directs RISC to an mRNA that has a complementary sequence. A protein called Ago2 in the RISC then cleaves the mRNA, or in some cases, represses translation of the mRNA, thus, leading to its destruction and an eventual reduction in the protein encoded by the mRNA. Thus, the shRNA leads to targeted gene silencing. shRNA is an advantageous mediator of siRNA in that it has relatively low rate of degradation and tu rnover.
  • In some embodiments, the methods described herein include treating TNBC in a subject using an shRNA. The methods may include administering to the subject a therapeutically effective amount of a vector, wherein the vector includes a polynucleotide encoding an shRNA capable of hybridizing to a portion of an mRNA transcript of the CDK19 gene. In some embodiments, the vector may also include appropriate expression control elements known in the art, including, e.g., promoters (e.g., tissue specific promoters), enhancers, and transcription terminators. Once the vector is delivered to the TNBC cell, the shRNA may be integrated into the cell’s genome and undergo downstream processing by Dicer and RISC (described in detail further herein) to eventually hybridize to the mRNA transcript of the CDK19 gene, leading to mRNA cleavage and degradation. In some embodiments, the shRNA may include a nucleic acid sequence that has at least 85% sequence identity to the sequence of GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1) or the sequence of ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2). In particular embodiments, the shRNA may target the amino acids at the N-terminus of an mRNA transcript of the CDK19 gene. In other embodiments, the shRNA may target the amino acids at an internal region of an mRNA transcript of the CDK19 gene.
  • As demonstrated in the Examples, e.g., FIGS. 1G-1J, both shRNAs (GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1) and ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2)) targeted against the CDK19 gene were able to knockdown the gene, which led to a significant reduction in the percentage of RFP positive cells in tumors from all three TNBC PDXs. Further, CDK19 knockdown also inhibited the growth of an aggressive PDX obtained from the brain metastasis of a patient with a chemotherapy-resistant inflammatory breast cancer (FIG. 1J), which was known to be aggressive, difficult to treat, and associated with extremely poor prognoses. In addition to inhibiting tumor growth, shRNAs also inhibited the lung metastases of these tumors in mice (FIG. 1L).
  • In some embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCGAGAATTGAAGTACCTTAA (SEQ ID NO: 1). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to ACCAGCAAATATCCTAGTAAT (SEQ ID NO: 2). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCTTGTAGAGAGATTGTACTT (SEQ ID NO: 3). In some embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GAGGACTGATAGTTCTTCTTT (SEQ ID NO: 4). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GATATTAGAAAGATGCCAGAA (SEQ ID NO: 5). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCCAACAGTAGCCTCATAAAG (SEQ ID NO: 6). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to CGTTCGTATTTATCTAGTTTC (SEQ ID NO: 7). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCATGACTTGTGGCATATTAT (SEQ ID NO: 8). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GCTTGTAGAGAGATTGCACTT (SEQ ID NO: 9). In other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to AGGACTGATAGCTCTTCTTTA (SEQ ID NO: 10). In yet other embodiments, an shRNA targeted against the CDK19 gene may have at least 85% sequence identity (e.g., 87%, 89%, 91%, 93%, 95%, 97%, or 99% sequence identity) to GTATGGCTGCTGTTTGATTAT (SEQ ID NO: 11). One of skill in the art has the knowledge and capability to design shRNAs that target different portions of the CDK19 gene (e.g., the 5′ UTR region or the 3′ UTR region) to achieve the desired reduction in expression of the gene. For example, available tools for designing shRNAs include, e.g., Project Insilico, Genomics and Bioinformatics Group, LMP, CCR, NIH. In some embodiments, an shRNA may be designed to knockout the CDK19 gene.
  • CDK8 and CDK19 shRNA
  • There are a number of structural elements that can affect shRNA efficacy. For specific RNAi knockdown of a desired target gene an shRNA can be designed in consideration of its multiple structural elements. Generally, an shRNA should be about 80 nucleotides in length and designed (from 5′ to 3′) to comprise of the following structural elements to make the hairpin structure of the shRNA: (1) a sense strand (e.g., upper stem); (2) followed by a hairpin loop; (3) an antisense strand (e.g., lower stem or guide strand) that has perfect or near perfect complementary to the target mRNA and is antisense to the target mRNA; (4-5) two cleavage motifs such as, “U” or “UH” at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand; and (6) arbitrary spacer nucleotides of about two nucleotides in length between the first nucleotide of guide strand “U” motif and the hairpin loop, and between the last nucleotide of the sense strand and the hairpin loop. The sense strand and antisense strand, making up the stem, may be designed to consist of a range from about 19 to 29 nucleotides in length, which will form the stem. The loop structure may be designed to consist of a range about 2 to 15 nucleotides in length, and preferably free of any internal secondary structure. Some examples of sequences that may be used for making the hairpin loop, include but are not limited to, a nine nucleotide loop comprising the sequence (TTCAAGAGA), and a seven nucleotide loop comprising the sequence (TCAAGAG). Other design strategies can be found in the relevant disclosure of Ros XB-D, Gu S. Guidelines for the optimal design of miRNA-based shRNAs. Methods (San Diego, Calif) 2016;103:157-166, which is herein incorporated by reference in its entirety for all purposes. There are also several design programs available such as, The RNAi Consortium software from The Broad Institute, which is made available through Sigma-Aldrich and Thermo-Fisher Scientific.
  • The specificity of the target sequence should also be considered, as many mRNAs can share similar sequences. Care should be taken in selecting target sequence that has low sequence homology to other genes in the genome to allow for gene-specific knockdown. Where a gene has multiple forms, to achieve complete knockdown of gene expression, shRNA should target sequences shared among all isoforms of the target mRNA.
  • An alignment of CDK19 and CDK8 mRNA sequences can identify not identical or low percent identity or similarity nucleotide sequence regions which can be used to design shRNAs that have a preference to target to CDK19 mRNA but not CDK8, see for example the 3′ UTR and 5′ UTR alignments in FIG. 16 and FIG. 17 .
  • In some embodiments, shRNA that targets a CDK19 mRNA transcript, and not of CDK8 mRNA transcript can be designed. In one approach the mRNA sequences for human CDK19 and CDK8 from National Center for Biotechnology Information (NCBI, found at Pubmed.gov) and an alignmenti is performed (e.g., with pairwise alignment program such as, LALIGN). A region of about 19 to 29 contiguous nucleotides (e.g., 19-20, 19-21, 19-22, 19-23, 19-24, 19-25, 19-26, 19-27, 19-28, or 19-29) in length is selected based on low sequence identity (e.g., less than 75%, identity, sometimes less than 70% identity, sometimes less than 60% identity. In some embodiments the 19 to 29 nt region has very low (e.g., less than 40%, less than 30% or less than 20% or sequence identity. The contiguous sequence can be in a protein coding region, the 5′-UTR, the 3′-UTR, or span two regions.
  • In one embodiment, target-specific knockdown of CDK19 can be accomplished by designing an shRNA with a guide strand that is complementary of the 3′ UTR region of CDK19 (SEQ ID NO:42) and has low or no homology to the 3′UTR of CDK8 (SEQ ID NO:44). The guide strand may be 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, or 29 nucleotides in length. Some exemplary sequence regions that may be used to design a CDK19 shRNA, include but are not limited to, CTCCAGCTCCCGTTGGGCCAGGCCAGCCC (SEQ ID NO: 20), AGCCCAGAGCACA GGCTCCAGCAATATGT (SEQ ID NO: 21), CTGCATTGAAAAGAACCAAAAAAATGCAA (SEQ ID NO: 22), ACTATGATGCCATTTCTATCTAAAACTCA (SEQ ID NO: 23), TACACATGGGAG GAAAACCTTATATACTG (SEQ ID NO: 24), AGCATTGTGCAGGACTGATAGCTCTTCTT (SEQ ID NO: 25), TATTGACTTAAAGAAGATTCTTGTGAAGT (SEQ ID NO: 26), TTCCCCTATCTCAGCA CCCCTTCCCTGCA (SEQ ID NO: 27), TGTGTTCCATTGTGACTTCTCTGATAAAG (SEQ ID NO: 28), CGTCTGATCTAATCCCAGCACTTCTGTAA (SEQ ID NO: 29), or CCTTCAGCATTTCTTT GAAGGATTCTATC (SEQ ID NO: 30). One of ordinary skill guided by this disclosure understands that other low homology sequence regions in the ‘3 UTR could also be used. See, for example, FIGS. 16A-D the low homology sequence regions from (1-1186) and (2418-4570). In one embodiment, the shRNA may be designed to be targeted to upstream of CDK19, downstream of CDK19, or in the exons of CDK19. In some cases the expression of the CDK19 shRNA results in knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%. In another embodiment the expression of the CDK19 shRNA can preferentially knockdown CDK19 compared to CDK8.
  • To make shRNAs that preferentially target CDK19 one would identify a unique region of CDK19, a region that does not have significant homology to other CDKs (e.g., CDK8) or other mRNAs in the genome. One would use this sequence to make a guide strand that is antisense to this target and comprises 19 to 29 nucleotides in length. To make the expression cassette one would add an appropriate promoter such as a pol II or pol III promotor at the beginning of the cassette, followed by the complementary sense strand (e.g., complementary to the targeting guide strand), which is them followed by the loop structure of about 2 to 15 nucleotides in length. In addition, the two Ago cleavage motifs, “U” or “UH” should be included at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand along to 1-2 spacer nucleotides at the end of the loop structure. See, for example US Application No. US2008/0293142 and Ros XB-D, Gu S. Guidelines for the optimal design of miRNA-based shRNAs. Methods (San Diego, Calif) 2016;103:157-166, which is herein incorporated by reference in its entirety for all purposes.
  • In another embodiment, target-specific knockdown of CDK8 can be performed by using an shRNA with a guide strand that comprises a complementary to the 5′UTR of CDK8 (SEQ ID NO: 43) and has low or no homology to the 5′ UTR of CDK19 (SEQ ID NO:41). The guide strand may be 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29, nucleotides in length. Some exemplary sequences that may be used to design a CDK8 shRNA include but are not limited to, TGGCCGCCCCGCCGCTCCCGCCGCAGCAG (SEQ ID NO: 31), GAGCAGAACGCGCGGCCGGAGA GAGCGGC (SEQ ID NO: 32), GGAGCCGGCGCCCAGGGAGCCCGCGGGGA (SEQ ID NO: 33), CAAGGGCAGAGACACCGCTCCCCACCCCC (SEQ ID NO: 34),AGCCCTCGTCCCTCGGCTCTCCTTCGCCG (SEQ ID NO: 35), GGGGATCCTCCCCGTTCCTCCACCCCCGG (SEQ ID NO: 36), CCGGCCTCTG CCCCGCCGTCCCCCTGGAT (SEQ ID NO: 37), GTCCCTGGCGCTTTCGCGGGGCCTCCTCC (SEQ ID NO: 38), TGCTCTTGCCGCATCAGTCGGGCTGGTGC (SEQ ID NO: 39), or TGCGGCCGGCGGGCGTAGAGC GGGCGGGT (SEQ ID NO: 40). One of ordinary skill in the art would understand that other low homology sequence regions in the ‘5 UTR could also be used. See, for example, FIG. 17 the low homology sequence regions from (1-33) or (223 -504). In another embodiment the shRNA may be designed to be targeted to upstream of CDK8, downstream of CDK8, or in the exons of CDK8. In some cases, the expression of the CDK8 shRNA can result in a knockdown of CDK8 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%..
  • To make shRNAs that preferentially target CDK8 one would identify a unique region of CDK8, a region that does not have significant homology to other CDKs (e.g., CDK19) or other mRNAs in the genome. One would use this sequence to make a guide strand that is antisense to this target and comprises 19 to 29 nucleotides in length. To make the expression cassette one would add an appropriate promoter such as a pol II or pol III promotor at the beginning of the cassette, followed by the complementary sense strand (e.g., complementary to the targeting guide strand), which is them followed by the loop structure of about 2 to 15 nucleotides in length. In addition, the two Ago cleavage motifs, “U” or “UH” should be included at the first position of the guide strand, and “UUC” or “CUUC” at the tail region of the guide strand along to 1-2 spacer nucleotides at the end of the loop structure. See, for example US Application No. US2008/0293142 and Ros XB-D, Gu S. Guidelines for the optimal design of miRNA-based shRNAs. Methods (San Diego, Calif) 2016;103:157-166, which is herein incorporated by reference in its entirety for all purposes.
  • The specificity or knockdown level of an shRNA or siRNA can be confirmed using real-time PCR analysis for mRNA level or ELISA assay for the protein level. Experimental controls may be run in parallel to assess knockdown. Some examples of experimental controls that may be used, include but are not limited to, a mock-infected or mock-transfected sample, an empty vector, an shRNA encoding a scrambled target or seed region, an shRNA targeting another gene entirely such as, housekeeping genes GAPDH or Actin, or a GFP positive control.
  • To determine if an siRNA or shRNA (e.g., RNAi agent) preferentially targets CDK19 over CDK8 one can transfect or transduce the shRNA or siRNA tagged to marker such as GFP in a cell line or other expression system, select the GFP positive cells (e.g. transformed cells), and determine the level of CDK19 knockdown relative to CDK19 expression in the cell system without transfection or transduction with the RNAi agent. In some embodiments, the expression of RNA is measured. In other embodiments, the expression of the protein is measured. In one example, mRNA may be measured by any PCR-based assay known in the art (e.g., RT-PCR or qRT-PCR or the like). In one example, the protein level may be measured by an immunoassay (e.g., ELISA assay or any antibody-based method known in the art).
  • In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 30% and CDK8 greater than about 70% relative to a system without transfection or transduction. In some other embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression at less than about 50% and CDK8 greater than about 95%. In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 5% and CDK8 greater than about 80%. In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression less than about 1% and CDK8 greater than about 60%. In some embodiments, a targeting CDK19 shRNA or siRNA results in CDK19 expression at less than about 0.5% and CDK8 greater than about 90%. In some embodiments, a targeting CDK19 shRNA results in CDK19 expression at about 0% and CDK8 at about 100% relative to a system without transfection or transduction. In some embodiments, the expression of RNA is measured. In other embodiments, the expression of the protein is measured.
  • CDK8 and CDK19 siRNA
  • The present disclosure also provides siRNA-based therapeutics for inhibiting expression of CDK8 and CDK19 in a patient with triple-negative breast cancer. The double stranded RNAi therapeutic includes a sense strand complementary to an antisense strand. The sense or antisense strands of the siRNA may be about 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. The antisense strand of the siRNA-based therapeutic includes a region complementary to a part of an mRNA encoding CDK8 or CDK19. Additional methods to make therapeutic siRNA can be found in U.S. Pat No. US9399775, which is incorporated by reference in its entirety for all purposes.
  • In some cases, the expression of CDK19 siRNA may result in a knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%. In another embodiment, the expression of CDK19 siRNA may preferentially knockdown CDK19 compared to CDK8. In some cases, the expression of CDK8 siRNA may result in a knockdown of CDK8 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • In a preferred embodiment, CDK19 siRNA may result in a knockdown of CDK19 at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% and CDK8 at least about 10%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, or 30%.
  • shRNA and siRNA Delivery
  • Depending on whether transient or stable expression is desired one can select an appropriate delivery vector. Examples of delivery vectors that may be used with the present disclosure are viral vectors, plasmids, exosomes, liposomes, bacterial vectors, or nanoparticles. The present disclosure also provides for delivery by any means known in the art.
  • For targeted delivery to triple-negative breast cancer cells, one skilled in the art would appreciate that delivery vectors may be genetically modified to target a specific cell type or to tissue type. To make a targeted delivery vector or plasmid one can identify a unique molecule expressed or associated with a triple-negative breast cancer (e.g., receptor, protein, glycoprotein, or combination thereof) and then create a delivery vector or plasmid that harbors or expresses these markers, preferably on the outside of the delivery vector or plasmid (e.g., cytosol facing). In addition, depending on the required therapeutic duration a viral delivery vector can be genetically modified to be continuously replicating, replication-defective, or conditionally replicating as described in, Sliva K, Schnierle BS. Selective gene silencing by viral delivery of short hairpin RNA. Virology Journal. 2010.
  • In one embodiment, the CDK8 or CDK19 shRNA or siRNA can be delivered by an adenovirus vector. Adenoviruses non-enveloped viruses with a nucleocapsid and a linear dsDNA genome. While they are able to replicate in the nucleus of mammalian cells, they do not efficiently integrate into the host’s genome and therefore pose only minimal risks of insertional mutagenesis but are inadequate for long-term therapy.
  • In another embodiment, the CDK8 or CDK19 shRNA or siRNA can be delivered by an adeno-associated viral vector (AAV). AAV is one of the smallest viruses and belongs to the genus Dependovirus. It has a small, single-stranded DNA genome and can accommodate about eight individual shRNA. AAV permits entry retargeting, allowing delivery of the shRNA to specific cell or tissue types. In a further embodiment, the present disclosure provides for a modified AAV that is targeted for delivery to a triple-negative breast cancer cell or tissue type.
  • In another embodiment, the CDK8 or CDK19 shRNA or siRNA can be delivered by a retrovirus vector. A retrovirus is a single-stranded RNA virus that belongs to the family of Retroviridae and replicate through a double-stranded DNA intermediate. They can integrate into a host’s genome thereby allowing long-term expression of a shRNA. The Env protein plays a central role in targeting retrovirus to a target cell. In a further embodiment, the present disclosure provides for a retrovirus vector with a modified env gene or its protein product for delivery to a triple-negative breast cancer cell or tissue type. In a further embodiment, the present disclosure provides for delivery of CDK8 or CDK19 shRNA of siRNA using a retrovirus vector with protease-activated Env proteins.
  • In another embodiment, the CDK8 or CDK19 shRNA or siRNA can be delivered by a lentivirus vector. Lentivirus is a subclass of retrovirus in the genus Lentivirinae which can accommodate large amounts of DNA. For some applications, it may be preferable to use a lentivirus vector engineered to be “self-inactivating” known as “SIN” vectors. In a further embodiment, the present disclosure provides for delivery of a CDK8 or CDK19 shRNA by a lentivirus vector with a modified env gene or its protein product for delivery to a triple-negative breast cancer cell or tissue type.
  • In another embodiment, the shRNA or siRNA can be delivered by a nanoparticle. Examples of nanoparticles that can be use with the present disclosure, include but are not limited to, exosomes, liposomes, organic nanoparticles, or inorganic nanoparticles. Other non-limiting examples of nanoparticles include, but are not limited to, e.g., those provided in Hong, Cheol Am, and Yoon Sung Nam. “Functional Nanostructures for Effective Delivery of Small Interfering RNA Therapeutics.” Theranostics 4.12 (2014): 1211-1232. PMC. Web. 13 Sept. 2018, which is hereby incorporated by reference in its entirety for all purposes. In some embodiments, the delivery of the shRNA or siRNA is mediated by receptor, protein, glycoprotein or combination thereof present or specific to triple-negative breast cancer cells.
  • In some embodiments, the siRNA CDK19 therapeutic is administered in a solution. The siRNA may be administered in an unbuffered solution. In one embodiment, the siRNA is administered in water. In other embodiments, the siRNA is administered with a buffer solution, such as an acetate buffer, a citrate buffer, a prolamine buffer, a carbonate buffer, or a phosphate buffer or any combination thereof. In some embodiments, the buffer solution is phosphate buffered saline.
  • 3.1.1.2. Rnase H-Mediated MRNA Degradation/Antisense
  • RNase H-dependent antisense oligonucleotides (ASOs) are single-stranded, chemically modified oligonucleotides that bind to complementary sequences in target mRNAs and reduce gene expression both by RNase H-mediated cleavage of the target RNA and by inhibition of translation by steric blockade of ribosomes.
  • RNase H is an endonuclease enzyme that catalyzes the cleavage of RNA in an RNA:DNA duplex. The most well studied endogenous function for this enzyme is the removal of Okazaki fragments (small RNAs) used to prime the DNA duplication during cell division. In some embodiments, to target the mRNA transcript of the CDK19 gene for degradation, a nucleic acid (e.g., DNA oligonucleotide) capable of hybridizing to a portion of the mRNA may be administered to the subject. Once inside the cell (e.g., a TNBC cell), the DNA oligonucleotide base pairs with its targeted mRNA transcript. RNase H may bind to the resulting duplex and cleave the mRNA transcript at one or more places. The DNA oligonucleotide may further bind to other mRNA transcripts to target them for RNase H degradation. Thus, the expression of the CDK19 gene may be greatly reduced in a subject with TNBC.
  • The DNA oligonucleotide capable of hybridizing to an mRNA transcript of a CDK19 gene may contain, e.g., between 10 and 30 nucleotides (e.g., 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, or 30 nucleotides). In some embodiments, the DNA oligonucleotide may have 100% complementarity to the portion of the mRNA transcript it binds. In other embodiments, the DNA oligonucleotide may have less than 100% complementarity (e.g., 95%, 90%, 85%, 80%, 75%, or 70% complementarity) to the portion of the mRNA transcript it binds, but can still form a stable RNA:DNA duplex for the RNase H to cleave the mRNA transcript. The DNA oligonucleotide may bind to the 5′ UTR or the 3′ UTR of the mRNA transcript of the CDK19 gene.
  • Further, the DNA oligonucleotide capable of hybridizing to an mRNA transcript of a CDK19 gene may contain modified nucleotides at the 5′ end and the 3′ end. The modified nucleotides at the termini may function to protect the internal portion of the DNA oligonucleotide from nuclease degradation and to increase the binding affinity for the target mRNA transcript. In some embodiments, the modified nucleotides at the termini may include a modified nucleobase (e.g., 5-methylcytosine) and/or a modified sugar (e.g., a locked sugar). In some embodiments, 3-5 nucleotides at each of the 5′ and 3′ ends of the DNA oligonucleotide may be modified.
  • 3.1.1.3. miRNA
  • A microRNA (miRNA) is a small non-coding RNA molecule that functions in RNA silencing and post-transcriptional regulation of gene expression. miRNAs base pair with complementary sequences within the mRNA transcript. As a result, the mRNA transcript may be silenced by one or more of the mechanisms such as cleavage of the mRNA strand, destabilization of the mRNA through shortening of its poly(A) tail, and decrease translation efficiency of the mRNA transcript into proteins by ribosomes. In some embodiments, miRNAs resemble the siRNAs of the shRNA pathway, except that miRNAs derive from regions of RNA transcripts that fold back on themselves to form short hairpins, which are also called pri-miRNA. Once transcribed as pri-miRNA, the hairpins are cleaved out of the primary transcript in the nucleus by an enzyme called Drosha. The hairpins, or pre-miRNA, are then exported from the nucleus into the cytosol. In the cytosol, the loop of the hairpin is cleaved off by an enzyme called Dicer. The resulting product is now a double strand RNA with overhangs at the 3′ end, which is then incorporated into RISC. Once in the RISC, the second strand is discarded and the miRNA that is now in the RISC is a mature miRNA, which binds to mRNAs that have complementary sequences.
  • The difference between miRNAs and siRNAs from the shRNA pathway is that base pairing with miRNAs comes from the 5′ end of the miRNA, which is also referred to as the seed sequence. Since the seed sequence is short, each miRNA may target many more mRNA transcript. In some embodiments, an miRNA targeting the CDK19 gene may be used in methods described herein.
  • 3.1.2. Crispr/Cas System
  • In some embodiments, the knocking out or knocking down of the CDK19 gene is performed using a gene editing system such as the CRISPR/Cas system. See Sanders and Joung, Nature Biotechnol 32:347-355, 2014, Huang et al., J Cell Physiol 10:1-17, 2017 and Mitsunobu et al., Trends Biotechnol 17:30132-30134, 2017. The CRISPR/Cas system includes a Cas protein and at least one or two ribonucleic acids that are capable of directing the Cas protein to and hybridizing to a target motif in the CDK19 sequence. The Cas protein then cleaves the target motif and results in a double-strand break or a single-strand break. Any CRISPR/Cas system that is capable of altering a target polynucleotide sequence in a cell can be used in methods described here. In some embodiments, the CRISPR/Cas system is a CRISPR type I system. In some embodiments, the CRISPR/Cas system is a CRISPR type II system. In some embodiments, the CRISPR/Cas system is a CRISPR type V system.
  • The Cas protein used in the methods described herein can be a naturally occurring Cas protein or a functional derivative thereof. A “functional derivative” includes, but are not limited to, fragments of a native sequence and derivatives of a native sequence polypeptide and its fragments, provided that they have a biological activity in common with the corresponding native sequence polypeptide. A biological activity contemplated herein is the ability of the functional derivative to hydrolyze a DNA substrate (e.g., a CDK19 gene) into fragments. The term “derivative” encompasses both amino acid sequence variants of polypeptide, covalent modifications, and fusions thereof. Suitable derivatives of a Cas protein or a fragment thereof include but are not limited to mutants, fusions, or covalent modifications of Cas protein.
  • In some embodiments, the Cas protein used in methods described herein is Cas9 or a functional derivative thereof. In some embodiments, the Cas9 protein is from Streptococcus pyogenes. Cas9 contains 2 endonuclease domains, including an RuvC-like domain which cleaves target DNA that is noncomplementary to crRNA, and an HNH nuclease domain which cleaves target DNA complementary to crRNA. The double-stranded endonuclease activity of Cas9 also requires that a short conserved sequence (e.g., 2-5 nucleotides), known as a protospacer-associated motif (PAM), follows immediately after the 3′ end of a target motif in the target sequence.
  • In some embodiments, the Cas protein is introduced into TNBC cells in polypeptide form. In certain embodiments, the Cas protein may be conjugated to a cell-penetrating polypeptide. Non-limiting examples of cell-penetrating peptides include, but are not limited to, e.g., those provided in Milletti et al., Drug Discov. Today 17: 850-860, 2012, the relevant disclosure of which is hereby incorporated by reference in its entirety. In other embodiments, a TNBC cell may be genetically engineered to produce the Cas protein.
  • In some embodiments, the target motif in the CDK19 gene, to which the Cas protein is directed by the guide RNAs, may be between 15 and 25 nucleotides in length (e.g., 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length). In some embodiments, the target motif is at least 20 nucleotides in length. In some embodiments, the target motif in the CDK19 gene immediately precedes a short conserved sequence known as a protospacer-associated motif (PAM), recognized by the Cas protein. In some embodiments, the PAM motif is an NGG motif. In some embodiments, the target motif of the CDK19 gene is within the first exon. In some embodiments, the target motifs can be selected to minimize off-target effects of the CRISPR/Cas systems. Those skilled in the art will appreciate that a variety of techniques can be used to select suitable target motifs for minimizing off-target effects (e.g., bioinformatics analyses).
  • The ribonucleic acids that are capable of directing the Cas protein to and hybridizing to a target motif in the CDK19 gene are referred to as single guide RNA (“sgRNA”). The sgRNAs can be selected depending on the particular CRISPR/Cas system employed, and the sequence of the target polynucleotide, as will be appreciated by those skilled in the art. In some embodiments, the one or two ribonucleic acids can also be selected to minimize hybridization with nucleic acid sequences other than the target polynucleotide sequence. In some embodiments, the one or two ribonucleic acids are designed to hybridize to a target motif immediately adjacent to a deoxyribonucleic acid motif recognized by the Cas protein. Guide RNAs can also be designed using available software, for example, CRISPR Design Tool (Massachusetts Institute of Technology). In some embodiments, the one or more sgRNAs can be transfected into TNBC cells, according to methods known in the art.
  • The use of antibodies for therapeutic purposes has been used to treat cancer. Passive immunotherapy involves the use of monoclonal antibodies (mAbs) in cancer treatments (see for example, Devita, Hellman, And Rosenberg’s Cancer: Principles & Practice Of Oncology, Eighth Edition (2008), DeVita, V. et al. Eds., Lippincott Williams & Wilkins, Philadelphia, Pa., pp. 537-547, 2979-2990). These antibodies can have inherent therapeutic biological activity both by direct inhibition of tumor cell growth or survival and by their ability to recruit the natural cell killing activity of the body’s immune system. The antibodies can be administered alone or in conjunction with radiation or chemotherapeutic agents. Trastuzumab, approved for treatment of breast cancer is an example of such a therapeutic. Alternatively, antibodies can be used to make antibody-drug conjugates in which the antibody is linked to a drug and directs that agent to the tumor by specifically binding to the tumor. Ado-Trastuzumab emtansine (T-DM1) is an example of an approved antibody-drug conjugate used for the treatment of breast cancer (see, Deng et al., Curr. Med. Chem., Vol. 24(23), 2505-2527 (2017). Another type of immunotherapy is active immunotherapy, or vaccination, with an antigen present on a specific cancer (e.g., TNBC cells) or a DNA construct that directs the expression of the antigen, which then evokes the immune response in the subject, i.e., to induce the subject to actively produce antibodies against their own cancer.
  • Antibodies have been highly effective in targeting cell surface proteins involved in disease. Though it is generally believed that their large size, complex architecture, and structural reliance on disulfide bonds preclude intracellular application, a number of examples of both in situ-expressed (see, e.g, Miersch and Sidhu, F1000Res doi: 10.12688/f1000research.8915.1, 2016) and exogenously supplied whole antibodies shown to maintain functional intracellular activity exist in the literature (see, e.g., Biocca et al., Expression and targeting of intracellular antibodies in mammalian cells. EMBO J. (1990); 9(1): 101-8 and Steinberger et al., Functional deletion of the CCR5 receptor by intracellular immunization produces cells that are refractory to CCRS-dependent HIV-1 infection and cell fusion. Proc Natl Acad Sci USA. (2000); 97(2): 805-10). Attempts to use smaller, less complex binding proteins such as antigen-binding fragments (Fabs) and single-chain variable fragments (scFvs) for intracellular application have similarly shown success in their ability to bind and modulate cytoplasmic protein function (See for example, Marasco et al., Design, intracellular expression, and activity of a human anti-human immunodeficiency virus type 1 gp120 single-chain antibody. Proc Natl Acad Sci USA. (1993); 90(16): 7889-93).
  • As used herein, the term “antibody” encompasses, but is not limited to, whole immunoglobulin (i.e., an intact antibody) of any class. Native antibodies are usually heterotetrameric glycoproteins, composed of two identical light (L) chains and two identical heavy (H) chains. Typically, each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies between the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (V(H)) followed by a number of constant domains. Each light chain has a variable domain at one end (V(L)) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains. The light chains of antibodies from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (κ) and lambda (A), based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG-1, IgG-2, IgG-3, and IgG-4; IgA-1 and IgA-2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • As used herein, the term “epitope” is meant to include any determinant capable of specific interaction with the provided antibodies. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Identification of the epitope that the antibody recognizes is performed as follows. First, various partial structures of the target molecule that the monoclonal antibody recognizes are prepared. The partial structures are prepared by preparing partial peptides of the molecule. Such peptides are prepared by, for example, known oligopeptide synthesis technique or by incorporating DNA encoding the desired partial polypeptide in a suitable expression plasmid. The expression plasmid is delivered to a suitable host, such as E. coli, to produce the peptides. For example, a series of polypeptides having appropriately reduced lengths, working from the C- or N-terminus of the target molecule, can be prepared by established genetic engineering techniques. By establishing which fragments react with the antibody, the epitope region is identified. The epitope is more closely identified by synthesizing a variety of smaller peptides or mutants of the peptides using established oligopeptide synthesis techniques. The smaller peptides are used, for example, in a competitive inhibition assay to determine whether a specific peptide interferes with binding of the antibody to the target molecule. If so, the peptide is the epitope to which the antibody binds. Commercially available kits, such as the SPOTs Kit (Genosys Biotechnologies, Inc., The Woodlands, TX) and a series of multipin peptide synthesis kits based on the multipin synthesis method (Chiron Corporation, Emeryvile, CA) may be used to obtain a large variety of oligopeptides.
  • The term antibody or fragments thereof can also encompass chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab′)2, Fab′, Fab and the like, including hybrid fragments. Thus, fragments of the antibodies that retain the ability to bind their specific antigens are provided. For example, fragments of antibodies which maintain CDK19 binding activity are included within the meaning of the term antibody or fragment thereof. Such antibodies and fragments can be made by techniques known in the art and can be screened for specificity and activity according to general methods for producing antibodies and screening antibodies for specificity and activity (See Harlow and Lane. Antibodies, A Laboratory Manual. Cold Spring Harbor Publications, New York (1988)).
  • Also included within the meaning of antibody or fragments thereof are conjugates of antibody fragments and antigen binding proteins (single chain antibodies) as described, for example, in U.S. Pat. No. 4,704,692, the contents of which are hereby incorporated by reference in their entirety.
  • In one embodiment, a therapeutic antibody (or antibody fragment) can be prepared using methods known in the art, having specificity for an antigen present in breast cancer, and in particular TNBC cells, that is absent or present only at low levels in any normal (non-cancerous) tissue. The therapeutic antibody would therefore have biological activity against TNBC cells and be able to recruit the immune system’s response to treat the disease. The therapeutic antibody can be administered as a therapeutic alone or in combination with current treatments (such as chemotherapy, radiation, or platinum-based therapies) or used to prepare immunoconjugates linked to toxic agents, such as drugs.
  • Monoclonal antibodies to CDK19 (e.g., anti-CKD19 antibodies), made by methods known in the art, can be used to identify the presence or absence of cancerous cells in breast tissue, for purposes of diagnosis or treatment. Anti-CKD19 antibodies can also be used to identify the presence or absence of cancerous cells, or the level thereof, which are circulating in the blood after their release from a solid tumor. Such circulating antigen can include an intact CDK19 antigen, or a fragment thereof that retains the ability to be detected according to the methods taught herein. Such detection may be effected for example, by FACS analysis using standard methods commonly used in the art.
  • In some embodiments, methods of targeting CDK19 can include administering to a subject in need thereof, a therapeutically effective amount of an antibody (e.g., an anti-CKD19 antibody) that is immunoreactive to CDK19 for the treatment of breast cancer, in particular treatment of TNBC. In one embodiment, the antibody having immunoreactivity to CDK19 targets intracellular signaling molecules, such as kinases, as opposed to cell surface molecules, whereby the specificity of the antibody is provided by neutralizing epitope(s) present on CDK19 that are not present on CDK8. In another embodiment, the anti-CDK19 antibody can target the Pl3K/mTOR/AKT pathway or ERK5 (see, Ocana and Pandiella, Oncotarget, 8(13), 22218-22234 (2017)). In one embodiment, the anti-CDK19 antibody can target multiple intracellular signaling molecules, for example, the Pl3K/mTOR and JAK/STAT pathway. In yet another embodiment, the anti-CDK19 antibody can comprise an engineered protein that binds to a neutralizing epitope present on CDK19 that is not present on CDK8.
  • In one embodiment, methods of targeting CDK19 can include administering to a subject in need thereof, a therapeutically effective amount of a tumor antigen (TA)-specific monoclonal antibody for the treatment of TNBC. In one embodiment, the TA-specific mAB can be directed to an intracellular antigen associated with TNBC (See for example, Wang et al., Molecular Oncology, Vol. 9(10), (2015) 1982-1993 and Just, FEBS letters, 2:21 (2014), 350-355).
  • In one aspect, provided is a method of treating a subject with breast cancer, the method including the step of administering to the subject a pharmaceutically effective amount of a composition comprising a CDK19 targeting agent. The CDK19 targeting agent may be a CDK19 targeted antibody, a CDK19 targeted peptide, a CDK19 targeted small molecule, a CDK19 targeted RNA molecule, or a combination thereof. In some instances, the CDK19 targeted agent may be conjugated to a therapeutic agent. In some instances, the method further includes administering a second form of cancer therapy (e.g., chemotherapy or radiation therapy) to the subject. In one embodiment, the breast cancer is TNBC. In another aspect, provided is a method of inhibiting expression of the CDK19 gene in a breast cancer cell, the method including the steps of contacting a breast cancer cell expressing the CDK19 gene with a synthetic CDK19 targeted RNA molecule.
  • In another aspect, provided is a method of assessing responsiveness of a subject with cancer to a CDK19 targeted agent including the steps of: (a) measuring in a tumor sample from a subject the amount of CDK19; (b) determining if a subject has a cancer characterized as having a high level of CDK19 expression; and (c) indicating that the subject is more likely to respond to the CDK19 targeted agent if the subject’s cancer is characterized as having a high level of CDK19 expression or that the subject is less likely to respond to the CDK19 targeted agent if the subject’s cancer is characterized as having a low level of CDK19 expression.
  • In one aspect, provided is a method of treating a subject with cancer, the method comprising administering to the patient a pharmaceutically effective amount of a composition comprising a CDK19 targeted agent. The CDK19 targeted agent is an agent that specifically binds to CDK19 protein or to CDK19 mRNA. CDK19 targeted agents include antibodies, or fragments thereof, peptides, small molecules, and polynucleotides (such as RNA molecules) that specifically bind to CDK19 protein or to CDK19 mRNA. The composition may further comprise a pharmaceutically acceptable carrier. In some instances, CDK19 targeted agents that bind to the CDK19 protein may directly inhibit CDK19 activity. In other instances, CDK19 targeted agents that bind to CDK19 mRNA may inhibit CDK19 expression and thereby inhibit CDK19 activity.
  • In one instance, the CDK19 targeted agent may comprise a CDK19 targeted antibody. The CDK19 targeted antibody may be a monoclonal antibody. In some instances, the CDK19 targeted antibody may be a humanized antibody. In another instance, the CDK19 targeted agent may be a CDK19 targeted peptide. In yet another instance, the CDK19 targeted agent may be a CDK19 targeted small molecule. The CDK19 targeted peptides and small molecules may be derived in a variety of manners as discussed further below. In some instances, the peptides are derived from the sequence of a CDK19 targeted antibody.
  • In some instances, treating a subject with the methods described herein inhibits at least one of: formation of a tumor, the proliferation of tumor cells, the growth of tumor cells, or metastasis of tumor cells in the subject. In another embodiment, treating a subject with the methods described herein may result in reduction of tumor size and, in some instances, elimination of one or more tumors in the subject.
  • 3.1.4. Small Molecule Inhibitors
  • In one approach, methods for treating TNBC include targeting the CDK19 protein using a small molecule inhibitor of CDK19 activity. Examples of small molecule inhibitors of CDK19 are described in U.S. Pat. No. 9,321,737, U.S. Pat. Publication No. US 20170071942, Mallinger et al., J. Med. Chem. 59:1078, 2016, and Czodrowski et al., J. Med. Chem. 59:9337, 2016. In some embodiments, the small molecule inhibitors bind to the ATP binding site of CDK19 to inhibit its activity.
  • The small molecule inhibitor of CDK19 may bind to the ATP binding site of CDK19 covalently or non-covalently to inhibit its activity. In other embodiments, the small molecule inhibitor may bind to other parts of CDK19 outside of the ATP binding site. For example, the small molecule inhibitor may form a covalent interaction with an amino acid (e.g., methionine, tyrosine, or serine) outside of the ATP binding site to inhibit CDK19 activity. In addition to occupying the ATP binding to inhibit kinase activity, a small molecule inhibitor may also bind to CDK19 to cause a conformational change in CDK19 that prevents CDK19 from functioning. In some embodiments, the small molecule inhibitor may bind to CDK19 with a higher affinity than to CDK8. As shown in FIG. 9 , the vast majority of amino acid differences between CDK19 and CDK8 are in the C-terminal domain. In some embodiments, without being bound by any theory, a small molecule inhibitor may bind to an amino acid or a portion in the C-terminal domain of CDK19, that is different from the corresponding amino acid or portion of CDK8, to achieve selective inhibition of CDK19 over CDK8.
  • In some embodiments the small molecule inhibitor is other than a compound described in U.S. Pat. No. 9,321,737. In some embodiments the small molecule inhibitor is other than a compound described in U.S. Pat. Publication No. US 20170071942. In some embodiments the small molecule inhibitor is other than a compound described in, Mallinger et al., J. Med. Chem. 59:1078, 2016. In some embodiments the small molecule inhibitor is other than a compound described in Czodrowski et al., J. Med. Chem. 59:9337, 2016. In some embodiments the small molecule inhibitor is other than one or more compounds selected from the group consisting of Cortistatin A, Sorafenib, Linifanib, Ponatinib, Senexin B, CCT251545, and CCT251921
  • 3.1.5. Cdk19 Inhibitors That Do Not Significantly Inhibit Expression or Activity of Cdk8 or Which Inhibits Expression or Activity of Cdk19 to a Greater Extent Than It Inhibits Expression or Activity of CDK8
  • Agents that inhibitors expression or activity of CDK19 but do not inhibit expression or activity of CDK8, or agents that inhibit expression or activity of CDK19 to a greater extent than expression or activity of CDK8 is inhibited can be designed based on differences in sequence and structure of the CDK19 and CDK8 proteins and their corresponding genes and mRNAs. For example, an alignment of CDK19 and CDK8 mRNA sequences can identify non-identical or low identity nucleotide sequences that can be used to design shRNAs or other nucleic acid agents that associate with CDK19 mRNA but not CDK8 sequences. (see, FIGS. 16 and 17 ). Likewise, aligning CDK19 and CDK8 amino acid sequences can identify divergent regions and antibodies or other binding agents can be produced to specifically bind the CDK19 protein. Likewise, small molecule agents can be identified (by rational drug design or screening) that specifically inhibit CDK19 activity or inhibit CDK19 activity to a greater degree that CDK8 activity.
  • The term “an agent that inhibits CDK19 activity but does not significantly inhibit activity of CDK8” as used herein, refers to an agent that is capable of specifically binding and inhibiting the activity of CDK19 such that minimal CDK19 activity is detected in vivo or in vitro; while the agent causes no significant decrease in CDK8 activity under the same conditions. For example, an agent that inhibits activity of CDK19 can specifically bind to CDK19 and fully or significantly inhibit CDK19 activity in vivo or in vitro. In some cases, a CDK19 inhibitor can be identified by its ability to preferentially bind to the CDK19 gene or a CDK19 gene product, and fully inhibit expression or activity of CDK19. Inhibition of CDK19 occurs when CDK19 activity, when exposed to an agent, is at least about 70% less, for example, at least about 75%, 80%, 90%, or 95% less than CDK19 activity in the presence of a control or in the absence of the agent. No significant decrease in CDK8 activity occurs when CDK8 activity, upon exposure to the agent, is at least about 90%, for example, at least 95%, 96%, 97%, 98%, 99%, or 100%, in comparison to CDK8 activity in the absence of the agent. As set forth herein, the agent can include small molecules (i.e., a molecule having a formula weight of 1000 Daltons or less), such as small molecule chemical inhibitors or large molecules, such as siRNA, shRNA, antisense oligonucleotides, or proteins.
  • Determining the effect of the agent on CDK19 and/or CDK8 activity can be measured using one or more methods known in the art, including but not limited to, half maximal inhibitory concentration (IC50), dissociation constant (KD), and inhibitor constant (Kl). For example, IC50 is a measure of the effectiveness of a substance in inhibiting a specific biological or biochemical function. This value indicates the concentration of the substance needed to inhibit a given biological process (or component of the biological process) by half. The IC50 values are typically expressed as molar concentration. According to the Food and Drug Administration (FDA), IC50 represents the concentration of a drug required for 50% inhibition in vitro. In one embodiment, an agent that inhibits CDK19 activity but does not significantly inhibit activity of CDK8 has an IC50 that is at least about 2-fold, 5-fold, 10- fold, 50-fold, 75-fold, or 100-fold, lower than the concentration of the agent required to effect CDK8 activity under the same conditions. In another embodiment, the IC50 for the agent to inhibit CDK19 activity is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the IC50 for the agent to inhibit the activity of CDK8.
  • In another embodiment, the effect of the agent on CDK19 and CDK8 activity can be determined by calculating the equilibrium dissociation constant (KD) of the agent to each CDK. For example, an agent that inhibits the activity of CDK19 but does not significantly inhibit activity of CDK8 has a KD that is at least about 2-fold, 5-fold, 10- fold, 50-fold, or 100-fold lower than the KD of the agent to CDK8 under the same conditions. In one embodiment, the KD for the agent (to CDK19) is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the Ko for the agent (to CDK8). In a preferred embodiment, the KD is lower for the agent to CDK19 as compared to the KD of the agent to CDK8. Said differently, the equilibrium dissociation constant of the agent (to CDK8) is greater than the equilibrium dissociation constant of the agent (to CDK19). In one embodiment, the agent can include an antibody having a KD value in the micromolar (10-6) to nanomolar (10-7 to 10-9) range. In another embodiment, the agent can include an antibody having a KD in the nanomolar range (10-9) to the picomolar (10-12) range. In yet another embodiment, the agent can have a nanomolar (nM) equilibrium dissociation constant to CDK19 and a micromolar (µM) equilibrium dissociation constant to CDK8. U.S. Pat. Publication No. US20120071477 describes kinase inhibition assays in which a compound at a single concentration (2,000 nM) to inhibit ATP pocket binding.
  • In another embodiment, the effect of the agent on CDK19 and CDK8 activity can be determined by calculating the inhibitor constant (Kl) of the agent to each CDK. The Kl is an indication of how potent an inhibitor is; it is the concentration required to produce half maximum inhibition. The lower the Ki, the greater the binding affinity between the agent and the CDK gene. For example, an agent that inhibits the activity of CDK19 but does not significantly inhibit activity of CDK8 has a Kl that is at least about 2-fold, 5-fold, 10- fold, 50-fold, 75-fold, or 100-fold lower than the Kl of the agent (to CDK8) under the same conditions. In one embodiment, the Kl for the agent to CDK19 is at least about 25%, 50%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%, lower than the Kl for the agent to CDK8. In a preferred embodiment, the Kl is lower for the agent to CDK19 as compared to the Kl of the agent to CDK8. Said differently, the inhibitor constant of the agent to CDK8 is greater than the inhibitor constant of the agent to CDK19. For example, an agent that inhibits activity of CDK19 can bind to CDK19 and significantly inhibit CDK19 activity in vivo or in vitro. In some cases, a CDK19 inhibitor can be identified by its ability to preferentially bind to CDK19 and fully inhibit activity of CDK19. Inhibition of CDK19 occurs when CDK19 activity, when exposed to an agent of the invention, is at least about 70% less, for example, at least about 75%, 80%, 90%, 95%, 96%, 97%, 98%, 99% less, or totally inhibited, in comparison to CDK19 activity in the presence of a control or in the absence of the agent. No significant decrease in CDK8 activity occurs when, CDK8 activity upon exposure to the agent, is at least about 90%, for example, at least 95%, 96%, 97%, 98%, 99%, or 100%, in comparison to CDK8 activity in the absence of the agent.
  • The term “an agent that inhibits activity of CDK19 to a greater extent than it inhibits activity of CDK8” as used herein, refers to an agent that is capable of binding and inhibiting the activity of CDK19 significantly more than the agent’s effect on inhibiting the activity of CDK8 under the same conditions. For example, an agent that inhibits activity of CDK19 to a greater extent than inhibiting the activity of CDK8, occurs when CDK19 activity, when exposed to an agent of the invention, is at least about 10% less, for example, at least about 15%, 20%, 30%, 40%, or 50% less, than the activity of CDK8 under the same conditions in vitro or in vivo. In a preferred embodiment, an agent inhibits the activity of CDK19 to a greater extent than the activity of CDK8, when the activity of CDK19 observed is at least 10% less than the activity of CDK8 under the same conditions. In another embodiment, an agent inhibits the activity of CDK19 to a greater extent than CDK8 activity, when at least 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold less CDK19 activity is observed as compared to CDK8 activity under the same conditions. The extent of inhibition (i.e., comparing CDK19 activity to CDK8 activity) can be determined using one or more methods known in the art, including but not limited to assays described herein in the Examples section of the specification and for example, “Percent Of Control (POC)” or “Normalized Percent Inhibition (NPI)”. POC and NPI are methods that normalize data and are often used when comparing multiple agents (e.g., various antibodies or small molecules) against multiple targets (e.g., CDK19 and CDK8). For example, POC is a method that corrects for plate-to-plate variability (for example in high-throughput drug screening) by normalizing an agent’s measurement relative to one or more controls present in the plate. Raw measurements for each agent can be divided by the “average” of within-plate controls. NPI is a control-based method in which the difference between the agent measurement and the mean of the positive controls is divided by the difference between the means of the measurements on the positive and the negative controls (Malo et al., Nature Biotechnology, Vol. 24, 167-175 (2006)). By normalizing the extent of inhibition observed, accurate conclusions can be made regarding which agent(s) are effective at inhibiting the activity of each target under investigation.
  • 3.1.6. Combination Therapy
  • In one approach the patient is treated with a combination therapy comprising an agent that inhibits expression or activity of CDK19 and (a) radiation therapy and/or chemotherapy. In one approach radiation or chemotherapy eliminates the bulk of the tumor mass and the CDK19 inhibitor reduces the number of viable cancer stem cells (e.g., EpCAMmed/high/CD10-/low) cells. In one approach the chemotherapy comprises administration of an anthracycline (e.g., Doxorubicin or Epirubicin), a taxane (e.g., Paclitaxel or Docetaxel), an anti-metabolite (e.g., Capecitabine or Gemcitabine), a platinum agent (e.g., Carboplatin or Cisplatin), Vinorelbine, or Eribulin.
  • 3.2 Methods of Assessing or Predicting Therapeutic Effect
  • A course of therapy with the CDK19 inhibitor will have a beneficial outcome for the patient, including, for example, a reduction in tumor volume, a reduction in metastases, and a reduction in tumor cells having the phenotype EpCAMmed/high and CD10-/low .
  • Tumor volume may be measured using art-known methods. See, e.g., Wapnir et al., Breast Cancer Res Treat 41:15-19, 1996; Sapi et al., PLoS One 10:e0142190, 2015. Tumor volume may be reduced by at least 10%, optionally at least 20% and sometimes by at least 50% after a course of treatment with a CDK19 inhibiting agent as monotherapy or in combination with other agent(s) or treatments. In some embodiments, the reduction in tumor volume (e.g., at least 10%, 20%, or 30% reduction in tumor volume) may be observed as soon as within 1 month of initiating therapy. In other embodiments, the reduction in tumor volume (e.g., at least 10%, 20%, 30%, 40%, 50%, or 60% reduction in tumor volume) may be observed within 2, 3, 4, 5, or 6 months of initiating therapy. In other embodiments, the methods described herein to treat TNBC may also slow down or inhibit the further growth of a tumor. In some embodiments a patient receives combination therapy and a therapeutic benefit is observed that exceeds that of monotherapy with the second agent.
  • A reduction in metastases in an individual may be determined as described in Makela et al., Sci Rep. 7:42109, 2017 and may be observed in a population according to standard methodology.
  • In some embodiments, the presence or amount of cancer cells having the expression profile EpCAMmed/high and CD10-/low in a TNBC tumor tissue obtained from a subject may be used to predict or assess the therapeutic responsiveness of the subject to treatments that target the CDK19 gene or its corresponding protein. As described and demonstrated herein, cells having the expression profile EpCAMmed/high/CD10-/low have a high tumor initiating capacity and are also enriched in CDK19. In some embodiments, subjects having a high percentage of EpCAMmed/high and CD10-/low TNBC cells may be especially responsive.
  • In one approach the likely therapeutic responsiveness of a subject with TNBC to a CDK19 targeting agent is determined by (a) quantitating EpCAM med/high /CD10-/low cells in a tumor sample obtained from the subject; (b) comparing the quantity of EpCAMmed/high/CD10- /low cells in (a) to a reference value characteristic of tumors responsive to a CDK19 targeting therapy, and treating the patient with an inhibitor of CDK19 expression or activity if the quantity of EpCAMmed/high/CD10-/low cells is equal to or exceeds the reference value. The reference value can be determined by quantitating EpCAMmed/high/CD10-/low cells in healthy and TNBC populations and calculating statistically significant ranges characteristic of healthy and tumor tissues. In another approach tumor tissue and healthy tissue from the same subject can be tested, and subjects with elevated EpCAMmed/high/CD10-low cells in tumor relative to healthy tissues can be identified as likely to respond to CDK19 targeted therapy.
  • 3.3 Delivery of Agents
  • The pharmaceutical compositions used in methods described herein may include an active ingredient and one or more pharmaceutically acceptable carriers or excipients, which can be formulated by methods known to those skilled in the art. In some embodiments, a pharmaceutical composition of the present invention includes, in a therapeutically effective amount, a DNA or RNA oligonucleotide that decreases the expression level of the CDK19 gene. In other embodiments, a pharmaceutical composition of the present invention includes, a pharmaceutical composition of the present invention includes a DNA or RNA oligonucleotide in a therapeutically effective amount, a small molecule that inhibits the activity of CDK19. The therapeutically effective amount of the active ingredient in a pharmaceutical composition is sufficient to prevent, alleviate, or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is within the capability of those skilled in the art.
  • In certain embodiments, a pharmaceutical composition of the present invention is formulated as a depot preparation. In general, depot preparations are typically longer acting than non-depot preparations. In some embodiments, such preparations are administered by implantation (for example subcutaneously) or by intramuscular injection. In some embodiments, depot preparations are prepared using suitable polymeric or hydrophobic materials (for example an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • In some embodiments, a pharmaceutical composition may include a delivery system. Examples of delivery systems include, but are not limited to, exosomes, liposomes, and emulsions. In some embodiments, an active ingredient may be loaded or packaged in exosomes that specifically target a cell type, tissue, or organ to be treated. Exosomes are small membrane-bound vesicles of endocytic origin that are released into the extracellular environment following fusion of mutivesicular bodies with the plasma membrane. Exosome production has been described for many immune cells including B cells, T cells, and dendritic cells. Techniques used to load a therapeutic compound into exosomes are known in the art and described in, e.g., U.S. Pat. Publication Nos. US 20130053426 and US 20140348904, and International Patent Publication No. WO 2015002956, which are incorporated herein by reference. In some embodiments, therapeutic compounds may be loaded into exosomes by electroporation or the use of a transfection reagent (i.e., cationic liposomes). In some embodiments, an exosome-producing cell can be engineered to produce the exosome and load it with the therapeutic compound. For example, exosomes may be loaded by transforming or transfecting an exosome-producing host cell with a genetic construct that expresses the active ingredient (i.e., a DNA or RNA oligonucleotide), such that the active ingredient is taken up into the exosomes as the exosomes are produced by the host cell. Various targeting moieties may be introduced into exosomes, so that the exosomes can be targeted to a selected cell type, tissue, or organ. Targeting moieties may bind to cell-surface receptors or other cell-surface proteins or peptides that are specific to the targeted cell type, tissue, or organ. In some embodiments, exosomes have a targeting moiety expressed on their surface. In some embodiments, the targeting moiety expressed on the surface of exosomes is fused to an exosomal transmembrane protein. Techniques of introducing targeting moieties to exosomes are known in the art and described in, e.g., U.S. Pat. Publication Nos. US 20130053426 and US 20140348904, and International Patent Publication No. WO 2015002956, which are incorporated herein by reference.
  • 4. Examples 4.1 Example 1- Materials and Experimental Methods Chemical Reagents
  • The following are the chemical names for the compounds used in this study. CCT152921 is4-[(2-Phenylethyl)amino]-6-quinazolinecarbonitrile (NIH NCAT). The compound was re-suspended in vehicle (PBS + 0.5% Methocel (w/v) + 0.25% Tween 20 (v/v)) to a concentration of 3 mg/mL and mice were dosed at 30 mg/kg. CCT251921 or vehicle was administered via daily oral gavage.
  • shRNA Expression Lentiviral Plasmids
  • Pairs of complementary ssDNA oligonucleotides containing the sense target sequence, a 15-mer loop sequence (5′-GTTAATATTCATAGC-3′ SEQ ID NO: 19), and the reverse complement of the sense sequence were synthesized (Elim Biopharmaceuticals). The oligonucleotides were annealed in 50 µM annealing buffer (10 mM Tris-HCl pH 8.0, 50 mM NaCl, 1 mM EDTA). The double-stranded DNA oligo templates were subsequently cloned into the pRSI12-U6-(sh)-HTS4-UbiC-TagRFP-2A-Puro shRNA expression vector (Cellecta) digested with Bbsl for constitutively active shRNA vector constructs and pRSITUR-U6Tet-(sh)-UbiC-TetRep-2A-TagRFP digested with Bbsl for inducible shRNA vector constructs. The sense strands in the shRNA vectors used in this study were: 5′-GCG AGA ATT GAA GTA CCT TAA-3′ (shCDK19-1 (SEQ ID NO: 1)), 5′-ACC AGC AAA TAT CCT AGT AAT-3′ (shCDK19-2 (SEQ ID NO:2)), and 5′-GCA GGG TAATAA CCA CATTAA-3′ (shCDK8-2 (SEQ ID NO: 3)). The unmodified pRSI12-U6-(sh)-HTS4-UbiC-TagRFP-2A-Puro shRNA expression vector above was used as the ‘empty’ control shRNA. The pHIV-ZsGreen expression vector (Addgene) was used to produce GFP positive tumor cells. The DECIPHER 27 K Pooled shRNA lentivirus library - Human Module 1 (Cellecta) used for the RNAi screen contains 27,500 unique shRNA constructs targeting 5,043 human genes (approximately five or six redundant shRNAs per gene) in the same pRSI12 shRNA expression vector.
  • Cell Lines
  • MDA-MB231, MDA-MB468, HS578T, and 293T cells were obtained from ATCC. HMEC cells were obtained from ThermoFisher Scientific. These cells were certified by the vendors to be mycoplasma free. None of the cell lines used are listed in the database of commonly misidentified cell lines maintained by ICLAC. All cell lines used were passaged less than 10 times from when the original cells from the vendors were thawed. All MDA-MB231, MDA-MB468, 293T, and HS578T cells were grown in DMEM (Invitrogen) supplemented with PSA (Life Technologies), 10% FBS (Hyclone), Glutamax (ThermoFisher Scientific), and sodium pyruvate (Life Technologies). HMEC cells were grown in HuMEC Ready Medium (ThermoFisher Scientific).
  • Mice
  • Nod scid gamma (NSG) mice (NOD.Cg-Prkdcscid IL2Rgtm1Wjl/SzJ) were purchased from the Jackson Laboratory. Mice used for PDX experiments were adult female mice between 8 and 10 weeks old. All the mice used in this study were maintained at the Stanford Animal Facility in accordance with a protocol approved by the Stanford University APLAC committee. Mice were maintained in-house under aseptic sterile conditions. Mice were administered autoclaved food and water. For PDX experiments utilizing doxycycline inducible constructs, mice were provided rodent feed containing 625 mg Doxycycline hyclate/kg diet (Envigo) in place of their normal rodent diet.
  • PDX Tumors and Their Pathological and Clinical Characteristics
  • For human samples, informed consent was obtained after the approval of protocols by the Institutional Review Boards of Stanford University and The City of Hope. See FIG. 15 for a full description of all the PDX tumors used in this study.
  • Single Cell Suspension of PDX Tumor Cells
  • Xenografts were mechanically chopped with a razor blade to approximately 1 mm pieces and then incubated at 37°-C for 3 to 4 hours with collagenase and hyaluronidase (Stem Cell Technologies) in Advanced DMEM/F12 (Invitrogen) with 120 µg/mL penicillin, 100 µg/mL streptomycin, 0.25 µg/mL amphotericin-B (PSA) (Life Technologies). Cells were then treated with ACK lysis buffer (Gibco) to lyse red blood cells, followed by 5 mins of treatment with pre-warmed dispase (Stem Cell Technologies) plus DNAsel (Sigma) and filtered through a 40 µm nylon mesh filter. Cells were finally washed with flow cytometry buffer (HBBS, 2% FCS, PSA).
  • Enrichment of PDX Tumor Cells
  • After PDX tumors were dissociated into single cells, the number of live cells were determined with Trypan blue staining and manually counted with a hemocytometer. Cells were resuspended with flow cytometry buffer to a concentration of 106 live cells/mL and incubated 1:50 (v/v) with Biotin anti-human CD326 (EpCAM) antibody (Biolegend) for 20 mins at 4°-C. Cells were washed with flow cytometry buffer and then resuspended to 80 µL and incubated with 20 µL anti-biotin microbeads (Miltenyi Biotec) for 20 mins at 4°-C. Cells were then washed with flow cytometry buffer and resuspended in 500 µL of buffer. Cells were applied to magnetized LS columns (Miltenyi Biotec), washed, and eluted off magnet per manufacturer’s protocol.
  • Lentivirus Production
  • Lentivirus was produced with Packaging Plasmid Mix (Cellecta) and subcloned pRSl12 shRNA expression plasmids using Lipofectamine 2000 (Thermofisher Scientific) in 293T cells per manufacturer’s instructions. Supernatants were collected at 48 h and 72 h, filtered with a 0.45 µm filter and precipitated with Lentivirus Precipitation Solution (Alstem LLC) per manufacturer’s instructions. Virus was resuspended in 1/100 original volume. Viral titers were determined by flow cytometry analyses of 293T cells infected with serial dilutions of concentrated virus.
  • Lentivirus Infection
  • For in vitro cell line experiments, concentrated lentiviral supernatant (to achieve an MOI of 3) was mixed with cells at the time of seeding. Cells were monitored by visualization of RFP under fluorescence microscopy. All flow cytometry analyses were performed after at least 72 hours of infection.
  • For in vivo PDX tumor growth and organoid colony formation experiments, concentrated lentiviral supernatant (to achieve an MOI of 10) was mixed with single cell suspensions of PDX tumor cells in organoid media with 4 µg/mL of Polybrene (Sigma-Aldrich). Organoid media consisted of: Advanced DMEM/F12 (Invitrogen), 10% FBS (Hyclone), 2.5% growth factor-reduced Matrigel (BD), 10 ng/mL mouse EGF (R&D), 100 ng/mL Noggin (R&D), 250 ng/mL RSPO-I (R&D), 1X B27 (Invitrogen), 1X N2 (Invitrogen), and PSA (Life Technologies). Cells were then spinoculated by centrifuging at 15 ºC for 2 hours at 1200xg. Cells were resuspended by pipetting and left overnight in 48-well ultra-low attachment cell culture plates (Corning).
  • For organoid colony formation assays, cells were transferred the next day to matrigel. For in vivo PDX assays, approximately 75% of the cells were injected into NSG mice as described in the PDX tumor engraftment section. The remainder 25% of cells were plated on matrigel and grown in organoid media for 72 hours until the cells became RFP positive. At that point media was removed and exchanged for dispase and incubated for 2-3 h until the matrigel dissolved. Dissociated cells were resuspended in flow cytometry buffer and analyzed by flow cytometry to determine the ‘baseline’ RFP percentage for cells that were injected into the mice.
  • Organoid Colony Formation Assay
  • Irradiated L1-Wnt3a feeder cells (generous gift of Dr. Roel Nusse) were mixed with growth factor reduced matrigel (BD Biosciences) and allowed to solidify at 37 ºC. Single cell suspensions of PDX tumor cells were transferred onto the solidified matrigel/feeder cell mix substrate and grown in organoid media. Cells were grown for approximately 2 weeks in a 37 ºC incubator with 5% CO2. 50% of media was exchanged with fresh media every 3-4 days. Colonies were counted under fluorescence microscopy. Only RFP positive colonies (which represent transduced cells) were counted. For experiments in which we induced expression of CDK19 shRNA, doxycycline hyclate was added to a final concentration of 100 ng/mL into the media.
  • Cell Viability Assay
  • For cell lines treated with chemical or infected with lentivirus, WST-1 Cell Proliferation Reagent (Roche) was added at 1:10 (v/v) final dilution to each well per manufacturer’s instructions. Cells were subsequently incubated at 37 ºC and 5% CO2. Between 1 and 4 hours after addition of reagent, plates were analyzed on a SpectraMax M3 Bioanalyzer (Molecular Devices). Absorbance for each well was measured at 450 nm (signal wavelength) and 650 nm (reference wavelength). Thus, the signal for each experimental sample was Absorbanceexperimental (A450nm-A650nm). To correct for the effect of media, Absorbancebackground (A450nm-A650nm) was obtained by measuring absorbance in a blank well. Thus, the background corrected signal for each sample Acorrected = Absorbanceexperimental -Absorbancebackground. All Acorrected values for the knockdowns were normalized to the Acorrected value for the control sample to obtain a ‘Relative Viability’.
  • Quantitative PCR RNA Expression Analyses
  • Cells were lysed with Trizol (Life Technologies) and RNA was extracted according to the manufacturer’s instruction. RNA was then treated with DNAsel to remove contaminating genomic DNA. RNA was reverse transcribed to cDNA using SuperScript III First Strand Synthesis kit (Life Technologies) according to the manufacturer’s instructions. TaqMan Gene Expression Master Mix (Applied Biosystems) and the following TaqMan Gene Expression Assays (Applied Biosystems) were used following manufacturer’s instructions: ACTB, Hs00357333_g1; CDK19, Hs01039931_m1; CDK8, Hs00993274_m1. Data was collected on a 7900HT Fast Real-Time PCR System (Applied Biosystems) and data analyzed with SDS 2.4 software (Applied Biosystems). Gene expression data in each sample was normalized against the expression of beta-actin.
  • PDX Tumor Cell Engraftment and Limiting Dilution Assays
  • Single cell suspensions of PDX cells were resuspended in 50% (v/v) mixtures of normal matrigel (BD Biosciences) and flow cytometry buffer in a total volume of 50-100 µL. Using an insulin syringe, cells were injected subcutaneously into the nipple of female NSG mice at the fourth abdominal fat pad. For limiting dilution assays, the specific number of cells injected into the mice were determined by flow cytometry and secondarily by manual counting with a hemocytometer.
  • PDX Tumor Growth and Total Body Weights
  • PDX tumors were detected by palpation. Tumor volumes were determined by measuring the length (l) and width (w) and calculating volumes using the ellipsoid formula ⅙ x l x w2 x π. Tumors volumes and mice weights were determined twice per week.
  • Mouse PDX Tumor and Lung Dissection
  • Xenograft tumors and mice lungs were surgically resected after the mice were euthanized. A 3 to 4 mm section is cut from each tumor and saved in ice cold PBS for imaging. The mice lungs and tumors were imaged on a M205FA Fluorescence Stereo Microscope (Leica) and images were captured with a DFC310FX camera (Leica).
  • Flow Cytometry to Determine RFP Percentage
  • Flow cytometry was performed with a 100 µm nozzle on a Flow Cytometry Aria II (BD Biosciences) with Diva software (BD Biosciences). Data analysis was performed using Flowjo software (Flowjo). For all experiments, side scatter and forward scatter profiles (area and width) were used to eliminate debris and cell doublets. Dead cells were eliminated by excluding 4′,6-diamidino-2-phenylindole (DAPI)-positive cells (Molecular Probes). For PDX tumor cells, they were gated for GFP positivity and then for RFP positivity. RFP percentage is the percentage of GFP positive cells that are also RFP positive. For each sample, we obtain the RFP fraction that is: the RFP % in the tumor divided by the baseline RFP % (see ‘Lentivirus infection’ section). RFP fraction for each sample is then normalized to the RFP fraction for the shRNA control sample which is set at 100% to obtain the ‘Normalized % RFP’.
  • Flow Cytometry Using EpCAM, CD10, and CD49f Cell Surface Markers for Analysis and Cell Sorting
  • Flow cytometry for analysis and cell sorting was performed as previously described. Human antibodies used included: EpCAM-Alexa Fluor 488 (clone 9C4, Biolegend); 1 µg mL-1, CD49f-APC (clone GoH3, Biolegend); CD10 PeCy7/Apc-Cy7 (clone H110a, Biolegend); 1 µg mL-1 and H-2Kd biotin/Pacific Blue (clone SF1-1.1, Biolegend); 1 µg mL-1.
  • RNAi Dropout Viability Screen
  • GFP positive PDX-T1 tumors grown in NSG mice were dissected, processed to single cells, and enriched with EpCAM as described previously. Analysis of cells at this point showed that they were approximately 98%-100% GFP positive.
  • For the in vitro RNAi dropout viability screen, 60 million dissociated PDX-T1 cells were transduced with the DECIPHER 27 K Pooled shRNA lentivirus library-Human Module 1 (Cellecta) at an MOI of 1 in the presence of polybrene and then spinoculated for 2 hours as described previously. The next day, half the cells were spun down and frozen as the in vitro baseline reference sample. A small number of cells were plated separately in organoid colony formation conditions to determine lentiviral infection percentage after 72 hours (cells were found to be approximately 80% RFP positive). The remainder of the cells were plated into twelve 150 mm dishes prepared with 12 mL matrigel containing irradiated L1-Wnt3a feeder cells at 250,000 cells/mL of matrigel. The cells were grown for 19 days with an exchange for fresh media every 3-4 days. On the final day, all the media was exchanged with dispase in order to dissolve the matrigel and to recover the cells. The cells from all the plates were pooled, washed, and frozen as the in vitro organoid growth experimental sample.
  • For the in vivo RNAi dropout viability screen, 30 million dissociated PDX-T1 cells were transduced with the DECIPHER 27 K Pooled shRNA lentivirus library-Human Module 1 (Cellecta) at an MOI of 1.25 in the presence of polybrene and then spinoculated for 2 hours as described previously. The next day, half the cells were spun down and frozen as the in vivo baseline reference sample. A small number of cells were plated separately in organoid colony formation conditions to determine lentiviral infection percentage after 72 hours (cells were found to be approximately 70% RFP positive). The remainder of the cells were resuspended in 50% (v/v) mixtures of normal matrigel (BD Biosciences) and flow cytometry buffer in a total volume of 1.8 mL. These cells were injected evenly into the right and left mammary fat pads of seventeen NSG mice. When tumors reached approximately 10 mm in diameter, the mice were euthanized and the tumors dissected as previously described. These tumors were then processed into single cells, pooled, washed, and frozen as the in vivo growth experimental sample.
  • The two pairs of samples, in vitro baseline reference sample and in vitro organoid growth experimental sample and in vivo baseline reference sample and in vivo growth experimental sample, were submitted to Cellecta, Inc. for genomic DNA extraction, bar code amplification, high-throughput sequencing and de-convolution. Twenty million barcode reads were performed for each sample.
  • ‘Hit’ Selection Algorithm From the In Vivo and In Vitro RNAi Dropout Viability Screens
  • Please see the schematic in FIG. 5C for an overview. We applied an algorithm to narrow our hits to a more manageable number for validation. 1) for each individual shRNA we determined a ‘dropout ratio’ that was shRNA barcode counts in the growth experimental sample divided by shRNA barcode counts in the baseline reference sample. In each screen, these were ranked from lowest to highest. 2) We examined the top 5% of the lowest dropout ratios in each experiment and identified genes targeted by ≥ 2 shRNA. 3) We cross-referenced the shRNA gene targets in the in vivo screen (208 genes) with those in the in vitro screen (150 genes) to identify genes that overlapped between the two experiments. These 46 overlapping ‘hit’ genes are shown in FIG. 5A.
  • Immunofluorescence of PDX Tumors
  • Sections of the PDX tumors were fixed in formalin overnight and then transferred to 70% ethanol. Samples were then embedded in paraffin and sectioned for histology. Formalin fixed paraffin embedded sections were de-parafinized in xylene and rehydrated in an ethanol gradient. Antigen retrieval was performed in a Tris-EDTA buffer by heating in a microwave for 20 min. The primary antibodies, polyclonal Rabbit anti-CDK19 (Sigma) and polyclonal chicken anti-CDK8 (Novus Biologicals), were diluted 1:50 and 1:100, respectively, in TBS + 1% BSA before applying to samples overnight. After overnight incubation, the secondary antibodies, Cy3 Donkey anti-Rabbit (Jackson ImmunoResearch) and Alexa 488 Goat anti-Chicken (Life Technologies) were diluted 1:500 in TBS + 1% BSA and incubated with the samples at room temperature. After DAPI staining, sections were mounted with Prolong® Gold antifade (Cell Signaling). A Zeiss LSM710 Confocal microscope (Carl Zeiss) was used to take the immunofluorescence images. Images for publication were processed with Fiji software.
  • Microarray Experiment
  • EpCAM enriched PDX-T1 cells were infected with shCDK19-2, shCDK8-2 or control shRNA and grown in organoid culture conditions for 72 hours. They were subsequently recovered from matrigel with dispase, resuspended in flow cytometry buffer and sorted by flow cytometry to obtain cells that were both GFP and RFP positive. RNA was extracted from these cells by RNeasy plus micro kit (Qiagen) according to manufacturer’s instructions and quantified on an Agilent 2100 Bioanalyzer. 50 ng of total RNA from each sample was used. In vitro transcription, fragmentation, labeling, hybridization to the microarray and scanning was performed by the Stanford Protein and Nucleic acid facility (PAN facility). Samples were hybridized on PrimeView Human Gene Expression Arrays (Affymetrix). Gene Level Differential Expression Analysis was performed with the Transcriptome Analysis Console (Affymetrix). Downregulated genes were defined as those for which log2 (sample/control) < -1.5 and upregulated genes log2 (sample/control) > 1.5.
  • H3K27Ac Chromatin Immunoprecipitations
  • ChIP assays were performed as described in, e.g., Zarnegar et al., Nucleic Acids Research, gkx648, July, 2017. Approximately 250,000 to 500,000 MDA-MB231 cells were used per ChIP. 1 µg of anti-H3K27ac (Active Motif #39133) were used per ChIP.
  • Library Construction
  • ChIP enriched DNA was quantified using a Qubit 3.0 and dsDNA HS assay. Up to 1 ng of DNA was used for library construction using transposition based NEXTERA XT (followed manufacturer’s protocol with ~14 PCR cycles for indexing). Indexed samples were pooled and submitted for sequencing on a NextSeq500 to obtain 75 bp single end reads with read depths of ~60 million reads.
  • Sequence Analysis
  • Raw sequence reads were uploaded to Galaxy (usegalaxy.org) and aligned to the human genome (hg19) using Bowtie2 (-very-fast-local). Only uniquely mapped reads were retained for further analysis. To visualize data, alignment files were used to produce signal tracks with DeepTools (100 bp bins with 200 bp read extensions and RPKM normalization) and BigWig files were loaded into Broad’s Integrated Genome Browser. MACS2 was used to call peaks (-nomodel, p=0.01, -broad, cuttoff 0.1, duplicates = auto, extension 200) for each replicate. A consensus peak list containing only those peaks occurring in all replicates, was generated using Bedtools. We performed differential peak analysis across consensus peaks using DiffBind. The DiffBind output peak list was annotated by fetching the nearest nonoverlapping feature of the human RefSeq table from UCSC. Data for aggregation plots of ChIP signal across various peaks sets were generated using DeepTools′ computeMatrix (scale-regions: 1000; 50 bp bins) and plotProfile. Data was then plotted with GraphPad Prism software.
  • GSEA Analysis
  • Gene set enrichment analysis (GSEA) was performed using the javaGSEA desktop application (GSEA 3.0) with log2 fold change values for CDK19 knockdown versus Control as the ranking metric and Hallmarks, CDK19KD-EnhancerUp and CDK19KD-EnhancerDOWN as the gene sets that were tested for enrichment.
  • Metascape Analysis
  • Metascape custom enrichment analysis of Hallmark gene sets using the CDK19KD-EnhancerUP ‘core’ genes and the CDK19KD-EnhancerDOWN ‘core’ genes (using the following parameters: H. Sapiens as the input species, p-value cutoffs of 0.01 and minimum enrichment 1.5) was performed online (www.metascape.org).
  • Statistical Analysis
  • Results are shown as mean ± s.d. Statistical calculations were performed with GraphPad Prism software (GraphPad Software Inc). Variance was analyzed using the F-test. To determine P-values, t-test was performed on homoscedastic populations, and t-test with Welch correction was applied on samples with different variances. For animal studies, sample size was not predetermined to ensure adequate power to detect a pre-specified effect size, no animals were excluded from analyses, experiments were not randomized and investigators were not blinded to group allocation during experiments.
  • 4.2 Example 2 - Identification of Genes Essential for TNBC Growth
  • To identify genes essential for the growth of TNBC, two pooled RNAi dropout viability screens were performed using a 27,500 shRNA library targeting 5000 genes in PDX-T1, a TNBC PDX (FIG. 15 ). The screens were performed in two different formats, in vitro as organoid cultures and in vivo as PDXs in nod scid gamma (NSG) mice (FIG. 1A). The abundance of individual shRNA in each experimental sample and the baseline reference samples were determined by high throughput sequencing of the shRNA barcodes. The goal was to identify genes whose knockdown by shRNA inhibited the growth of PDX tumor cells across different experimental conditions. Consistent with screens in other tumors, the in vivo screen had a more significant shRNA dropout rate (FIG. 5A) compared to the in vitro screen (FIG. 5B). FIGS. 5A and 5B are graphs showing the shRNA counts in the in vivo growth experimental sample (FIG. 5A) and in the in vitro growth experimental sample (FIG. 5B) versus the shRNA counts in the baseline sample. Control shRNA targeting luciferase (light gray dots) and shRNA targeting CDK19 (dark gray dots) are highlighted. All other shRNA are shown as black dots (each experiment performed once). The final candidate list was restricted to genes with the lowest 5% of shRNA ratios in each screen that were targeted by more than two shRNAs and were also identified both in vitro and in vivo (FIG. 5C). This resulted in the identification of 46 candidate genes (FIG. 5D).
  • CDK19 was chosen because data from the Cancer Genome Atlas (TCGA) showed that CDK19 copy number amplifications and mRNA upregulation were more prevalent in TNBC patient samples (23%) compared to samples from other breast cancer subtypes (see, e.g., Cancer Genome Atlas Research, N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45:1113-1120, 2013; FIG. 6A). Additionally, high CDK19 expression has been reported to correlate with poor relapse free survival in breast cancer patients (see, e.g., Broude et al., Current cancer drug targets 15, 739-749, 2015 and Porter et al., Proc Natl Acad Sci U S A 109: 13799-13804, 2012). CDK19 belongs to a subset of the CDK family that is reportedly more associated with regulation of RNA polymerase II (RNAPII) transcription than cell cycle progression. CDK19 and its paralog, CDK8, can both form the CDK module (CKM) by binding with three other proteins: MED12, MED13, and Cyclin C. The presence and nuclear localization of CDK19 in our PDX cells were confirmed by immunofluorescence (FIG. 6B). In FIGS. 6A and 6B, the percentage shows the percentage of samples with CDK19 copy number amplifications or CDK19 mRNA upregulation in triple-negative, HER2 positive, estrogen receptor positive, and all breast cancers. The fractions show the number of positive samples and total samples in each group. Data obtained from cBioPortal (see, e.g., Gao et al., SciSignal 6, pl1, 2013).
  • 4.3 Example 3 - Growth Inhibitory Effects of CDK19 Knockdown
  • To validate the growth inhibitory effect of CDK19 knockdown, three commonly used TNBC cell lines: MDA-MB231, MDA-MB468, and HS578T were used. Using two different shRNAs (shCDK19-1 (SEQ ID NO: 1) and shCDK19-2 (SEQ ID NO: 2)) that independently target CDK19, the knockdown of CDK19 (FIGS. 7A and 7B) was confirmed. For both FIGS. 7A and 7B, the relative expression of CDK19 in CDK19 knockdown cells is normalized to the mean expression of CDK19 in cells transduced with control shRNA. Gene expression in each condition is normalized to beta-actin as a housekeeping gene (**P < 0.01; ****P < 0.0001, mean ± s.d., (FIGS. 7A and 7B) n = 3 (FIG. 7C) n =2, experiments performed twice). The knockdown of CDK19 also showed that it caused decreased proliferation in all three TNBC cell lines (FIGS. 1B-1D). FIGS. 1B-1D demonstrate that CDK19 knockdown significantly decreased the viability of TNBC cells (viability of MDA-MB231 cells, ****P < 0.0001 (FIG. 1B), MDA-MB468 cells, ***P < 0.001; ****P < 0.0001 (FIG. 1C), or HS578T cells, *P < 0.05; ****P < 0.0001 (FIG. 1D) assessed 4 days after transduction with control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2)). All values in FIGS. 1B-1D were normalized to control shRNA sample (mean ± s.d., n = 3, experiment performed twice, P values determined by unpaired t-test).
  • In the same TNBC PDX used in the initial dropout screen (PDX-T1), CDK19 knockdown (FIG. 7C) also inhibited the formation of organoid colonies (FIG. 1E). In FIG. 1E, colonies were counted 2 weeks after transduction with either control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2), ***P < 0.001 (unpaired t-test) (mean ± s.d., n = 6, experiment performed twice). To determine the effects of CDK19 knockdown in non-transformed mammary cells, human mammary epithelial cells (HMEC) were infected with shRNA targeting CDK19. In HMECs, neither of the two CDK19 knockdowns affected the viability of the cells (FIG. 1F). In FIG. 1F, viability of HMEC cells was assessed 4 days after transduction with control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2). All values are normalized to control shRNA sample, ns is P > 0.05 (mean ± s.d., n = 6, experiment performed twice, P values determined by unpaired t-test). Collectively, the studies show that in vitro, CDK19 knockdown inhibits the proliferation of multiple TNBC cell lines and the formation of PDX organoid colonies but does not adversely affect the growth of non-transformed mammary epithelial cells.
  • We extended our studies to more physiologically relevant in vivo systems by knocking down CDK19 in three different TNBC PDXs grown in NSG mice. These PDXs: PDX-T1, PDX-T2, and PDX-T3 were derived from chemotherapy naive patients (FIG. 15 ). In these studies, all PDX tumor cells were first labeled with green fluorescent protein (GFP) and cells subsequently infected with either CDK19 shRNA or control shRNA were additionally labeled with red fluorescent protein (RFP). Measuring the percentage of GFP-labeled tumor cells that were also RFP positive allowed us to determine the effect the shRNA had on the PDX tumor cells. With each of the two CDK19 shRNAs tested, CDK19 knockdown led to a significant reduction in the percentage of RFP positive cells in tumors from all three TNBC PDXs (FIGS. 1G-1I and FIG. 1M). Tumor growth was monitored and tumors were analyzed when they exceeded 17 mm. The percentage of RFP positive cells in PDX-T1, ***P < 0.001; ****P < 0.0001 (FIG. 1G), PDX-T2, ****P < 0.0001 (FIG. 1H), PDX-T3, **P < 0.01 (FIG. 1I), or PDX-T4, **P < 0.01 (FIG. 1J) were determined by flow cytometry and normalized to the mean RFP percentage of the control shRNA sample that was set to 100%. Each data point represents one mouse. For FIGS. 1H and 1H, mean ± s.d., n = 9, experiment performed three times. For FIGS. 1I and 1J, mean ± s.d., n = 3, experiment performed once. For all, P values determined by unpaired t-test).
  • FIG. 1M shows representative images of PDX-T1 tumors transduced with control shRNA (top row), shCDK19-1 (middle row), or shCDK19-2 (bottom row). Bright field images (left column) show gross tumor morphology, FITC images (middle column) identify tumor cells labeled with GFP and Texas-Red images (right column) identify shRNA-transduced cells labeled with RFP.
  • These results confirmed that CDK19 is critical for tumor growth in vivo. CDK19 knockdown prevented transduced (RFP positive) TNBC cells from metastasizing to the lungs in mice. Percentage of mice with RFP positive lung metastases from mice bearing PDX-T1 (FIG. 1K) or PDX-T4 (FIG. 1L) tumor xenografts are shown. Number of mice with RFP positive lung metastases and total number of mice in each treatment group is shown as a fraction for each condition. PDX tumor cells were transduced with either control shRNA or CDK19 targeting shRNA (shCDK19-1, shCDK19-2) (For FIG. 1K, n = 9, experiment performed three times; For FIG. 1I, n = 3, experiment performed once). Furthermore, in PDX-T1, which normally metastasizes to lung, CDK19 knockdown eliminated the detection of any lung metastases by those cells (FIG. 1K and FIG. 1N). In FIG. 1N, bright field images (left column) show gross lung morphology, FITC images (middle column) identify metastatic tumor cells labeled with GFP, and Texas-Red images (right column) identify shRNA-transduced metastatic cells labeled with RFP. We also tested the effect of CDK19 knockdown on PDX-T4, an aggressive PDX obtained from the brain metastasis of a patient with a chemotherapy-resistant inflammatory breast cancer. Since inflammatory breast cancers are known to be aggressive, difficult to treat, and associated with extremely poor prognoses, it is notable that CDK19 knockdown inhibited both the growth of the PDX (FIG. 1J) and the lung metastases in these mice (FIG. 1L and FIG. 7D). These data show that in vivo, CDK19 knockdown not only affected primary tumor growth, but also inhibited tumor metastasis.
  • 4.4 Example 4 - Identification of Tumor Initiating Cells (TICs) Within the TNBC PDXs
  • Given that CDK19 knockdown inhibited growth in two independent assays commonly used to assess tumorigenicity (PDX growth in vivo and organoid colony formation in vitro) and genes critical for tumor initiation are frequently amplified or overexpressed in a subset of cancers, it is hypothesized that the tumor initiating cells (TICs) might be sensitive to CDK19 inhibition. Thus, we sought to identify the TICs within the TNBC PDXs. Previously, EpCAM and CD49f were utilized to isolate cell sub-populations in normal breast tissue and in breast cancers. However, in many TNBC PDXs, EpCAM and CD49f often cannot clearly separate cells into distinct sub-populations (FIG. 2A, left). Thus, we utilized the basal cell marker, CD10 with EpCAM to FACS-sort breast cancer PDXs. We discovered that CD10 and EpCAM can separate PDX cells into three distinct sub-populations, EpCAMmed/high/CD10-/low, EPCAMlow/med/CD10low/+, and EpCAM-/CD10- (FIG. 2A, right). In FIG. 2A, the large inseparable cell population (left) seen using EpCAM and CD49f, becomes three distinct sub-populations using EpCAM and CD10 (right): EpCAMmed/high/CD10-/low (gate (1)), EPCAMlow/med/CD10low/+ (gate (2)) and EpCAM-/CD10- (gate (3)). The overlap of these three sub-populations using EpCAM and CD49f is also shown (FIG. 8A).
  • To test the tumor initiating capacity of the three EpCAM/CD10 separated sub-populations, we performed organoid colony formation assays in vitro and transplantation limiting dilution assays (LDA) in vivo. In organoid colony forming assays, the EpCAMmed/high/CD10-/low cells formed significantly more organoid colonies than the EpCAMlow/medCD10low/+ cells (FIG. 2B). In FIG. 2B, the EpCAMmed/high/CD10-/low cells formed significantly more organoid colonies than the EPCAMlow/med/CD10low/+ cells, *P < 0.05 (unpaired t-test) (mean ± s.d., n = 3, experiment performed twice). In transplantation assays performed in NSG mice, injection of EpCAMmed/high/CD10-/low cells from all six PDXs consistently formed tumors (FIG. 2C), sometimes with the transplant of as little as 100 cells (PDX-T1 and PDX-T2). In contrast, transplant of EPCAMlow/med/CD10low/+ cells only formed tumors in two PDXs (PDX-T1 and PDX-T2), and only when transplanting high cell numbers (i.e. 2500 cells) (FIG. 2C). Furthermore, no tumors formed from the transplant of EpCAM-/CD10- cells from any PDX. Hence, TIC’s are enriched in the EpCAMmed/high/CD10-/low sub-population of all PDX breast tumors we examined.
  • Having identified these distinct subpopulations, we next investigated whether CDK19 expression was enriched in the more tumorigenic EpCAMmed/high/CD10-/low cells compared to the less tumorigenic EPCAMlow/med/CD10low/+ cells. In three of the four PDXs examined, CDK19 expression was higher in the more tumorigenic EpCAMmed/high/CD10-/low cells compared to the less tumorigenic EPCAMlow/med/CD10low/+ cells (FIGS. 2D-2G). To generate the data in FIGS. 2D-2G, relative expression of CDK19 in the EPCAMlow/med/CD10low/+ and the EpCAMmed/high/CD10-/low cells as determined by RT-qPCR. Gene expression in each condition is normalized to beta-actin as a housekeeping gene. Relative expression of CDK19 is normalized to the mean expression of CDK19 in the EPCAMlow/med/CD10low/+ cells. *P < 0.05 (unpaired t-test) (PDX-T1: mean + s.d., n = 2; PDX-T2: mean + s.d., n = 6 (EpCAMlow/med/CD10low/+) and n = 3 (EpCAMmed/high/CD10-/low); PDX-T3: mean + s.d., n = 6 (EpCAMlow/med/CD10low/+) and n = 3 (EpCAMmed/high/CD10-/low); PDX-T8: mean + s.d., n = 3. All experiments performed at least twice). Thus, while CDK19 was expressed in all the PDX tumors we examined, it was expressed at higher levels in the more tumorigenic EpCAMmed/high/CD10-/low sub-population in three of the four tumors that we investigated.
  • To determine tumor initiating frequencies in the setting of CDK19 knockdown, we performed LDA using PDX-T1 cells transduced with a doxycycline-inducible CDK19 knockdown construct to produce inducCDK19KD-PDX-T1 cells where we can control CDK19 expression (FIG. 8B). In FIG. 8B, the relative expression of CDK19 in doxycycline treated inducCDK19KD-PDX-T1 cells is normalized to the mean expression of CDK19 in control inducCDK19KD-PDX-T1 cells. Gene expression in each condition is normalized to beta-actin as a housekeeping gene (*P < 0.05, mean ± s.d., n =2, experiments performed twice). By comparing the in vivo transplantation of inducCDK19KD-PDX-T1 cells in the presence of doxycycline (+Dox) with inducCDK19KD-PDX-T1 cells without doxycycline (No Dox), we find that CDK19 knockdown eliminates tumor formation in all the cell transplantation conditions examined (FIG. 8C). inducCDK19KD-PDX-T1 cells were injected into the mammary fat pads of NSG mice at 50, 250 and 1250 cells. Mice in the doxycycline group were fed a doxycycline containing rodent feed to induce CDK19 shRNA, while mice in the control group were fed a normal rodent diet. Tumors were detected by palpation of tumors. The number of tumors that formed and the number of injections that were performed are indicated for each population. Populations and injections where tumors formed are bolded (n = 5 per group) in FIG. 8C. Using ELDA, we discovered that the tumor initiating frequencies significantly decreased from 1 in 342 cells (95%Cl: 1 in 828 to 1 in 142) in the control (No Dox) group to 1 in ∞ cells (95%Cl: 1 in ∞ to 1 in 2587) in the CDK19 knockdown (+Dox) group (FIG. 8D). Both the significant decrease in tumor initiating frequency caused by CDK19 knockdown and CDK19′s higher expression in the TIC sub-population suggests that TIC inhibition is likely responsible for the impaired tumor growth observed with CDK19 knockdown.
  • 4.5 Example 5 - Identification of Genes and Pathways Regulated by CDK19
  • There is an 84% amino acid sequence homology between CDK19 and its well described paralog, CDK8 (FIG. 9 ). CDK8 has been shown to play a role in a variety of malignancies including colon cancer, acute myeloid leukemia, and melanoma. Higher expression of CDK8 has been associated with worse prognosis in colon cancer (Firestein et al., Nature 455:547-551, 2008). CDK8 knockout in embryonic stem cells was shown to prevent embryonic development (Porter et al., Proc Natl Acad Sci USA, 109:13799-13804, 2012) due to its essential role in the pluripotent stem cell phenotype. The known cancer-relevant activities of CDK8 may include positive regulation of Wnt/β- catenin pathway, growth factor-induced transcription, and TGFP signaling. Depending on context, CDK8 has also been shown to either negatively or positively regulate transcription. However, recent evidence has suggested that CDK19 may function differently from CDK8. In vitro studies showed that CDK19 and CDK8 participate mutually exclusively of each other in binding to other CKM components, while gene knockdown studies in cell lines of cervical cancer and colon cancer showed that CDK19 and CDK8 regulate different genes. Our goal was to investigate in TNBC whether CDK19 and CDK8 have distinct biological functions by examining global gene expression changes resulting from targeted knockdown of CDK19 or CDK8.
  • To understand whether the molecular targets of CDK19 in TNBC are unique from CDK8, we knocked down each gene in MDA-MB231 and examined the respective gene expression changes relative to control. Overall, CDK19 knockdown affected 3909 genes and CDK8 knockdown affected 4233 genes (FIG. 3A). However, only 12% of upregulated and 5% of downregulated genes in the CDK19 knockdown experiment were also affected by CDK8 knockdown. This suggested that CDK19 and CDK8 largely regulate distinct genes (FIG. 3A).
  • Gene set enrichment analysis (GSEA) of the CDK19 and CDK8 knockdown genes allowed us to identify enriched Hallmark gene sets amongst the most upregulated or downregulated genes (FIG. 3B and FIG. 10 ). In FIG. 10 , the Hallmark gene sets uniquely enriched in the knockdown of CDK19 or CDK8 are shown in black, enriched in both the knockdown of CDK19 and CDK8 are marked by “*” and enriched by genes expressed in opposite directions between the knockdown of CDK19 and CDK8 are marked by “**”. Normalized enrichment scores and FDR q-value are determined by the GSEA software. An FDR cutoff of < 0.25 was used to select significant Hallmarks. These Hallmark gene sets consist of genes that are specifically involved in certain biological states or pathways. Genes associated with known breast cancer-related Hallmarks such as mitosis (E2F targets, G2M Checkpoint, Mitotic Spindle), PI3K-AKT-MTOR signaling, MYC pathways (Myc Targets v1), glycolysis, apoptosis, and oxidative phosphorylation were changed in the same direction by CDK19 and CDK8 knockdowns (FIG. 3B, middle overlap region), demonstrating a co-regulatory relationship between CDK19 and CDK8. Further, genes associated with early estrogen response, epithelial to mesenchymal transition (EMT), cholesterol homeostasis, MYC pathways (Myc Targets v2), interferon alpha response, and fatty acid metabolism changed in the opposite direction in response to knockdown by CDK19 compared to CDK8 (FIG. 3B, boxes), which suggests a counter-regulatory relationship exists between CDK19 and CDK8. Hallmark gene sets enriched by the expression of genes in opposite directions by CDK19 knockdown compared to CDK8 knockdown are boxed. A number of the Hallmark gene sets were only enriched in the genes that uniquely changed due to CDK19 knockdown (FIG. 3B, left region). Hallmarks reflected by these gene sets included P53 signaling, KRAS signaling, androgen response, NOTCH signaling, TGF BETA signaling, and IL6-JAK-STAT3 signaling, which may be potential biological pathways for targeted therapies for TNBC. All of these biological pathways represent active areas of clinical investigation in the evaluation of targeted therapies for TNBC. Consistent with our findings, a number of the pathways found enriched in our CDK19 knockdown experiments, such as cholesterol homeostasis, P53 signaling, mitosis, and NFKB pathways have been shown previously in other cell types to also be regulated by CDK19.
  • In summary, these analyses showed that CDK19 and CDK8 have the potential to co-regulate certain pathways, while counter-regulating others. Furthermore, CDK19, like CDK8, is capable of positively or negatively regulating biological pathways. The multitude of clinically relevant TNBC pathways regulated by CDK19 suggests that targeting CDK19 can provide the opportunity to modulate multiple pathways simultaneously and at the same time, avoid potential toxicity because of the advantageous limited tissue distribution of CDK19. This approach could overcome the resistance to single agent therapy commonly seen in TNBC and also potentially enable the targeting of ‘undruggable’ processes such as those involving P53 or MYC.
  • 4.6 Example 6 - Effects of CDK19 and CDK8 on Epigenetic Modifications
  • Recent studies have highlighted the role of CDK19 and CDK8, as well as other transcriptional CDKs (CDK7, CDK12/CDK13), in regulating the transcription of critical oncogenic genes by acting at large clusters of enhancers (also called ‘super-enhancers’) that are marked by histone 3 lysine 27 acetylation (H3K27Ac). The exact mechanism for this gene regulation is unclear, but is believed to occur in part through interactions of the CKM with Mediator to regulate RNAPII-Mediator interactions and in part by phosphorylating serine residues in the C-terminal domain of RNAPII. Given the propensity of transcriptional CDKs to function at enhancers, we wanted to investigate whether CDK19 and CDK8 can also regulate the epigenetic modifications at enhancer sites as a mechanism to control gene expression. While enhancer modification through other signaling pathways have been identified, this mechanism of gene control has not yet been reported for the CDKs.
  • To explore the role of CDK19 in epigenetic regulation, chromatin immunoprecipitation and sequencing (CHIP-Seq) for the H3K27Ac modification was performed on MDA-MB231 cells under three different conditions: Control (empty vector transduction), CDK19 knockdown, and CDK8 knockdown. Genome-wide analysis of all H3K27Ac modified regions showed that both CDK19 knockdown and CDK8 knockdown had similar global H3K27Ac levels compared to control (FIG. 11 ). In FIG. 11 , H3K27Ac CHIP-Seq signals across all identified H3K27Ac peak regions are normalized to 1-Kb and centered on the middle of those regions. Signals of the flanking 2-Kb regions are also shown. To compare relative signal changes, the total signal of each biological replicate was determined by summing the signals of each 50-base window 1-Kb around the center of each region. P-values between total CHIP-Seq signals of each sample were determined by unpaired t-test. Through comparative analysis of H3K27Ac levels in the CDK19 knockdown compared to the control, we identified 3034 peak regions with increased H3K27Ac signal (All-H3K27UP) and 502 peak regions with decreased H3K27Ac signal (All-H3K27DOWN). By excluding peak regions that were also different in CDK8 knockdown compared to control, we identified 2309 peak regions with increased H3K27Ac signal (CDK19KD-H3K27UP) and 432 regions with decreased H3K27Ac signal (CDK19KD-H3K27DOWN) that were unique to CDK19 knockdown. The specificity of these regions for CDK19 was investigated by comparing the H3K27Ac levels at these regions in CDK19 knockdown, CDK8 knockdown, and control. Compared to control, enrichment of H3K27Ac levels across the CDK19KD-H3K27UP regions (FIG. 3C) and depletion of H3K27Ac levels across the CDK19KD-H3K27DOWN regions (FIG. 3D) were significant only for CDK19 knockdown and not for CDK8 knockdown. In FIGS. 3C and 3D, ***P < 0.001; ns is P > 0.05 (all samples n = 3, experiments performed three times). H3K27Ac CHIP-Seq signals of the CDK19KD-H3K27AcUP or CDK19KD-H3K27AcDOWN regions are normalized to 1-Kb and centered on the middle of those regions. Signals of the flanking 2-Kb regions are also shown. To compare relative signal changes, the total signal of each biological replicate was determined by summing the signals of each 50-base window 1-Kb around the center of each region. P-values between total CHIP-Seq signals of each sample were determined by unpaired t-test. Thus, CDK19KD-H3K27UP and CDK19KD-H3K27DOWN define peak regions where the H3K27Ac signal is more specific for, and most sensitive to, knockdown of CDK19 compared to knockdown of CDK8.
  • We next assessed whether increases or decreases in H3K27Ac levels as a result of CDK19 knockdown corresponded to changes in gene output. For this, the previously defined All-H3K27UP and All-H3K27DOWN peak regions were annotated by proximity to the nearest gene to establish two gene sets: CDK19KD-EnhancerUP (1593 genes) and CDK19KD-EnhancerDOWN (341 genes) for further analysis (Table 1 and Table 2). GSEA of these gene sets with our CDK19 knockdown gene expression data indicated that genes most upregulated by CDK19 knockdown were enriched for the CDK19KD-EnhancerUP genes (NES 1.68, FDR q-value = 0.000) (FIG. 3E), while genes most downregulated by CDK19 knockdown were enriched for the CDK19KD-EnhancerDOWN genes (NES -1.84, FDR q-value = 0.000) (FIG. 3F). Thus, as a result of CDK19 knockdown, perturbations to the H3K27Ac signal at the putative enhancer elements of genes correlated well and in the expected direction with changes in gene expression.
  • TABLE 1
    CHIPSEQ_CDK19-KD ENHANCERDOWN
    NDRG3 TTLL11 CYB561 KAZN PPM1A SLC25A32 GRAMD4 S100Z
    SNRK YWHAZ FAM168A KIAA1524 CDH4 PAQR5 KCNK12 NSMAF
    RNF169 SLC35F3 HDAC8 KCNAB1 CDKAL1 ZFYVE9 AK7 DDX31
    WDHD1 RNF144B DGKB FKTN C6orf203 EPB41L2 RUNX2 CXCL8
    PLXNA4 TOX2 XPO6 PGM2 TRIM60 PKP2 TWSG1 RGCC
    AZU1 NORAD ARFIP1 SSH2 ALKBH8 TMBIM4 IPO5 TTC39C
    KITLG C11orf87 SCN5A ZCCHC24 FBXO11 RAI14 ABCA8 PRNP
    OC90 STX8 LOC341056 MAGT1 FOS LPA OPHN1 FGF9
    MPP4 IQCJ RPL7L1 ZFAT ABCA13 CSGALNACT2 KIAA0586 RNF114
    TOX G6PC2 BACH2 RGMB C1QTNF3 MOK MED27 WWC1
    SPRED1 C11orf63 C12orf75 HRH1 NTNG1 GGCX ADCK2 PDE7B
    UBASH3B ZNF281 LOC100506797 SLCO4A1-AS1 WDR27 RBM5 AKR1B15 ENKUR
    CACNA1A WDR89 SLIT2 SHTN1 ALK TLE1 FAM107B ELOVL5
    FZD8 CSTF2 XRRA1 ARSF STX18 KIF3C SLC25A12 HIVEP1
    SATB2-AS1 SNX14 IDNK OXCT1 ZNF133 TAPT1 STK38 STRA8
    TMEM18 UTP18 VAPA CCR1 SPPL3 MBP ASAP3 SEMA4D
    TBL1X SMYD3 ITGB1BP1 CRTAM MDM1 TRHR FAF1 STK4
    SMIM19 DNAJA3 PDE8B TSNARE1 KCNV1 AVEN FAM20B CDH13
    KIAA1109 KHDC1 DAP HIPK3 OR10V1 VTI1A FIP1L1 AKAP1
    C20orf85 PPP4R1L IL10 PIK3CB ALG10B ATAD1 ZBTB10 TNRC6A
    COMMD2 MLEC NCK2 FAM171A1 SGPL1 NFATC1 GRB10 NECAB1
    AMOTL1 RHOH HDAC9 PDE4B RFX8 NR2F1-AS1 RNF34 TMSB10
    KYNU TMEM235 SLC26A8 SIK3 CHI3L2 PPP3CA HESX1 CORIN
    ARHGAP18 SYAP1 OLIG2 THG1L MAST2 PPA2 BTBD9 GPR68
    EPB41L1 OLFML2A CFAP36 KLHL5 PRDMS COMMD7 CEP112 SVIL
    C1orf21 PUM2 ST3GAL6 MTCL1 RPAP2 ATG5 PLEKHM3 EDEM3
    SAP18 PANX1 MAB21L2 PTPN20 DSCR9 SIPA1L1 SUMF1 CDK5RAP2
    UBR5 GBF1 UBE3A INHBC EPS15L1 CD226 TCF7 TGFBR2
    HTR7 BCAP29 PRLR USP43 ATP6AP1L RPS6KA5 EXOSC7 RAB10
    KCNG1 CPD KIAA1147 RPS3A CCDC152 ATF7IP CCDC88A CASS4
    ADM2 GTF2H5 FER1L6 DDR2 PARD3 PREP RPL5 C1GALT1C1
    GJD4 WWP2 SVIP FZD4 BPGM ARMC9 ERICH6B MAP1B
    TCP11 PLS3 NT5DC3 CBLN1 C5orf42 LIN7A FIBP TSEN2
    CSNK2A1 UBE2V2 CMTM8 ARHGAP25 KAT7 BLCAP IFI44 TMEM38B
    EDNRA LOC285696 GOLIM4 NEK1 C3orf67 PRDM8 TBXAS1 SND1
    ANAPC10 TSPAN9 ARC ETV1 CTDSPL NDRG1 WWTR1 WASF2
    ADH7 NNT SLC46A3 CTNND2 MBD2 HYPM RNF217 CHST11
    CLDN2 STAG2 INTS6 ZMIZ2 CHSY3 MRPS28 CBFA2T2 BTD
    CEP290 RIN2 COX7A2L TMEM30B WASF3 APCDD1L PARP12 FAM46C
    TCF12 FKBP1A ARFGAP2 PUDP LDHD ADGRL3 TMEM50A TRDMT1
    TSEN15 BAZ2A TANC1 NANS TAOK1 MAPK8 PPP4R3B FAM196A
    OAT AGA DNAH6 ARHGEF4 PSMC4 ANTXR2 BASP1 TPTE2P1
    OR2AT4 MMAB DENND2D C7orf73 ST18
  • TABLE 2
    CHIPSEQ_CDK19-KD ENHANCERUP
    HLCS EFCAB13 FBXL20 AGR2 ABCC11 MFSD7 RIC8B KCNT2
    IGF1 SLC12A8 AZIN1 LYSMD4 AVIL ATP2B4 ASS1 MARCKSL1
    CDYL CRABP2 ERCC8 OSR2 CASQ2 ACTL7B TNFRSF11A NAV2
    LHFPL2 TEX35 SLC22A16 LUM PRKCZ RDH16 ERICH2 STPG2
    HGC6.3 PTPRE GPCPD1 BEGAIN BEST3 ABCG1 ZFPM2 SOWAHC
    MYL4 TCF7L2 HAS2 IGSF22 BDKRB1 MYL12A DNAJB11 LOC10050679 7
    NNAT SCAF8 LOC10026816 8 PPP1R36 CDC42EP5 EDN1 SP4 SOWAHB
    NEURL1 TSC1 MIS18A RALGPS1 SH3BP4 C15orf53 GJA4 FOPNL
    RPIA STOM VEGFA AHDC1 DBX1 PHACTR1 ALDH1A3 DACT1
    SLC1A2 SRPX2 PLXNA2 TBC1D14 RAD23B MAP1A ECHDC3 GLI2
    IQSEC1 ANKRD16 CHAT MAGEF1 NOL6 SUB1 RFK CHRNE
    DENND3 NEK6 S1PR1 C12orf76 DIEXF DHRS9 ERICH5 SCCPDH
    TAF1B XPR1 RYBP ANP32C MCHR1 DLX4 OSBPL11 ARHGAP12
    FGD2 SNTG1 PTGER4 AGMO PTRHD1 FANCA AES KRBA2
    ZC3HC1 TRIM24 HMHB1 IRF2BP2 INPP5F CACNG2 HHLA3 CFI
    TTLL5 ACBD3 PLB1 EDIL3 IGFN1 TROAP HAUS8 NOV
    HPSE2 YARS PROC LEPROTL1 EFHD1 GALNT12 KANK4 JAK3
    TMEM170 B DCLK1 PTPRN SPATA16 CCDC97 ZNF787 TPRG1 DAPK3
    KIF25 LMCD1 AADACL4 RFXAP ALOX5AP BIRC7 GBA3 C1R
    TMPRSS5 TMEM100 OR1M1 ENO2 PTPN3 FAM196B CLEC14A TSPAN 1
    NPC1L1 TBL1X PTPRR LOC10013087 2 FAM136A HSPH1 STK17B GSTA3
    ACKR3 OPTC CREB5 PHTF2 SMIM20 SPRED2 SHE AGAP1
    CTAGE1 KIF16B TRAF4 FAM57A KIAA1211L CORO6 SPNS2 MAOB
    SOAT1 TRIB1 KCTD4 CELF2 TWIST2 C19orf38 TMEM40 THEM4
    GSX2 ADAT2 USF2 NRP2 NSUN7 SEMA3E ZNF462 SUGCT
    BCAT1 CSNK1A1 RAD51AP2 FFAR4 NINJ1 SHH SPIB PSAT1
    CLDN1 ERGIC1 SLC15A1 KISS1 C11orf49 NAT2 HECW1 EXOC6B
    KLHL31 YAE1D1 PIM3 DGKZ MEF2A USB1 CAB39L PCSK1N
    MAST2 STON2 HIP1R ELF3 C4orf26 ZNF429 DISC2 CENPB
    MTCH1 PALLD GLRA3 ZSWIM3 NMBR C14orf37 GPR108 GSTP1
    ATP1A2 RBM47 SORBS3 RAB14 RPS29 ACVR2A C11orf94 DAW1
    THADA CKAP4 SF3B5 ANO6 BTBD16 XRCC2 OTOS EMX1
    EPS8L3 PTK2 ZNF318 RTN2 CAMK2D MRPL4 VGLL3 LMNTD2
    CAB39 PAPSS2 TRIML1 ZSCAN18 HCAR1 RPS3A FAM81A FIZ1
    NEK2 EHF NEDD4L SYT2 LEPROT MAP7D3 PRTFDC1 SEC14L5
    HYI SLC44A1 BAG1 GFI1 MFSD4B GCG PPEF1 LAM B4
    NANOS1 YWHAEP7 ATG9B GCNT3 ATXN1 LIMD1 P2RY1 TMEM120B
    SLC37A1 GRHL3 OTUD3 VWA2 IGFL1 P2RX7 TLR10 KIAA1324
    MAPK8IP1 SLC2A8 RHOB CAMSAP2 TMEM95 FAT1 TFAM APIP
    PPM1L ETS2 KPNA7 HRK ACOT11 RGS7 TMEM106B CERS4
    NXPH2 SLC30A6 RREB1 EML5 WFIKKN2 PAK1 FJX1 HMGCR
    RCAN1 GUCY1A2 LAMC3 RBFOX2 BMP6 DSG3 PITPNM3 ISL1
    PACSIN2 TSN BCOR HES1 NIPBL STAT4 CDH3 PSG2
    SLC39A10 XIRP1 NAB1 DYNC2H1 TMEM51-AS1 ARRB2 CCL20 MINK1
    MRPL15 LY86 PLEKHA1 M ETTL6 LRRC8D SPR SCRT2 RALA
    MAPK1IP1 L EGLN3 CRISPLD2 PAPLN MOAP1 COL24A1 MYO5C SLC28A3
    MAP3K7CL RB1CC1 SERPINB10 TPD52L1 PPARA MZT1 ATP8B2 RASSF6
    PIGU ADTRP CYP1B1 LRRFIP2 NLN ZC3HAV1L NECTIN1 CELA2A
    SYT14 CDCA4 FBXO3 ASCC3 SH2B2 C3orf58 ENOX2 PLEKHG4
    DAAM1 TINAGL1 YIPF6 GPR135 ZNF160 ANXA1 ERCC3 SLC39A11
    CDKN3 CBX4 RALGDS TUBA1A PMAIP1 MN1 ADAMTS10 FGFR3
    EPAS1 ZCCHC10 LRRC4C DUSP18 CXCR5 CRABP1 MAST3 ABLIM2
    INO80C TLR2 AKAP10 RASAL2 NR4A1 PNOC SCN3A NOCT
    DDC TACC2 IFNLR1 COL4A5 FOXQ1 DSG2 PPFIBP2 MAD2L1
    FILIP1L ASH2L TJP3 NID2 DAOA-AS1 CAPZA2 RMND5A SLC8A2
    STC1 DDX47 RXFP3 COL6A3 PDE8A RGS1 TMEM119 MXRA5
    KCTD16 WDFY3 EMILIN2 PSAP SETBP1 GPRC5C MAST4 DNAH1
    RPUSD4 KCNJ15 CCDC9 COX6B2 MEDAG IL6R NUAK1 ZP4
    CD276 EVA1C DPEP2 ABHD5 MRPS22 GLDN RPH3AL AQP7
    LRRFIP1 GHSR NME9 SALL4 F5 MCOLN3 GPATCH1 VSTM2L
    PDLIM1 KIAA0753 STK39 TNFRSF11B HSPBAP1 SLC9B2 PEX26 CNGB1
    CDKAL1 SLC34A3 KERA UBAP1 JADE1 IQCA1 FKBP6 SARM1
    DLX1 P3H2 ITPR2 BTBD10 FBLN2 HES2 C1orf100 KRT10
    NEDD9 GATA6 PLAC8 FAM198B FBP2 BSN SPINK2 PFKP
    C11orf88 SIN3B ORMDL3 TBX21 GPR173 KAZN KIAA0040 HDAC11
    FAM96A DCHS2 UPF2 KCNMA1 PLA2G4E ARL4C HCN3 COL14A1
    BEST1 ACSBG2 NPFFR1 TMEM178A CDC123 NDUFA12 CDNF RBM45
    CBLB PIM1 CTSO DUSP6 LHFPL5 BCL2L10 DIXDC1 TCF12
    TNFAIP8 PPM1H SMARCD3 RAD54B C4orf45 CREB3L2 NPVF OR6B1
    HMGCS1 SSR3 CXCL13 TTC8 MAPRE3 SLC2A6 SERPINB7 HTR3A
    USP36 VIT C17orf99 CYP27C1 NFKBIZ AHCYL2 DHRS7C KRT32
    COL19A1 NOL10 MUC1 SYT17 GRHL1 DENND2C CLCA1 WNT11
    INTS10 CRELD2 LGR6 FHL1 ARRDC5 PARVB CRK NECAP2
    KRTAP4-5 CLMP NCF2 GGT7 INSR PIK3C2B C11orf65 CLUAP1
    MBL2 ASPSCR1 YBX3 SLC35F2 NEMP2 CLDN22 DAB2IP MAMDC2
    IL37 CDCA7 GAREM1 DCN ATP12A REPS2 FAM216B CTTN
    KLHDC9 CDKL2 AGRN ATP9A OXT OLIG2 TSEN54 KIRREL2
    HECTD1 ME3 INTU PGRMC2 RFX2 DSPP LSM8 TEX9
    MYEOV POLR1A PKIG NEBL SOX4 HFM1 OSBPL5 TANK
    CALHM3 TLE1 TRIM66 SACS SLC10A7 MTM1 KLHL38 SHCBP1L
    SRP19 TMC1 TOR2A FNDC3B XIAP JARID2 ALG1L NYX
    BMP10 TRAF3IP2 WISP2 SCN1B C15orf56 ARHGEF3 EPB41L4A AFG3L2
    MON1A PSMA6 TRIP13 ACTR10 GJD3 EFCAB11 IRF4 SLC22A23
    INPP4B HNF1B KIAA1522 ALPP KRT37 MMP24 CMTR2 ARHGAP29
    SSUH2 NUBPL RRAD CDH2 CREB1 ZNF621 APOBEC1 HIPK3
    METAP1D SPOCK2 PTPN1 INHBE ACHE UGP2 PITX2 RPS5
    PTAFR P2RX4 GJB4 PRKCSH C12orf71 ZNF292 PKP1 MAOA
    YPEL5 NKAIN1 KCNG1 EXD1 KRT39 STAU2 AFAP1L2 MIER3
    ATG14 HRH1 CYB5B QPCT TRAK2 IL12B AP4S1 ACSL1
    LAMA3 GJA1 OR51B6 FOXS1 RPTOR VAPA ASIP SPIN1
    ZNF542P THRB COX11 RPS23 PIGC CREBL2 PIN1 UNC13A
    GPBP1 ATOH8 PPFIA2 DMKN ZBTB43 INPP5K STRA6 ABR
    DEF6 DACH1 LILRB3 POLE4 CAPZA3 SNX13 MMP16 MOGAT2
    NRCAM NRARP GATA2 TMEM65 FBN2 INTS7 INPP4A TMEM38B
    C10orf67 ATXN7L1 GPM6A GSTZ1 GARNL3 USP38 AKTIP NR2F1-AS1
    KMT5B LGALS9 ZFP36L2 CD200R1L GPATCH2 DLL1 CNGA2 GAS7
    RIPPLY3 FAM161A C10orf113 TRPC4 RAB27B CD109 TNS3 CDHR2
    TNFRSF21 FAM50B CAGE1 RNF220 ARFGEF3 RALB INTS1 VWA3B
    SLC30A1 ITCH UPP2 LZTS3 YTHDF1 AKR1D1 TAS2R16 DPF3
    IL15 LGMN ST8SIA4 PROSER2 SHC3 PLEKHH1 GRIN2D CCDC184
    PLEKHG6 METTL25 CYP26B1 SHC4 GSG1L BMPER C3orf38 STEAP4
    FA2H TMEM88B PPARGC1A IGFBP3 HSD17B14 RNF112 CXCR4 TESC
    AHR CDK14 CD36 CLTC TFCP2 PRRX2 FOXD2 ATP6V1H
    GALNT15 FSCB YTHDC2 C10orf35 ZNF92 COMMD1 0 RPTN RGS11
    C9orf135 IFT81 TTI1 AMTN LPIN1 IRS2 SLTM MYO1G
    FGFBP1 LRRC25 RPS6KA3 BCR EEPD1 RSPH1 LAMC1 KLF5
    HRASLS2 FOXN3 ANO10 GTF2E2 TRIM9 PAPPA RABEP2 PCLO
    ATF3 PAFAH1B2 PRSS57 FAM3B CYP24A1 CARTPT NPEPPS NTF3
    TLR3 CABLES1 SLN ANGPT1 TUFT1 CLCNKA ANXA4 NCALD
    LTBP1 CPPED1 DYNLT1 MREG C4orf19 AIG1 THNSL2 RBM3
    SMG6 OR2S2 AMER3 NAV3 RNF13 PPP1R2 SLC43A3 WIPI2
    PANK1 TAMM41 ST6GALNAC4 TRIM54 NAT1 PTPRC COX20 CCDC65
    NRDE2 SPAG9 DCTN6 GABBR2 UPP1 ARHGEF18 C1orf226 TTC39C
    TGFB3 EIF4A2 UNC5A TPK1 LBH PRKAR1A SERBP1 TOPBP1
    LARP4B SERPINB1 C12orf74 GOSR2 OSCP1 PSTK KNOP1 C1orf228
    CD9 ACSL5 CT62 DENND5A BAIAP3 SLIT2 NFATC1 DNAJC6
    ADAMTS6 ADAM29 FRMD3 RAB8B OR10H1 RAB11FIP 4 TMEM45B FABP3
    NNMT WDFY1 CEP152 ARHGAP42 HIC1 SHQ1 TARBP2 SNX7
    C10orf90 RAD51B LRRC20 GNLY TIMM22 SMARCA2 DUSP8 SOX8
    NCOA6 CCNY COX7A2 TRAF3IP1 SLC6A3 KBTBD12 ARHGAP39 MDFIC
    PPM1B SEMA3A PLA2G2E DNAH11 EGR4 DUSP27 TMPRSS7 NFE2
    CACUL1 TMEM86B TRABD2A REEP3 NDUFB11 CCDC186 C7orf57 CC2D2A
    LEKR1 ATP6V1G1 MPZL2 SLC4A1 MGAT4C TMPRSS9 COL21A1 LBP
    TMEM247 CCDC34 IGF2R CLDN4 WNT7A APBB1IP CPA4 DTWD1
    NSMCE4A GDPD5 ANKRD33 PLEKHG3 CYP11A1 CDCA7L NID1 MAF
    NUP155 CAMK2B ZEB2 ARID5A FRMD4B ATXN7 LSM3 FGGY
    ABI3BP PNPLA5 ATXN3L ZNF396 SOCS2 M BTPS2 HS1BP3 C9orf3
    MUC20 IL7R FIGN PPP2R2C USP2 ENKUR RALBP1 MRPS18A
    CNIH3 ULK1 ADGRF1 FLJ23867 CRIP2 PTHLH FAM187B SH2D3C
    SH3GL3 ODC1 LGALSL PRRC1 GC NEK10 MAMLD1 C4o rf3 2
    SH3TC1 LGI1 SLC6A20 CD180 PLCE1 THY1 CTSH SLCO4A1
    SLC26A9 MPL AACSP1 COL26A1 SSR2 CMAHP ID4 ALCAM
    TAGLN2 COBL SCNN1G TRAF7 MYOZ1 AKR1C3 CER1 AREG
    ABCG2 DCK CCDC174 PRKCE CBLC SYNM BCAS2 BDKRB2
    NABP1 TBC1D1 DHRS3 TES USHBP1 UBQLN4 ETV7 CCR8
    AGPAT2 MLXIPL SLC13A1 ADAD1 NOSTRIN QRFPR RHOBTB1 SCFD2
    PPP4R3A E2F6 CDK4 PABPC1P2 COL5A3 RAB31 PPP1R14D CASK
    ADAMTS15 CRTAC1 HRC KCNQ4 UBE3D MIEN1 KIF18A BPI
    KIAA0895 SIM2 LITAF RNF165 CCDC77 DIO2 ABCA6 ZNF473
    FHAD1 BRINP1 GRAMD1C TAF1L EMC7 TRIM29 AGXT2 CD300LF
    PPP1R12B GPR37L1 WAPL AQP3 LZTFL1 YIPF5 ENOX1 ZC2HC1A
    GIN1 FHDC1 PBOV1 DERA FGD4 TYK2 ACP6 NLGN1
    ULK4 BANK1 PER1 ITGA2 LLGL2 ALDH8A1 FBRSL1 TPPP3
    TNNI2 TMEM167A RGS4 PDGFB ZDHHC17 APOBEC2 THBD HGF
    BTN3A1 EXOC3-AS1 NAA20 VAV1 ZNF664 TRMO TMEM139 PRR15
    PHLPP1 GINS2 GMDS PCDH1 PARD3B MYH13 C1orf43 ARSB
    TMEM217 SLC22A2 IL1RN FMN1 KCNJ12 RASAL3 HTR1B PCDH8
    BRDT NEK7 MCM10 NPSR1-AS1 ARID5B SEMA3C UBB TACR2
    VSX1 LOC10013221 5 MMD2 MEF2C SPON1 FLVCR2 SNX25 GLOD5
    STK38L ZNF555 YKT6 NR5A1 DNAJC10 SYNPO2L APEH ALDH3A1
    DPYSL2 ETFB GCM2 FGF19 GRN GNAS FCHO1 DBN1
    TCEANC SOCS6 CEP128 RBM24 HEATR5A ASAH1 CHMP6 RPS26
    MRVI1 PLA2R1 CDC14B SCARB1 SLC7A10 SLC13A2 WDR89 VPS45
    INSIG2 GJA3 MCM5 TRPS1 CHMP4B ZNF366 SRMS CNTN6
    MYO5B AGTPBP1 TMC8 FAM173A PITX3 TRAFD1 PNPLA8 CD28
    YWHAQ C9orf116 SLC16A3 VPS37D ASB5 JSRP1 UGT8 WBP2NL
    TSPAN2 EGFR SRD5A3 CDC16 NDUFA10 SPOPL NR5A2 ZC3H4
    KLF4 C9orf153 GADD45A C18orf12 EMX2 BMF PPP2R5A MKL2
    TIMM17A CMIP METTL4 FEM1C ST6GALNAC 5 PIWIL3 SRL CCBE1
    CIT ASB7 C15orf54 TMEM71 TGIF1 ARVCF MEGF6 TPPP
    TNFRSF19 RAB11FIP1 MRPS36 FTH1 ETS1 MAN1A1 PELO OXER1
    DYM SLC23A3 MMP20 KCMF1 TRY2P RPS6KA5 NPAS2 SLC25A19
    CCDC112 SOX9 RGS9 NUTF2 OXSR1 MAGEB2 AVP TMEM59
    C9orf50 ABHD11-AS1 GPR132 PLCD1 NATD1 OTUD1 PLA2G4D BHLHE41
    AAED1 TMIE NDUFB6 SPCS3 PRRG4 GCLC CEACAM22P LIN28A
    KIF5C PHLDB1 E2F8 EPHA5 CITED2 SLC5A1 TBC1D23 PLEKHG4B
    BANF2 GLP2R HSD17B2 PTPRK SLC7A7 SLC9A7 SNX9 SND1-IT1
    OLA1 PEBP1 TAPT1 LOC401052 CLIC5 CPEB4 KDM4C SLC20A1
    RAPGEF2 SGK1 TANGO6 SNCB SEMA3D FLRT2 NTRK2 LEPR
    C9orf131 IFI6 LVRN ZNF214 C14orf2 SSFA2 PABPC4L TMEM244
    C1QTNF1 TMC5 WDR18 BRMS1L CTNNB1 PDE1A SH3PXD2B NTN4
    LIMCH1 PSD3 SLC38A11 HTRA1 DIRAS1 EPHB6 HTRA3 PTGIR
    YY1AP1 TFAP2A GTPBP4 ARFGAP3 LDHAL6A ZNF331 EPC1 SNRPC
    CREG2 ZBTB7C CDK20 KIAA0825 RXFP2 GPR182 CASZ1 ZBED2
    ASAP2 INPP5A UBE2O WNT7B TNFRSF8 RANBP3L SORBS1 GUSB
    CFAP126 SNHG7 COL18A1 CACNA1A FKBP8 TEKT3 RPEL1 GNAT2
    FAM107B LCA5 MAP1S RHOD ADSSL1 SLC8A1 PKP2 ABHD15
    FAM86B3P SNRNP35 SLC1A4 CLDN23 INHBB FAM110B TMEM207 HMCN1
    ADAM12 PRF1 CD38 METRNL OPCML RAP1GAP2 IQUB TP63
    RECQL5 PIK3R1 KRT20 CYP1A1 DUSP14 FTHL17 EPYC CCDC134
    B4GALNT2 FOXE1 ADAMTSL1 SCIN POPDC2 NXNL1 RFX7 VTCN1
    CPA2 IL21 PPP2R2A RAPGEF4 ARNT2 GSN SIGLEC8 LRRC29
    ZNF385B NLRC5 FUZ CCR3 VLDLR MELTF BDNF ACSL3
    ZNF488 FRAT2 BATF3 C11orf96 SULT4A1 ITGAV ADGRL3 SKIL
    SIRT4 MORN3 RIPK2 KLLN MYO6 MTCL1 UBA7 JPH2
    DYSF TYROBP CCDC83 RHOU NFIL3 FKBP11 LRPAP1 CLDN10
    ERP44 IPMK LTBP4 BBS10 RNLS SPAG17 YOD1 BPTF
    FERMT2 SYT12 CCDC150 S1PR2 PRSS41 FAM120B TPH2 CDKL3
    SFXN4 NDRG4 FAM171A1 ANKRD10 SLC29A3 IRAK3 KCNA10 ZBTB16
    MICAL3 C5orf51 NAA16 EDNRA PRMT9 DCST1 PDC VCL
    RAD51C OSER1 SFRP2 VSTM5 BCLAF1 CXCL16 BFSP1 SHISA2
    LGALS3BP SCG2 TYMP NENF TEX36 C17orf107 ST6GALNAC 1 C5orf30
    KCNJ6 AGTR2 SHANK2 GPR156 MICALL1 ZNF608 CCDC63 AQP9
    MSX2 GPC1 GFPT1 GPRC5B LACC1 NPFFR2 FBXO7 PARP11
    TIGD2 ANKRD9 LRRN3 UBASH3A CCDC68 TDRD7 ARHGAP24 SH3BGRL2
    PNMA2 SLC1A3 ABCA13 CIPC SPIRE2 H3F3C EFHC2 VILL
    CACNA1H KCTD12 UBE4B NYAP2 DUSP23 CCDC124 RHOBTB2 ERBB4
    RAB35 ITPK1 PIK3R3 SPTSSA MMP27 UBASH3B PYM1 SPAG16
    TOMM5 TLE6 MRPL21 JPH1 PKD1L2 TMEM94 LANCL3 IL2RG
    FUNDC1
  • The aforementioned GSEA also enabled us to identify the leading edge ‘core’ genes that contribute the most to each enrichment (FIGS. 12A and 12B). At these ‘core’ genes, differences in H3K27Ac enhancer signals due to CDK19 knockdown (FIGS. 13A-13D) result in large corresponding changes in gene expression (FIG. 13E). The gene tracks at the ELF3 (FIG. 13A) and ETV7 (FIG. 13B) loci show enrichment of H3K27Ac signals in the CDK19 knockdown samples, whereas the gene tracks at the CHI3L2 (FIG. 13C) and CRTAM (FIG. 13D) loci show enrichment of H3K27Ac signals in the Control samples. Upper tracks denote Control samples, while lower tracks denote CDK19 knockdown samples. Gray bars denote regions identified by DiffBind to be different between control and CDK19 knockdown samples (FDR < 0.05). Metascape analysis was then used to evaluate Hallmark gene sets enriched within the CDK19KD-EnhancerUP ‘core’ and the CDK19KD-EnhancerDOWN ‘core’ genes. Within the CDK19KD-EnhancerUP ‘core’ genes, early Estrogen Response (p-value = 8.72e-5) and Epithelial Mesenchymal Transition (p-value = 1.08e-3) were Hallmarks identified as enriched (FIG. 3G, dark gray bars). Similarly, within the CDK19KD-EnhancerDOWN ‘core’ genes Androgen Response (p-value = 1.89e-3) was the Hallmark found to be enriched (FIG. 3G, light gray bar). Thus, a subset of genes (FIG. 3G) within the early Estrogen Response, Epithelial to Mesenchymal Transition, and Androgen Response gene sets have changes in H3K27Ac enhancer signals and strong corresponding changes in gene expression. These genes constitute a small fraction of the total genes in each Hallmark gene set (5-10%), but highlight key genes within these biological processes where CDK19 can epigenetically regulate gene transcription.
  • 4.7 Example 7 - Effects of CDK19 Knockdown on the Growth of Pre-Established Organoids
  • We explored the effect of CDK19 knockdown on the growth of pre-established organoids in vitro and in pre-established PDX tumors in vivo. This aimed to model the treatment of patients’ pre-existing tumors. In vitro, adding doxycycline to the treatment group (to induce CDK19 shRNA) significantly reduced the number of pre-established organoids compared to the control (no doxycycline) (FIGS. 4A and 4B). In FIGS. 4A and 4B, number of organoid colonies at Day 0 (FIG. 4A) and Day 16 (FIG. 4B) after initiating doxycycline treatment is shown, ****P < 0.0001; ns is P > 0.05 (mean ± s.d., n = 6, experiment performed twice, P values determined by unpaired t-test). In vivo, feeding doxycycline to mice with pre-established inducCDK19KD-PDX-T1 or inducCDK19KD-PDX-T3 (PDX-T3 cells transduced with a doxycycline-inducible CDK19 knockdown construct) tumors significantly impacted the growth of these tumors (FIGS. 4C and 4D). In FIGS. 4C and 4D, the growth of pre-established tumors in the doxycycline fed NSG mice and control NSG mice are shown for inducCDK19KD-PDX-T1, ****P < 0.0001 ; ***P < 0.001 (mean ± s.d., n = 5, experiment performed twice, P values determined by unpaired t-test) (FIG. 4C) and inducCDK19KD-PDX-T3, ****P < 0.0001; ***P < 0.001 (mean ± s.d., n = 5, experiment performed once, P values determined by unpaired t-test) (FIG. 4D). CDK19 shRNA induced tumors were ultimately 82% smaller in inducCDK19KD-PDX-T1 tumors and 38% smaller in inducCDK19KD-PDX-T3 tumors when compared to control tumors (FIGS. 4C and 4D). In both inducCDK19KD-PDX-T1 and inducCDK19KD-PDX-T3 experiments, mouse total body weights were not significantly different between the treatment and control groups (FIGS. 14A and 14B). Finally, survival studies showed that overall survival was significantly longer in mice whose PDX-T1 tumors were transduced with CDK19 shRNA compared to mice transduced with control shRNA (FIG. 4E). Shown in FIG. 4E are Kaplan-Meir survival curves for mice engrafted with PDX-T1 xenografts transduced with control shRNA (black line), shCDK19-1 (solid gray line) or shCDK19-2 (dashed gray line). Mice were followed with weekly measurements of tumor diameters. Mice were sacrificed when the longest diameter of their tumor exceeded 17 mm. Two mice in the shCDK19-2 group did not develop PDX tumors and were sacrificed at the end of the experiment. These mice were censored when constructing the survival curve for the shCDK19-2 group, ***P < 0.001 (n = 9, experiment performed three times, log-rank (Mantel-Cox) test used to determine P values). In summary, these experiments showed that even in pre-established tumors, specifically knocking down CDK19 can significantly decrease tumor growth and that CDK19 knockdown can prolong survival in mice.
  • 4.8 Example 8 - Effects of CCT251921 on Pre-Established PDX Tumors
  • To model the use of a CDK19 targeted therapy clinically, we treated mice with pre-established PDX tumors with CCT251921 (FIG. 4F), an orally bioavailable inhibitor of both CDK19 and the closely related paralog, CDK8. PDX-T1 tumors were pre-established in mice before starting daily oral administration (30 mg/kg) of CCT251921 or vehicle. Treatment with CCT251921 resulted in a significant reduction in tumor growth by day 14 (FIG. 4G). Final volumes of the tumors in CCT251921 treated mice were over 30% smaller than the tumors of vehicle treated mice (FIG. 4G). NSG mice with pre-established PDX-T1 xenograft tumors were treated with daily oral gavage of CCT251921 or vehicle. Mice were followed with twice weekly determinations of tumor volume, ****P < 0.0001; ***P < 0.001 (mean ± s.d., n = 5, experiment performed once, P values determined by unpaired t-test). Mice in both the CCT251921 and vehicle cohorts suffered an overall weight loss, but this was not significantly different between the two groups and most likely due to the effect of daily oral gavage on their feeding habits (FIG. 14C). It is well known that different biological outcomes can arise from gene knockdown versus chemical inhibition. We show here in pre-established tumors that chemical inhibition of CDK19 kinase activity can recapitulate the effects of total CDK19 loss shown in our knockdown studies.
  • From our data, we conclude that CDK19 regulates multiple cancer relevant pathways and that it is a potential therapeutic target in TICs. Thus, CDK19 inhibition is useful both to therapeutic strategies targeting transcriptional co-factors such as CDK8, CDK9, and BRD4, and to those targeting TICs and their self-renewal pathways such as Hedgehog, Wnt/β-catenin, and Notch. However, some therapeutic approaches may be limited by toxicity caused to normal cells. This can be attributed to the ubiquitous expression of transcriptional co-factors in normal tissues and the importance of self-renewal pathways in normal stem cells. BRD4 inhibition, for example, resulted in a disruption of tissue homeostasis in multiple organs in mice. Similarly, due to the challenge of narrow therapeutic indices, Hedgehog, Notch, and Wnt pathway inhibitors have had limited clinical success thus far. The biology of CDK19 points towards potential advantages as a therapeutic target. Compared to other ubiquitous transcriptional co-factors such as its paralog CDK8, CDK9, and BRD4, CDK19 has more limited tissue distribution (see, e.g., Tsutsui et al., Genes to cells : devoted to molecular & cellular mechanisms 16:1208-1218, 2011), potentially limiting the toxicity from CDK19 inhibition, while CDK8, CDK9, and BRD4 knockouts are lethal (see, e.g., Brown et al., Mamm Genome 23:632-640, 2012; Westerling, Molecular and Cellular Biology 27:6177-6182, 2007; and Houzelstein et al., Molecular and Cellular Biology 22, 3794-3802, 2002). In addition, the limited expression of CDK19 in tissues could broaden the therapeutic window to enable the otherwise toxic inhibition of stem cell pathways such as NOTCH, or critical processes, such as G2/M checkpoint. Our studies showingthat small molecule inhibition of CDK19 impaired PDX growth affirms the potential of therapeutically targeting CDK19 in TNBC.
  • 5. References
  • 1 Bauer, K. R., Brown, M., Cress, R. D., Parise, C. A. & Caggiano, V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer Registry. Cancer 109, 1721-1728, doi:10.1002/cncr.22618 (2007).
  • 2 Tsutsui, T., Fukasawa, R., Tanaka, A., Hirose, Y. & Okhuma, Y. Identification of target genes for the CDK subunits of the Mediator complex. Genes to cells : devoted to molecular & cellular mechanisms 16, 1208-1218, doi:10.1111/j.1365-2443.2011.01565.x (2011).
  • 3 Brown, S. D. & Moore, M. W. The International Mouse Phenotyping Consortium: past and future perspectives on mouse phenotyping. Mamm Genome 23, 632-640, doi:10.1007/s00335-012-9427-x (2012).
  • 4 Diehl, P., Tedesco, D. & Chenchik, A. Use of RNAi screens to uncover resistance mechanisms in cancer cells and identify synthetic lethal interactions. Drug Discov Today Technol 11, 11-18, doi:10.1016/j.ddtec.2013.12.002 (2014).
  • 5 Lee, C. Y. et al. Neuregulin autocrine signaling promotes self-renewal of breast tumor-initiating cells by triggering HER2/HER3 activation. Cancer Res 74, 341-352, doi:10.1158/0008-5472.CAN-13-1055 (2014).
  • 6 Nolan-Stevaux, O. et al. Measurement of Cancer Cell Growth Heterogeneity through Lentiviral Barcoding Identifies Clonal Dominance as a Characteristic of Tumor Engraftment. PloS one 8, e67316, doi:10.1371/journal.pone.0067316 (2013).
  • 7 Cancer Genome Atlas Research, N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113-1120, doi:10.1038/ng.2764 (2013).
  • 8 Broude, E. V. et al. Expression of CDK8 and CDK8-interacting Genes as Potential Biomarkers in Breast Cancer. Current cancer drug targets 15, 739-749 (2015).
  • 9 Porter, D. C. et al. Cyclin-dependent kinase 8 mediates chemotherapy-induced tumor-promoting paracrine activities. Proc Natl Acad Sci U S A 109, 13799-13804, doi:10.1073/pnas.1206906109 (2012).
  • 10 Galbraith, M. D., Donner, A. J. & Espinosa, J. M. CDK8: a positive regulator of transcription. Transcription 1, 4-12, doi:10.4161/trns.1.1.12373 (2010).
  • 11 Robertson, F. M. et al. Inflammatory breast cancer: the disease, the biology, the treatment. CA Cancer J Clin 60, 351-375, doi:10.3322/caac.20082 (2010).
  • 12 Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J. & Clarke, M. F. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100, 3983-3988, doi:10.1073/pnas.0530291100 (2003).
  • 13 Lim, E. et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nature medicine 15, 907-913, doi:10.1038/nm.2000 (2009).
  • 14 Prat, A. et al. Characterization of cell lines derived from breast cancers and normal mammary tissues for the study of the intrinsic molecular subtypes. Breast cancer research and treatment 142, 237-255, doi:10.1007/s10549-013-2743-3 (2013).
  • 15 Scheeren, F. A. et al. A cell-intrinsic role for TLR2-MYD88 in intestinal and breast epithelia and oncogenesis. Nature cell biology 16, 1238-1248, doi:10.1038/ncb3058 (2014).
  • 16 Bachelard-Cascales, E. et al. The CD10 enzyme is a key player to identify and regulate human mammary stem cells. Stem cells 28, 1081-1088, doi:10.1002/stem.435 (2010).
  • 17 Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. Journal of immunological methods 347, 70-78, doi:10.1016/j.jim.2009.06.008 (2009).
  • 18 Sato, S. et al. A set of consensus mammalian mediator subunits identified by multidimensional protein identification technology. Molecular cell 14, 685-691, doi:10.1016/j.molcel.2004.05.006 (2004).
  • 19 Firestein, R. et al. CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity. Nature 455, 547-551, doi:10.1038/nature07179 (2008).
  • 20 Pelish, H. E. et al. Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature 526, 273-276, doi:10.1038/nature14904 (2015).
  • 21 Kapoor, A. et al. The histone variant macroH2A suppresses melanoma progression through regulation of CDK8. Nature 468, 1105-1109, doi:10.1038/nature09590 (2010).
  • 22 Galbraith, M. D. et al. HIF1A employs CDK8-mediator to stimulate RNAPII elongation in response to hypoxia. Cell 153, 1327-1339, doi:10.1016/j.cell.2013.04.048 (2013).
  • 23 Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102, 15545-15550, doi:10.1073/pnas.0506580102 (2005).
  • 24 Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell systems 1, 417-425, doi:10.1016/j.cels.2015.12.004 (2015).
  • 25 Kalimutho, M. et al. Targeted Therapies for Triple-Negative Breast Cancer: Combating a Stubborn Disease. Trends in pharmacological sciences 36, 822-846, doi:10.1016/j.tips.2015.08.009 (2015).
  • 26 Chen, M. et al. CDK8/19 Mediator kinases potentiate induction of transcription by NFkappaB. Proc Natl Acad Sci USA, doi:10.1073/pnas.1710467114 (2017).
  • 27 Audetat, K. A. et al. A Kinase-Independent Role for Cyclin-Dependent Kinase 19 in p53 Response. Molecular and cellular biology 37, doi:10.1128/MCB.00626-16 (2017).
  • 28 Melotte, V. et al. The N-myc downstream regulated gene (NDRG) family: diverse functions, multiple applications. FASEB J 24, 4153-4166, doi:10.1096/fj.09-151464 (2010).
  • 29 Zarnegar, M. A., Reinitz, F., Newman, A.M., Clarke, M.F. Targeted chromatin ligation, a robust epigenetic profiling technique for small cell numbers. Nucleic Acids Research, doi:10.1093/nar/gkx648 (2017).
  • 30 Bae, D. H. et al. The role of NDRG1 in the pathology and potential treatment of human cancers. J Clin Pathol 66, 911-917, doi:10.1136/jclinpath-2013-201692 (2013).
  • 31 Brown, J. D. et al. NF-kappaB directs dynamic super enhancer formation in inflammation and atherogenesis. Molecular cell 56, 219-231, doi:10.1016/j.molcel.2014.08.024 (2014).
  • 32 Nabet, B. et al. Deregulation of the Ras-Erk Signaling Axis Modulates the Enhancer Landscape. Cell Rep 12, 1300-1313, doi:10.1016/j.celrep.2015.06.078 (2015).
  • 33 Tripathi, S. et al. Meta- and Orthogonal Integration of Influenza “OMICs” Data Defines a Role for UBR4 in Virus Budding. Cell host & microbe 18, 723-735, doi:10.1016/j.chom.2015.11.002 (2015).
  • 34 Mallinger, A. et al. Discovery of Potent, Selective, and Orally Bioavailable Small-Molecule Modulators of the Mediator Complex-Associated Kinases CDK8 and CDK19. Journal of medicinal chemistry 59, 1078-1101, doi:10.1021/acs.jmedchem.5b01685 (2016).
  • 35 Poss, Z. C. et al. Identification of Mediator Kinase Substrates in Human Cells using Cortistatin A and Quantitative Phosphoproteomics. Cell Rep 15, 436-450, doi:10.1016/j.celrep.2016.03.030 (2016).
  • 36 Bolden, J. E. et al. Inducible in vivo silencing of Brd4 identifies potential toxicities of sustained BET protein inhibition. Cell Rep 8, 1919-1929, doi:10.1016/j.celrep.2014.08.025 (2014).
  • 37 Takebe, N. et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol 12, 445-464, doi:10.1038/nrclinonc.2015.61 (2015).
  • 38 Westerling, T., Kuuluvainen, E. & Makela, T. P. Cdk8 is essential for preimplantation mouse development. Molecular and cellular biology 27, 6177-6182, doi:10.1128/MCB.01302-06 (2007).
  • 39 Houzelstein, D. et al. Growth and early postimplantation defects in mice deficient for the bromodomain-containing protein Brd4. Molecular and cellular biology 22, 3794-3802 (2002).
  • 40 Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat Methods 9, 676-682, doi:10.1038/nmeth.2019 (2012).
  • 41 Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6, pl1, doi:10.1126/scisignal.2004088 (2013).
  • While the foregoing invention has been described in some detail for purposes of clarity and understanding, it will be appreciated by those skilled in the relevant arts, once they have been made familiar with this disclosure, that various changes in form and detail can be made without departing from the true scope of the invention in the appended claims. The invention is therefore not to be limited to the exact components or details of methodology or construction set forth above. Except to the extent necessary or inherent in the processes themselves, no particular order to steps or stages of methods or processes described in this disclosure, including the Figures, is intended or implied. In many cases the order of process steps may be varied without changing the purpose, effect, or import of the methods described.
  • All publications and patent documents cited herein are incorporated herein by reference as if each such publication or document was specifically and individually indicated to be incorporated herein by reference. Citation of publications and patent documents (patents, published patent applications, and unpublished patent applications) is not intended as an admission that any such document is pertinent prior art, nor does it constitute any admission as to the contents or date of the same.
  • CDK19 Transcript Variant 1 (NM_015076.4) (SEQ ID NO: 12)
  • 1 tgtggccgcc gaggagtccc ttgctgaagg cggaccgcgg agcggcgggc ggcgggcggc
      61 gcgcgcgcgc gcgcgagagg cggctgttgg agaagtggag cggcggtcgc ggggggagga
     121 ggaggaggga ctgagcggcg gcggcccccg cgtcccgtgc ctctatgggg gaagcagaca
     181 atggattatg atttcaaggc gaagctggcg gcggagcggg agcgggtgga ggatttgttt
     241 gagtacgaag ggtgcaaagt gggacgcggc acctacggtc acgtctacaa ggcgaggcgg
     301 aaagatggaa aagatgaaaa ggaatatgca ttgaagcaaa ttgaaggcac aggaatatcc
     361 atgtcggctt gtagagagat tgcacttttg cgagaattga agcaccctaa tgtgattgca
     421 ttgcagaagg tgttcctttc tcacagtgac aggaaggtat ggctgctgtt tgattatgca
     481 gagcatgact tgtggcatat tattaagttt caccgtgcat caaaagcaaa taaaaagccc
     541 atgcagttgc caagatctat ggttaaatcc ttactttacc agattcttga tggtatccat
     601 tacctccatg caaattgggt gcttcacaga gacttgaaac cagcaaatat cctagtaatg
     661 ggagaaggtc ctgagagggg gagagtcaaa atagctgaca tgggttttgc cagattattc
     721 aattctcctc taaagccact agcagatttg gatccagtag ttgtgacatt ttggtatcgg
     781 gctccagaac ttttgcttgg tgcaaggcat tatacaaagg ccattgatat atgggcaata
     841 ggttgtatat ttgctgaatt gttgacttcg gaacctattt ttcactgtcg tcaggaagat
     901 ataaaaacaa gcaatccctt tcatcatgat caactggatc ggatatttag tgtcatgggg
     961 tttcctgcag ataaagactg ggaagatatt agaaagatgc cagaatatcc cacacttcaa
    1021 aaagacttta gaagaacaac gtatgccaac agtagcctca taaagtacat ggagaaacac
    1081 aaggtcaagc ctgacagcaa agtgttcctc ttgcttcaga aactcctgac catggatcca
    1141 accaagagaa ttacctcgga gcaagctctg caggatccct attttcagga ggaccctttg
    1201 ccaacattag atgtatttgc cggctgccag attccatacc ccaaacgaga attccttaat
    1261 gaagatgatc ctgaagaaaa aggtgacaag aatcagcaac agcagcagaa ccagcatcag
    1321 cagcccacag cccctccaca gcaggcagca gcccctccac aggcgccccc accacagcag
    1381 aacagcaccc agaccaacgg gaccgcaggt ggggctgggg ccggggtcgg gggcaccgga
    1441 gcagggttgc agcacagcca ggactccagc ctgaaccagg tgcctccaaa caagaagcca
    1501 cggctagggc cttcaggcgc aaactcaggt ggacctgtga tgccctcgga ttatcagcac
    1561 tccagttctc gcctgaatta ccaaagcagc gttcagggat cctctcagtc ccagagcaca
    1621 cttggctact cttcctcgtc tcagcagagc tcacagtacc acccatctca ccaggcccac
    1681 cggtactgac cagctcccgt tgggccaggc cagcccagcc cagagcacag gctccagcaa
    1741 tatgtctgca ttgaaaagaa ccaaaaaaat gcaaactatg atgccattta aaactcatac
    1801 acatgggagg aaaaccttat atactgagca ttgtgcagga ctgatagctc ttctttattg
    1861 acttaaagaa gattcttgtg aagtttcccc agcacccctt ccctgcatgt gttccattgt
    1921 gacttctctg ataaagcgtc tgatctaatc ccagcacttc tgtaaccttc agcatttctt
    1981 tgaaggattt cctggtgcac ctttctcatg ctgtagcaat cactatggtt tatcttttca
    2041 aagctctttt aataggattt taatgtttta gaaacaggat tccagtggtg tatagtttta
    2101 tacttcatga actgatttag caacacaggt aaaaatgcac cttttaaagc actacgtttt
    2161 cacagacaat aactgttctg ctcatggaag tcttaaacag aaactgttac tgtcccaaag
    2221 tactttacta ttacgttcgt atttatctag tttcagggaa ggtctaataa aaagacaagc
    2281 ggtgggacag agggaaccta caaccaaaaa ctgcctagat ctttgcagtt atgtgcttta
    2341 tgccacgaag aactgaagta tgtggtaatt tttatagaat cattcatatg gaactgagtt
    2401 cccagcatca tcttattctg aatagcattc agtaattaag aattacaatt ttaaccttca
    2461 tgtagctaag tctaccttaa aaagggtttc aagagctttg tacagtctcg atggcccaca
    2521 ccaaaacgct gaagagagta acaactgcac taggatttct gtaaggagta attttgatca
    2581 aaagacgtgt tacttccctt tgaaggaaaa gtttttagtg tgtattgtac ataaagtcgg
    2641 cttctctaaa gaaccattgg tttcttcaca tctgggtctg cgtgagtaac tttcttgcat
    2701 aatcaaggtt actcaagtag aagcctgaaa attaatctgc ttttaaaata aagagcagtg
    2761 ttctccattc gtatttgtat tagatataga gtgactattt ttaaagcatg ttaaaaattt
    2821 aggttttatt catgtttaaa gtatgtatta tgtatgcata attttgctgt tgttactgaa
    2881 acttaattct atcaagaatc tttttcattg cactgaatga tttcttttgc ccctaggaga
    2941 aaacttaata attgtgccta aaaactatgg gcggatagta taagactata ctagacaaag
    3001 tgaatatttg catttccatt atctatgaat tagtggctga gttctttctt agctgcttta
    3061 aggagcccct cactccccag agtcaaaagg aaatgtaaaa acttagagct cccattgtaa
    3121 tgtaaggggc aagaaatttg tgttcttctg aatgctacta gcagcaccag ccttgtttta
    3181 aatgttttct tgagctagaa gaaatagctg attattgtat atgcaaatta catgcatttt
    3241 taaaaactat tctttctgaa cttatctacc tggttatgat actgtgggtc catacacaag
    3301 taaaataaga ttagacagaa gccagtatac attttgcact attgatgtga tactgtagcc
    3361 agccaggacc ttactgatct cagcataata atgctcacta ataatgaagt ctgcatagtg
    3421 acactcatca agactgaaga tgaagcaggt tacgtgctcc attggaagga gtttctgata
    3481 gtctcctgct gttttacccc ttccattttt taaaataaga aattagcagc cctctgcata
    3541 atgtagctgc ctatatgcag ttttatcctg tgccctaaag cctcactgtc cagagctgtt
    3601 ggtcatcaga tgcttattgc accctcacca tgtgcctggt gccctgctgg gtagagaaca
    3661 cagaggacag ggcatacttc ttgtccttaa ggagcttgtg atctgtgaca gtaagccctc
    3721 ctgggatgtc tgtgccatgt gattgactta caagtgaaac tgtcttataa tatgaaggtc
    3781 tttttgttta cttctaaacc cacttgggta gttactatcc ccaaatctgt tctgtaaata
    3841 atattatgga agggtttcta tgtcagtcta ccttagagaa agccagtgat tcaatatcac
    3901 aaaaggcatt gacgtatctt tgaaatgttc acagcagcct tttaacaaca actgggtggt
    3961 ccttgtaggc agaacatact ctcctaagtg gttgtaggaa attgcaagga aaatagaagg
    4021 tctgttcttg ctctcaagga ggttaccttt aataaaagaa gacaaaccca gatagatatg
    4081 taaaccaaaa tactatgccc cttaatactt tataagcagc attgttaaat agttcttacg
    4141 cttatacatt cacagaacta ccctgttttc cttgtatata atgacttttg ctggcagaac
    4201 tgaaatataa actgtaaggg gatttcgtca gttgctccca gtatacaata tcctccagga
    4261 catagccaga aatctccatt ccacacatga ctgagttcct atccctgcac tggtactggc
    4321 tcttttctcc tctttccttg cctcagggtt cgtgctaccc actgattccc tttaccctta
    4381 gtaataattt tggatcattt tctttccttt aaaggggaac aaagcctttt ttttttttga
    4441 gacggagtgt tgctctgtca cccaagctgg agtgcagtgg cacgatcttg gctcactcca
    4501 acctccacct tccaggttca agtgattctc ctgcctcagc ctcccgagta gctgggacta
    4561 cgggcacgca ccaccacgtc tggctaattt ttgtattttt agtagagatg gggtttcacc
    4621 ctattggtca ggctggtctt gaattcctca cctcaggtca tccgcctgtc tcggcctccc
    4681 gaagtgctgg gattataggt gtgagccacc gcacccagtt gggaacaaag cctttttaac
    4741 acacgtaagg gccctcaaac cgtgggacct ctaaggagac ctttgaagct ttttgagggc
    4801 aaactttacc tttgtggtcc ccaaatgatg gcatttctct ttgaaattta ttagatactg
    4861 ttatgtcccc caagggtaca ggaggggcat ccctcagcct atgggaacac ccaaactagg
    4921 aggggttatt gacaggaagg aatgaatcca agtgaaggct ttctgctctt cgtgttacaa
    4981 accagtttca gagttagctt tctggggagg tgtgtgtttg tgaaaggaat tcaagtgttg
    5041 caggacagat gagctcaagg taaggtagct ttggcagcag ggctgatact atgaggctga
    5101 aacaatcctt gtgatgaagt agatcatgca gtgacataca aagaccaagg attatgtata
    5161 tttttatatc tctgtggttt tgaaacttta gtacttagaa ttttggcctt ctgcactact
    5221 cttttgctct tacgaacata atggactctt aagaatggaa agggatgaca tttacctatg
    5281 tgtgctgcct cattcctggt gaagcaactg ctacttgttc tctatgcctc taaaatgatg
    5341 ctgttttctc tgctaaaggt aaaagaaaag aaaaaaatag ttggaaaata agacatgcaa
    5401 cttgatgtgc ttttgagtaa atttatgcag cagaaactat acaatgaagg aagaattcta
    5461 tggaaattac aaatccaaaa ctctatgatg atgtcttcct agggagtaga gaaaggcagt
    5521 gaaatggcag ttagaccaac agaggcttga aggattcaag tacaagtaat attttgtata
    5581 aaacatagca gtttaggtcc ccataatcct caaaaatagt cacaaatata acaaagttca
    5641 ttgttttagg gtttttaaaa aacgtgttgt acctaaggcc atacttactc ttctatgcta
    5701 tcactgcaaa ggggtgatat gtatgtatta tataaaaaaa aaaaccctta atgcactgtt
    5761 atctcctaaa tatttagtaa attaatacta tttaattttt ttaaagattt gtctgtgtag
    5821 acactaaaag tattacacaa aatctggact gaaggtgtcc tttttaacaa caatttaaag
    5881 tactttttat atatgttatg tagtatatcc tttctaaact gcctagtttg tatattccta
    5941 taattcctat ttgtgaagtg tacctgttct tgtctctttt ttcagtcatt ttctgcacgc
    6001 atcccccttt atatggttat agagatgact gtagcttttc gtgctccact gcgaggtttg
    6061 tgctcagagc cgctgcaccc cagcgaggcc tgctccatgg agtgcaggac gagctactgc
    6121 tttggagcga gggtttcctg cttttgagtt gacctgactt ccttcttgaa atgactgtta
    6181 aaactaaaat aaattacatt gcatttattt tatattcttg gttgaaataa aatttaattg
    6241 actttg
  • CDK19 Transcript Variant 2 (NM_001300960.1) (SEQ ID NO: 13)
  •    1 tgtggccgcc gaggagtccc ttgctgaagg cggaccgcgg agcggcgggc ggcgggcggc
      61 gcgcgcgcgc gcgcgagagg cggctgttgg agaagtggag cggcggtcgc ggggggagga
     121 ggaggaggga ctgagcggcg gcggcccccg cgtcccgtgc ctctatgggg gaagcagaca
     181 atggattatg atttcaaggc gaagctggcg gcggagcggg agcgggtgga ggatttgttt
     241 gagtacgaag ggtgcaaagt gggacgcggc acctacggtc acgtctacaa ggcgaggcgg
     301 aaagatggaa aagatgaaaa ggaatatgca ttgaagcaaa ttgaaggcac aggaatatcc
     361 atgtcggctt gtagagagat tgcacttttg cgagaattga agcaccctaa tgtgattgca
     421 ttgcagaagg tgttcctttc tcacagtgac aggaaggtat ggctgctgtt tgattatgca
     481 gagcatgact tgtggcatat tattaagttt caccgtgcat caaaagcaaa taaaaagccc
     541 atgcagttgc caagatctat ggttaaatcc ttactttacc agattcttga tggtatccat
     601 tacctccatg caaattgggt gcttcacaga gacttgaaac cagcaaatat cctagtaatg
     661 ggagaaggtc ctgagagggg gagagtcaaa atagatatat gggcaatagg ttgtatattt
     721 gctgaattgt tgacttcgga acctattttt cactgtcgtc aggaagatat aaaaacaagc
     781 aatccctttc atcatgatca actggatcgg atatttagtg tcatggggtt tcctgcagat
     841 aaagactggg aagatattag aaagatgcca gaatatccca cacttcaaaa agactttaga
     901 agaacaacgt atgccaacag tagcctcata aagtacatgg agaaacacaa ggtcaagcct
     961 gacagcaaag tgttcctctt gcttcagaaa ctcctgacca tggatccaac caagagaatt
    1021 acctcggagc aagctctgca ggatccctat tttcaggagg accctttgcc aacattagat
    1081 gtatttgccg gctgccagat tccatacccc aaacgagaat tccttaatga agatgatcct
    1141 gaagaaaaag gtgacaagaa tcagcaacag cagcagaacc agcatcagca gcccacagcc
    1201 cctccacagc aggcagcagc ccctccacag gcgcccccac cacagcagaa cagcacccag
    1261 accaacggga ccgcaggtgg ggctggggcc ggggtcgggg gcaccggagc agggttgcag
    1321 cacagccagg actccagcct gaaccaggtg cctccaaaca agaagccacg gctagggcct
    1381 tcaggcgcaa actcaggtgg acctgtgatg ccctcggatt atcagcactc cagttctcgc
    1441 ctgaattacc aaagcagcgt tcagggatcc tctcagtccc agagcacact tggctactct
    1501 tcctcgtctc agcagagctc acagtaccac ccatctcacc aggcccaccg gtactgacca
    1561 gctcccgttg ggccaggcca gcccagccca gagcacaggc tccagcaata tgtctgcatt
    1621 gaaaagaacc aaaaaaatgc aaactatgat gccatttaaa actcatacac atgggaggaa
    1681 aaccttatat actgagcatt gtgcaggact gatagctctt ctttattgac ttaaagaaga
    1741 ttcttgtgaa gtttccccag caccccttcc ctgcatgtgt tccattgtga cttctctgat
    1801 aaagcgtctg atctaatccc agcacttctg taaccttcag catttctttg aaggatttcc
    1861 tggtgcacct ttctcatgct gtagcaatca ctatggttta tcttttcaaa gctcttttaa
    1921 taggatttta atgttttaga aacaggattc cagtggtgta tagttttata cttcatgaac
    1981 tgatttagca acacaggtaa aaatgcacct tttaaagcac tacgttttca cagacaataa
    2041 ctgttctgct catggaagtc ttaaacagaa actgttactg tcccaaagta ctttactatt
    2101 acgttcgtat ttatctagtt tcagggaagg tctaataaaa agacaagcgg tgggacagag
    2161 ggaacctaca accaaaaact gcctagatct ttgcagttat gtgctttatg ccacgaagaa
    2221 ctgaagtatg tggtaatttt tatagaatca ttcatatgga actgagttcc cagcatcatc
    2281 ttattctgaa tagcattcag taattaagaa ttacaatttt aaccttcatg tagctaagtc
    2341 taccttaaaa agggtttcaa gagctttgta cagtctcgat ggcccacacc aaaacgctga
    2401 agagagtaac aactgcacta ggatttctgt aaggagtaat tttgatcaaa agacgtgtta
    2461 cttccctttg aaggaaaagt ttttagtgtg tattgtacat aaagtcggct tctctaaaga
    2521 accattggtt tcttcacatc tgggtctgcg tgagtaactt tcttgcataa tcaaggttac
    2581 tcaagtagaa gcctgaaaat taatctgctt ttaaaataaa gagcagtgtt ctccattcgt
    2641 atttgtatta gatatagagt gactattttt aaagcatgtt aaaaatttag gttttattca
    2701 tgtttaaagt atgtattatg tatgcataat tttgctgttg ttactgaaac ttaattctat
    2761 caagaatctt tttcattgca ctgaatgatt tcttttgccc ctaggagaaa acttaataat
    2821 tgtgcctaaa aactatgggc ggatagtata agactatact agacaaagtg aatatttgca
    2881 tttccattat ctatgaatta gtggctgagt tctttcttag ctgctttaag gagcccctca
    2941 ctccccagag tcaaaaggaa atgtaaaaac ttagagctcc cattgtaatg taaggggcaa
    3001 gaaatttgtg ttcttctgaa tgctactagc agcaccagcc ttgttttaaa tgttttcttg
    3061 agctagaaga aatagctgat tattgtatat gcaaattaca tgcattttta aaaactattc
    3121 tttctgaact tatctacctg gttatgatac tgtgggtcca tacacaagta aaataagatt
    3181 agacagaagc cagtatacat tttgcactat tgatgtgata ctgtagccag ccaggacctt
    3241 actgatctca gcataataat gctcactaat aatgaagtct gcatagtgac actcatcaag
    3301 actgaagatg aagcaggtta cgtgctccat tggaaggagt ttctgatagt ctcctgctgt
    3361 tttacccctt ccatttttta aaataagaaa ttagcagccc tctgcataat gtagctgcct
    3421 atatgcagtt ttatcctgtg ccctaaagcc tcactgtcca gagctgttgg tcatcagatg
    3481 cttattgcac cctcaccatg tgcctggtgc cctgctgggt agagaacaca gaggacaggg
    3541 catacttctt gtccttaagg agcttgtgat ctgtgacagt aagccctcct gggatgtctg
    3601 tgccatgtga ttgacttaca agtgaaactg tcttataata tgaaggtctt tttgtttact
    3661 tctaaaccca cttgggtagt tactatcccc aaatctgttc tgtaaataat attatggaag
    3721 ggtttctatg tcagtctacc ttagagaaag ccagtgattc aatatcacaa aaggcattga
    3781 cgtatctttg aaatgttcac agcagccttt taacaacaac tgggtggtcc ttgtaggcag
    3841 aacatactct cctaagtggt tgtaggaaat tgcaaggaaa atagaaggtc tgttcttgct
    3901 ctcaaggagg ttacctttaa taaaagaaga caaacccaga tagatatgta aaccaaaata
    3961 ctatgcccct taatacttta taagcagcat tgttaaatag ttcttacgct tatacattca
    4021 cagaactacc ctgttttcct tgtatataat gacttttgct ggcagaactg aaatataaac
    4081 tgtaagggga tttcgtcagt tgctcccagt atacaatatc ctccaggaca tagccagaaa
    4141 tctccattcc acacatgact gagttcctat ccctgcactg gtactggctc ttttctcctc
    4201 tttccttgcc tcagggttcg tgctacccac tgattccctt tacccttagt aataattttg
    4261 gatcattttc tttcctttaa aggggaacaa agcctttttt ttttttgaga cggagtgttg
    4321 ctctgtcacc caagctggag tgcagtggca cgatcttggc tcactccaac ctccaccttc
    4381 caggttcaag tgattctcct gcctcagcct cccgagtagc tgggactacg ggcacgcacc
    4441 accacgtctg gctaattttt gtatttttag tagagatggg gtttcaccct attggtcagg
    4501 ctggtcttga attcctcacc tcaggtcatc cgcctgtctc ggcctcccga agtgctggga
    4561 ttataggtgt gagccaccgc acccagttgg gaacaaagcc tttttaacac acgtaagggc
    4621 cctcaaaccg tgggacctct aaggagacct ttgaagcttt ttgagggcaa actttacctt
    4681 tgtggtcccc aaatgatggc atttctcttt gaaatttatt agatactgtt atgtccccca
    4741 agggtacagg aggggcatcc ctcagcctat gggaacaccc aaactaggag gggttattga
    4801 caggaaggaa tgaatccaag tgaaggcttt ctgctcttcg tgttacaaac cagtttcaga
    4861 gttagctttc tggggaggtg tgtgtttgtg aaaggaattc aagtgttgca ggacagatga
    4921 gctcaaggta aggtagcttt ggcagcaggg ctgatactat gaggctgaaa caatccttgt
    4981 gatgaagtag atcatgcagt gacatacaaa gaccaaggat tatgtatatt tttatatctc
    5041 tgtggttttg aaactttagt acttagaatt ttggccttct gcactactct tttgctctta
    5101 cgaacataat ggactcttaa gaatggaaag ggatgacatt tacctatgtg tgctgcctca
    5161 ttcctggtga agcaactgct acttgttctc tatgcctcta aaatgatgct gttttctctg
    5221 ctaaaggtaa aagaaaagaa aaaaatagtt ggaaaataag acatgcaact tgatgtgctt
    5281 ttgagtaaat ttatgcagca gaaactatac aatgaaggaa gaattctatg gaaattacaa
    5341 atccaaaact ctatgatgat gtcttcctag ggagtagaga aaggcagtga aatggcagtt
    5401 agaccaacag aggcttgaag gattcaagta caagtaatat tttgtataaa acatagcagt
    5461 ttaggtcccc ataatcctca aaaatagtca caaatataac aaagttcatt gttttagggt
    5521 ttttaaaaaa cgtgttgtac ctaaggccat acttactctt ctatgctatc actgcaaagg
    5581 ggtgatatgt atgtattata taaaaaaaaa aacccttaat gcactgttat ctcctaaata
    5641 tttagtaaat taatactatt taattttttt aaagatttgt ctgtgtagac actaaaagta
    5701 ttacacaaaa tctggactga aggtgtcctt tttaacaaca atttaaagta ctttttatat
    5761 atgttatgta gtatatcctt tctaaactgc ctagtttgta tattcctata attcctattt
    5821 gtgaagtgta cctgttcttg tctctttttt cagtcatttt ctgcacgcat ccccctttat
    5881 atggttatag agatgactgt agcttttcgt gctccactgc gaggtttgtg ctcagagccg
    5941 ctgcacccca gcgaggcctg ctccatggag tgcaggacga gctactgctt tggagcgagg
    6001 gtttcctgct tttgagttga cctgacttcc ttcttgaaat gactgttaaa actaaaataa
    6061 attacattgc atttatttta tattcttggt tgaaataaaa tttaattgac tttg
  • CDK19 Transcript Variant 3 (NM_001300963.1) (SEQ ID NO: 14)
  •   1 gaggggcggc cctggtacgc aggcgcgcat gctttgtggg ggcgaggctg tggtggcccg
      61 agattccagg agggcttcgt gtatggacct caagcgttgg aggtagcaga cttttcagca
     121 gaagaaaaga tgaaaaggaa tatgcattga agcaaattga aggcacagga atatccatgt
     181 cggcttgtag agagattgca cttttgcgag aattgaagca ccctaatgtg attgcattgc
     241 agaaggtgtt cctttctcac agtgacagga aggtatggct gctgtttgat tatgcagagc
     301 atgacttgtg gcatattatt aagtttcacc gtgcatcaaa agcaaataaa aagcccatgc
     361 agttgccaag atctatggtt aaatccttac tttaccagat tcttgatggt atccattacc
     421 tccatgcaaa ttgggtgctt cacagagact tgaaaccagc aaatatccta gtaatgggag
     481 aaggtcctga gagggggaga gtcaaaatag ctgacatggg ttttgccaga ttattcaatt
     541 ctcctctaaa gccactagca gatttggatc cagtagttgt gacattttgg tatcgggctc
     601 cagaactttt gcttggtgca aggcattata caaaggccat tgatatatgg gcaataggtt
     661 gtatatttgc tgaattgttg acttcggaac ctatttttca ctgtcgtcag gaagatataa
     721 aaacaagcaa tccctttcat catgatcaac tggatcggat atttagtgtc atggggtttc
     781 ctgcagataa agactgggaa gatattagaa agatgccaga atatcccaca cttcaaaaag
     841 actttagaag aacaacgtat gccaacagta gcctcataaa gtacatggag aaacacaagg
     901 tcaagcctga cagcaaagtg ttcctcttgc ttcagaaact cctgaccatg gatccaacca
     961 agagaattac ctcggagcaa gctctgcagg atccctattt tcaggaggac cctttgccaa
    1021 cattagatgt atttgccggc tgccagattc cataccccaa acgagaattc cttaatgaag
    1081 atgatcctga agaaaaaggt gacaagaatc agcaacagca gcagaaccag catcagcagc
    1141 ccacagcccc tccacagcag gcagcagccc ctccacaggc gcccccacca cagcagaaca
    1201 gcacccagac caacgggacc gcaggtgggg ctggggccgg ggtcgggggc accggagcag
    1261 ggttgcagca cagccaggac tccagcctga accaggtgcc tccaaacaag aagccacggc
    1321 tagggccttc aggcgcaaac tcaggtggac ctgtgatgcc ctcggattat cagcactcca
    1381 gttctcgcct gaattaccaa agcagcgttc agggatcctc tcagtcccag agcacacttg
    1441 gctactcttc ctcgtctcag cagagctcac agtaccaccc atctcaccag gcccaccggt
    1501 actgaccagc tcccgttggg ccaggccagc ccagcccaga gcacaggctc cagcaatatg
    1561 tctgcattga aaagaaccaa aaaaatgcaa actatgatgc catttaaaac tcatacacat
    1621 gggaggaaaa ccttatatac tgagcattgt gcaggactga tagctcttct ttattgactt
    1681 aaagaagatt cttgtgaagt ttccccagca ccccttccct gcatgtgttc cattgtgact
    1741 tctctgataa agcgtctgat ctaatcccag cacttctgta accttcagca tttctttgaa
    1801 ggatttcctg gtgcaccttt ctcatgctgt agcaatcact atggtttatc ttttcaaagc
    1861 tcttttaata ggattttaat gttttagaaa caggattcca gtggtgtata gttttatact
    1921 tcatgaactg atttagcaac acaggtaaaa atgcaccttt taaagcacta cgttttcaca
    1981 gacaataact gttctgctca tggaagtctt aaacagaaac tgttactgtc ccaaagtact
    2041 ttactattac gttcgtattt atctagtttc agggaaggtc taataaaaag acaagcggtg
    2101 ggacagaggg aacctacaac caaaaactgc ctagatcttt gcagttatgt gctttatgcc
    2161 acgaagaact gaagtatgtg gtaattttta tagaatcatt catatggaac tgagttccca
    2221 gcatcatctt attctgaata gcattcagta attaagaatt acaattttaa ccttcatgta
    2281 gctaagtcta ccttaaaaag ggtttcaaga gctttgtaca gtctcgatgg cccacaccaa
    2341 aacgctgaag agagtaacaa ctgcactagg atttctgtaa ggagtaattt tgatcaaaag
    2401 acgtgttact tccctttgaa ggaaaagttt ttagtgtgta ttgtacataa agtcggcttc
    2461 tctaaagaac cattggtttc ttcacatctg ggtctgcgtg agtaactttc ttgcataatc
    2521 aaggttactc aagtagaagc ctgaaaatta atctgctttt aaaataaaga gcagtgttct
    2581 ccattcgtat ttgtattaga tatagagtga ctatttttaa agcatgttaa aaatttaggt
    2641 tttattcatg tttaaagtat gtattatgta tgcataattt tgctgttgtt actgaaactt
    2701 aattctatca agaatctttt tcattgcact gaatgatttc ttttgcccct aggagaaaac
    2761 ttaataattg tgcctaaaaa ctatgggcgg atagtataag actatactag acaaagtgaa
    2821 tatttgcatt tccattatct atgaattagt ggctgagttc tttcttagct gctttaagga
    2881 gcccctcact ccccagagtc aaaaggaaat gtaaaaactt agagctccca ttgtaatgta
    2941 aggggcaaga aatttgtgtt cttctgaatg ctactagcag caccagcctt gttttaaatg
    3001 ttttcttgag ctagaagaaa tagctgatta ttgtatatgc aaattacatg catttttaaa
    3061 aactattctt tctgaactta tctacctggt tatgatactg tgggtccata cacaagtaaa
    3121 ataagattag acagaagcca gtatacattt tgcactattg atgtgatact gtagccagcc
    3181 aggaccttac tgatctcagc ataataatgc tcactaataa tgaagtctgc atagtgacac
    3241 tcatcaagac tgaagatgaa gcaggttacg tgctccattg gaaggagttt ctgatagtct
    3301 cctgctgttt taccccttcc attttttaaa ataagaaatt agcagccctc tgcataatgt
    3361 agctgcctat atgcagtttt atcctgtgcc ctaaagcctc actgtccaga gctgttggtc
    3421 atcagatgct tattgcaccc tcaccatgtg cctggtgccc tgctgggtag agaacacaga
    3481 ggacagggca tacttcttgt ccttaaggag cttgtgatct gtgacagtaa gccctcctgg
    3541 gatgtctgtg ccatgtgatt gacttacaag tgaaactgtc ttataatatg aaggtctttt
    3601 tgtttacttc taaacccact tgggtagtta ctatccccaa atctgttctg taaataatat
    3661 tatggaaggg tttctatgtc agtctacctt agagaaagcc agtgattcaa tatcacaaaa
    3721 ggcattgacg tatctttgaa atgttcacag cagcctttta acaacaactg ggtggtcctt
    3781 gtaggcagaa catactctcc taagtggttg taggaaattg caaggaaaat agaaggtctg
    3841 ttcttgctct caaggaggtt acctttaata aaagaagaca aacccagata gatatgtaaa
    3901 ccaaaatact atgcccctta atactttata agcagcattg ttaaatagtt cttacgctta
    3961 tacattcaca gaactaccct gttttccttg tatataatga cttttgctgg cagaactgaa
    4021 atataaactg taaggggatt tcgtcagttg ctcccagtat acaatatcct ccaggacata
    4081 gccagaaatc tccattccac acatgactga gttcctatcc ctgcactggt actggctctt
    4141 ttctcctctt tccttgcctc agggttcgtg ctacccactg attcccttta cccttagtaa
    4201 taattttgga tcattttctt tcctttaaag gggaacaaag cctttttttt ttttgagacg
    4261 gagtgttgct ctgtcaccca agctggagtg cagtggcacg atcttggctc actccaacct
    4321 ccaccttcca ggttcaagtg attctcctgc ctcagcctcc cgagtagctg ggactacggg
    4381 cacgcaccac cacgtctggc taatttttgt atttttagta gagatggggt ttcaccctat
    4441 tggtcaggct ggtcttgaat tcctcacctc aggtcatccg cctgtctcgg cctcccgaag
    4501 tgctgggatt ataggtgtga gccaccgcac ccagttggga acaaagcctt tttaacacac
    4561 gtaagggccc tcaaaccgtg ggacctctaa ggagaccttt gaagcttttt gagggcaaac
    4621 tttacctttg tggtccccaa atgatggcat ttctctttga aatttattag atactgttat
    4681 gtcccccaag ggtacaggag gggcatccct cagcctatgg gaacacccaa actaggaggg
    4741 gttattgaca ggaaggaatg aatccaagtg aaggctttct gctcttcgtg ttacaaacca
    4801 gtttcagagt tagctttctg gggaggtgtg tgtttgtgaa aggaattcaa gtgttgcagg
    4861 acagatgagc tcaaggtaag gtagctttgg cagcagggct gatactatga ggctgaaaca
    4921 atccttgtga tgaagtagat catgcagtga catacaaaga ccaaggatta tgtatatttt
    4981 tatatctctg tggttttgaa actttagtac ttagaatttt ggccttctgc actactcttt
    5041 tgctcttacg aacataatgg actcttaaga atggaaaggg atgacattta cctatgtgtg
    5101 ctgcctcatt cctggtgaag caactgctac ttgttctcta tgcctctaaa atgatgctgt
    5161 tttctctgct aaaggtaaaa gaaaagaaaa aaatagttgg aaaataagac atgcaacttg
    5221 atgtgctttt gagtaaattt atgcagcaga aactatacaa tgaaggaaga attctatgga
    5281 aattacaaat ccaaaactct atgatgatgt cttcctaggg agtagagaaa ggcagtgaaa
    5341 tggcagttag accaacagag gcttgaagga ttcaagtaca agtaatattt tgtataaaac
    5401 atagcagttt aggtccccat aatcctcaaa aatagtcaca aatataacaa agttcattgt
    5461 tttagggttt ttaaaaaacg tgttgtacct aaggccatac ttactcttct atgctatcac
    5521 tgcaaagggg tgatatgtat gtattatata aaaaaaaaaa cccttaatgc actgttatct
    5581 cctaaatatt tagtaaatta atactattta atttttttaa agatttgtct gtgtagacac
    5641 taaaagtatt acacaaaatc tggactgaag gtgtcctttt taacaacaat ttaaagtact
    5701 ttttatatat gttatgtagt atatcctttc taaactgcct agtttgtata ttcctataat
    5761 tcctatttgt gaagtgtacc tgttcttgtc tcttttttca gtcattttct gcacgcatcc
    5821 ccctttatat ggttatagag atgactgtag cttttcgtgc tccactgcga ggtttgtgct
    5881 cagagccgct gcaccccagc gaggcctgct ccatggagtg caggacgagc tactgctttg
    5941 gagcgagggt ttcctgcttt tgagttgacc tgacttcctt cttgaaatga ctgttaaaac
    6001 taaaataaat tacattgcat ttattttata ttcttggttg aaataaaatt taattgactt
    6061 tg
  • CDK19 Transcript Variant 4 (NM_001300964.1) (SEQ ID NO: 15)
  •    1 agaaaagaaa caagctgcgg tacaactgtc ctcaccagcc ctcgcctccc gagtcactgc
      61 agccaaccct tcagcaagaa aagatgaaaa ggaatatgca ttgaagcaaa ttgaaggcac
     121 aggaatatcc atgtcggctt gtagagagat tgcacttttg cgagaattga agcaccctaa
     181 tgtgattgca ttgcagaagg tgttcctttc tcacagtgac aggaaggtat ggctgctgtt
     241 tgattatgca gagcatgact tgtggcatat tattaagttt caccgtgcat caaaagcaaa
     301 taaaaagccc atgcagttgc caagatctat ggttaaatcc ttactttacc agattcttga
     361 tggtatccat tacctccatg caaattgggt gcttcacaga gacttgaaac cagcaaatat
     421 cctagtaatg ggagaaggtc ctgagagggg gagagtcaaa atagctgaca tgggttttgc
     481 cagattattc aattctcctc taaagccact agcagatttg gatccagtag ttgtgacatt
     541 ttggtatcgg gctccagaac ttttgcttgg tgcaaggcat tatacaaagg ccattgatat
     601 atgggcaata ggttgtatat ttgctgaatt gttgacttcg gaacctattt ttcactgtcg
     661 tcaggaagat ataaaaacaa gcaatccctt tcatcatgat caactggatc ggatatttag
     721 tgtcatgggg tttcctgcag ataaagactg ggaagatatt agaaagatgc cagaatatcc
     781 cacacttcaa aaagacttta gaagaacaac gtatgccaac agtagcctca taaagtacat
     841 ggagaaacac aaggtcaagc ctgacagcaa agtgttcctc ttgcttcaga aactcctgac
     901 catggatcca accaagagaa ttacctcgga gcaagctctg caggatccct attttcagga
     961 ggaccctttg ccaacattag atgtatttgc cggctgccag attccatacc ccaaacgaga
    1021 attccttaat gaagatgatc ctgaagaaaa aggtgacaag aatcagcaac agcagcagaa
    1081 ccagcatcag cagcccacag cccctccaca gcaggcagca gcccctccac aggcgccccc
    1141 accacagcag aacagcaccc agaccaacgg gaccgcaggt ggggctgggg ccggggtcgg
    1201 gggcaccgga gcagggttgc agcacagcca ggactccagc ctgaaccagg tgcctccaaa
    1261 caagaagcca cggctagggc cttcaggcgc aaactcaggt ggacctgtga tgccctcgga
    1321 ttatcagcac tccagttctc gcctgaatta ccaaagcagc gttcagggat cctctcagtc
    1381 ccagagcaca cttggctact cttcctcgtc tcagcagagc tcacagtacc acccatctca
    1441 ccaggcccac cggtactgac cagctcccgt tgggccaggc cagcccagcc cagagcacag
    1501 gctccagcaa tatgtctgca ttgaaaagaa ccaaaaaaat gcaaactatg atgccattta
    1561 aaactcatac acatgggagg aaaaccttat atactgagca ttgtgcagga ctgatagctc
    1621 ttctttattg acttaaagaa gattcttgtg aagtttcccc agcacccctt ccctgcatgt
    1681 gttccattgt gacttctctg ataaagcgtc tgatctaatc ccagcacttc tgtaaccttc
    1741 agcatttctt tgaaggattt cctggtgcac ctttctcatg ctgtagcaat cactatggtt
    1801 tatcttttca aagctctttt aataggattt taatgtttta gaaacaggat tccagtggtg
    1861 tatagtttta tacttcatga actgatttag caacacaggt aaaaatgcac cttttaaagc
    1921 actacgtttt cacagacaat aactgttctg ctcatggaag tcttaaacag aaactgttac
    1981 tgtcccaaag tactttacta ttacgttcgt atttatctag tttcagggaa ggtctaataa
    2041 aaagacaagc ggtgggacag agggaaccta caaccaaaaa ctgcctagat ctttgcagtt
    2101 atgtgcttta tgccacgaag aactgaagta tgtggtaatt tttatagaat cattcatatg
    2161 gaactgagtt cccagcatca tcttattctg aatagcattc agtaattaag aattacaatt
    2221 ttaaccttca tgtagctaag tctaccttaa aaagggtttc aagagctttg tacagtctcg
    2281 atggcccaca ccaaaacgct gaagagagta acaactgcac taggatttct gtaaggagta
    2341 attttgatca aaagacgtgt tacttccctt tgaaggaaaa gtttttagtg tgtattgtac
    2401 ataaagtcgg cttctctaaa gaaccattgg tttcttcaca tctgggtctg cgtgagtaac
    2461 tttcttgcat aatcaaggtt actcaagtag aagcctgaaa attaatctgc ttttaaaata
    2521 aagagcagtg ttctccattc gtatttgtat tagatataga gtgactattt ttaaagcatg
    2581 ttaaaaattt aggttttatt catgtttaaa gtatgtatta tgtatgcata attttgctgt
    2641 tgttactgaa acttaattct atcaagaatc tttttcattg cactgaatga tttcttttgc
    2701 ccctaggaga aaacttaata attgtgccta aaaactatgg gcggatagta taagactata
    2761 ctagacaaag tgaatatttg catttccatt atctatgaat tagtggctga gttctttctt
    2821 agctgcttta aggagcccct cactccccag agtcaaaagg aaatgtaaaa acttagagct
    2881 cccattgtaa tgtaaggggc aagaaatttg tgttcttctg aatgctacta gcagcaccag
    2941 ccttgtttta aatgttttct tgagctagaa gaaatagctg attattgtat atgcaaatta
    3001 catgcatttt taaaaactat tctttctgaa cttatctacc tggttatgat actgtgggtc
    3061 catacacaag taaaataaga ttagacagaa gccagtatac attttgcact attgatgtga
    3121 tactgtagcc agccaggacc ttactgatct cagcataata atgctcacta ataatgaagt
    3181 ctgcatagtg acactcatca agactgaaga tgaagcaggt tacgtgctcc attggaagga
    3241 gtttctgata gtctcctgct gttttacccc ttccattttt taaaataaga aattagcagc
    3301 cctctgcata atgtagctgc ctatatgcag ttttatcctg tgccctaaag cctcactgtc
    3361 cagagctgtt ggtcatcaga tgcttattgc accctcacca tgtgcctggt gccctgctgg
    3421 gtagagaaca cagaggacag ggcatacttc ttgtccttaa ggagcttgtg atctgtgaca
    3481 gtaagccctc ctgggatgtc tgtgccatgt gattgactta caagtgaaac tgtcttataa
    3541 tatgaaggtc tttttgttta cttctaaacc cacttgggta gttactatcc ccaaatctgt
    3601 tctgtaaata atattatgga agggtttcta tgtcagtcta ccttagagaa agccagtgat
    3661 tcaatatcac aaaaggcatt gacgtatctt tgaaatgttc acagcagcct tttaacaaca
    3721 actgggtggt ccttgtaggc agaacatact ctcctaagtg gttgtaggaa attgcaagga
    3781 aaatagaagg tctgttcttg ctctcaagga ggttaccttt aataaaagaa gacaaaccca
    3841 gatagatatg taaaccaaaa tactatgccc cttaatactt tataagcagc attgttaaat
    3901 agttcttacg cttatacatt cacagaacta ccctgttttc cttgtatata atgacttttg
    3961 ctggcagaac tgaaatataa actgtaaggg gatttcgtca gttgctccca gtatacaata
    4021 tcctccagga catagccaga aatctccatt ccacacatga ctgagttcct atccctgcac
    4081 tggtactggc tcttttctcc tctttccttg cctcagggtt cgtgctaccc actgattccc
    4141 tttaccctta gtaataattt tggatcattt tctttccttt aaaggggaac aaagcctttt
    4201 ttttttttga gacggagtgt tgctctgtca cccaagctgg agtgcagtgg cacgatcttg
    4261 gctcactcca acctccacct tccaggttca agtgattctc ctgcctcagc ctcccgagta
    4321 gctgggacta cgggcacgca ccaccacgtc tggctaattt ttgtattttt agtagagatg
    4381 gggtttcacc ctattggtca ggctggtctt gaattcctca cctcaggtca tccgcctgtc
    4441 tcggcctccc gaagtgctgg gattataggt gtgagccacc gcacccagtt gggaacaaag
    4501 cctttttaac acacgtaagg gccctcaaac cgtgggacct ctaaggagac ctttgaagct
    4561 ttttgagggc aaactttacc tttgtggtcc ccaaatgatg gcatttctct ttgaaattta
    4621 ttagatactg ttatgtcccc caagggtaca ggaggggcat ccctcagcct atgggaacac
    4681 ccaaactagg aggggttatt gacaggaagg aatgaatcca agtgaaggct ttctgctctt
    4741 cgtgttacaa accagtttca gagttagctt tctggggagg tgtgtgtttg tgaaaggaat
    4801 tcaagtgttg caggacagat gagctcaagg taaggtagct ttggcagcag ggctgatact
    4861 atgaggctga aacaatcctt gtgatgaagt agatcatgca gtgacataca aagaccaagg
    4921 attatgtata tttttatatc tctgtggttt tgaaacttta gtacttagaa ttttggcctt
    4981 ctgcactact cttttgctct tacgaacata atggactctt aagaatggaa agggatgaca
    5041 tttacctatg tgtgctgcct cattcctggt gaagcaactg ctacttgttc tctatgcctc
    5101 taaaatgatg ctgttttctc tgctaaaggt aaaagaaaag aaaaaaatag ttggaaaata
    5161 agacatgcaa cttgatgtgc ttttgagtaa atttatgcag cagaaactat acaatgaagg
    5221 aagaattcta tggaaattac aaatccaaaa ctctatgatg atgtcttcct agggagtaga
    5281 gaaaggcagt gaaatggcag ttagaccaac agaggcttga aggattcaag tacaagtaat
    5341 attttgtata aaacatagca gtttaggtcc ccataatcct caaaaatagt cacaaatata
    5401 acaaagttca ttgttttagg gtttttaaaa aacgtgttgt acctaaggcc atacttactc
    5461 ttctatgcta tcactgcaaa ggggtgatat gtatgtatta tataaaaaaa aaaaccctta
    5521 atgcactgtt atctcctaaa tatttagtaa attaatacta tttaattttt ttaaagattt
    5581 gtctgtgtag acactaaaag tattacacaa aatctggact gaaggtgtcc tttttaacaa
    5641 caatttaaag tactttttat atatgttatg tagtatatcc tttctaaact gcctagtttg
    5701 tatattccta taattcctat ttgtgaagtg tacctgttct tgtctctttt ttcagtcatt
    5761 ttctgcacgc atcccccttt atatggttat agagatgact gtagcttttc gtgctccact
    5821 gcgaggtttg tgctcagagc cgctgcaccc cagcgaggcc tgctccatgg agtgcaggac
    5881 gagctactgc tttggagcga gggtttcctg cttttgagtt gacctgactt ccttcttgaa
    5941 atgactgtta aaactaaaat aaattacatt gcatttattt tatattcttg gttgaaataa
    6001 aatttaattg actttg
  • Cyclin dependent kinase 8 (CDK8), transcript variant 1 (NM_001260.2) (SEQ ID NO: 16)
  •    1 gagtgccctc cctcctcctc tctttgagga ggtaccggct gttgtgcggc tctgcccttc
      61 tgtttgagtg tatgggagag tgagtgagtg agtgagtgtg agcgtgtgtg tgagagcgtg
     121 aggcgtgagt gcgcgtgtga gaggacgaga gcccgcctgg ccgccccgcc gctcccgccg
     181 cagcaggagc agaacgcgcg gccggagaga gcggcggagc cggcgcccag ggagcccgcg
     241 gggacaaggg cagagacacc gctccccacc cccagccctc gtccctcggc tctccttcgc
     301 cgggggatcc tccccgttcc tccacccccg gccggcctct gccccgccgt ccccctggat
     361 gtccctggcg ctttcgcggg gcctcctcct gctcttgccg catcagtcgg gctggtgctg
     421 cggccggcgg gcgtagagcg ggcgggttcc cgggggctgc ggctgcccgt gcttccccgg
     481 tccccacccc tgccccccgg ccccccgacc cagctctccg gcctcagagg ctgtgacaat
     541 ggactatgac tttaaagtga agctgagcag cgagcgggag cgggtcgagg acctgtttga
     601 atacgagggc tgcaaagttg gccgaggcac ttatggtcac gtctacaaag ccaagaggaa
     661 agatgggaag gatgataaag actatgcttt aaaacaaata gaaggaactg ggatctctat
     721 gtcggcatgt agagaaatag cattacttcg agagcttaag catccaaacg tcatttctct
     781 tcaaaaggtg tttctgtctc atgctgatag gaaggtgtgg cttctgtttg actatgctga
     841 acatgacctc tggcatataa tcaagtttca cagagcttct aaagcaaaca agaagccagt
     901 tcagttacct cggggaatgg tgaagtcact attatatcag atcctagatg gtattcacta
     961 cctgcatgct aactgggtgt tgcacagaga tttgaaacct gctaatattt tagttatggg
    1021 tgaaggtcct gagcgaggaa gagtaaaaat tgctgacatg ggctttgccc gattatttaa
    1081 ttcacctttg aagcctttag cagatttgga tccagtggtt gttacattct ggtaccgagc
    1141 ccctgaacta cttcttggag caaggcatta taccaaagct attgatattt gggctatagg
    1201 gtgtatattt gcagaactac taacgtcaga accaatattt cactgtcgac aagaggacat
    1261 caaaactagt aatccttatc accatgacca gctggacaga atattcaatg taatgggatt
    1321 tcctgcagat aaagattggg aagatataaa aaagatgcct gaacattcaa cattaatgaa
    1381 agatttcaga agaaatacgt ataccaactg cagccttatc aagtatatgg aaaaacataa
    1441 agttaaacca gatagtaaag cattccactt gcttcagaag ctgcttacca tggacccaat
    1501 aaagcgaatt acctcagaac aggctatgca ggacccctat ttcttagaag acccacttcc
    1561 tacatcagac gtttttgccg gttgtcaaat cccttaccca aaacgagaat ttttaacgga
    1621 agaagaacct gatgacaaag gagacaaaaa gaaccagcag cagcagcagg gcaataacca
    1681 cactaatgga actggccacc cagggaatca agacagcagt cacacacagg gacccccgtt
    1741 gaagaaagtg agagttgttc ctcctaccac tacctcaggt ggacttatca tgacctcaga
    1801 ctatcagcgt tccaatccac atgctgccta tcccaaccct ggaccaagca catcacagcc
    1861 gcagagcagc atgggatact cagctacctc ccagcagcct ccacagtact cacatcagac
    1921 acatcggtac tgagctgcat cggaatcttg tccatgcact gttgcgaatg ctgcagggct
    1981 gactgtgcag ctctctgcgg gaacctggta tgggccatga gaatgtactg tacaaccaca
    2041 tcttcaaaat gtccagtagc caagttccac cacttttcac agattggggt agtggcttcc
    2101 aagttgtacc tattttggag ttagacttga aaagaaagtg ctagcacagt ttgtgttgtg
    2161 gatttgctac ttccatagtt tacttgacat ggttcagact gaccaatgca tttttttcag
    2221 tgacagtctg tagcagttga agctgtgaat gtgctagggg caagcatttg tctttgtatg
    2281 tggtgaattt tttcagtgta acaacattat ctgaccaata gtacacacac agacacaaag
    2341 tttaactggt acttgaaaca tacagtatat gttaacgaaa taaccaagac tcgaaatgag
    2401 attattttgg tacacctttc tttttagtgt cttatcagtg ggctgattca ttttctacat
    2461 taatcagtgt tttctgacca agaatattgc ttggattttt ttgaaagtac aaaaagccac
    2521 atagtttttc cagaaaggtt tcaaaactcc caaagattaa cttccaactt ataagtttgt
    2581 ttttattttc aatctatgac ttgactggta ttaaagctgc tatttgatag taattaaata
    2641 tgttgtcatt gatataaacc tgtttggttc agcaaacaaa ctaaaatgat tgtcatagac
    2701 agtgttttat ttttcctgtt ggtgttgctg atttgtgagc atgctttaag atgaaaaaag
    2761 catgaatgat aacttcctta aaaaggtgcg gcatccaatt caaatatttt cgtcctgatt
    2821 ttaaagctgg ttggtgtagt gctattaaaa tttcgttcag ttaattttcc ttttgaaaac
    2881 ttgttcgcac gttgtttagg gtgcccttac ttcagcaaag gagaaggagt aggagagcct
    2941 tagaattttt gaggaaaaaa aaacctataa catacaatgt actgtatcaa actattttac
    3001 atgaatgaca caagtattct gaataaaaaa taattgaaca ttgttaaaaa caaggtgtta
    3061 tgtaataaat ttatttttca taaatcaaaa aaaaaaaaaa a
  • Cyclin dependent kinase 8 (CDK8), transcript variant 2 (NM_001318368.1) (SEQ ID NO: 17)
  •    1 gagtgccctc cctcctcctc tctttgagga ggtaccggct gttgtgcggc tctgcccttc
      61 tgtttgagtg tatgggagag tgagtgagtg agtgagtgtg agcgtgtgtg tgagagcgtg
     121 aggcgtgagt gcgcgtgtga gaggacgaga gcccgcctgg ccgccccgcc gctcccgccg
     181 cagcaggagc agaacgcgcg gccggagaga gcggcggagc cggcgcccag ggagcccgcg
     241 gggacaaggg cagagacacc gctccccacc cccagccctc gtccctcggc tctccttcgc
     301 cgggggatcc tccccgttcc tccacccccg gccggcctct gccccgccgt ccccctggat
     361 gtccctggcg ctttcgcggg gcctcctcct gctcttgccg catcagtcgg gctggtgctg
     421 cggccggcgg gcgtagagcg ggcgggttcc cgggggctgc ggctgcccgt gcttccccgg
     481 tccccacccc tgccccccgg ccccccgacc cagctctccg gcctcagagg ctgtgacaat
     541 ggactatgac tttaaagtga agctgagcag cgagcgggag cgggtcgagg acctgtttga
     601 atacgagggc tgcaaagttg gccgaggcac ttatggtcac gtctacaaag ccaagaggaa
     661 agatgggaag gatgataaag actatgcttt aaaacaaata gaaggaactg ggatctctat
     721 gtcggcatgt agagaaatag cattacttcg agagcttaag catccaaacg tcatttctct
     781 tcaaaaggtg tttctgtctc atgctgatag gaaggtgtgg cttctgtttg actatgctga
     841 acatgacctc tggcatataa tcaagtttca cagagcttct aaagcaaaca agaagccagt
     901 tcagttacct cggggaatgg tgaagtcact attatatcag atcctagatg gtattcacta
     961 cctgcatgct aactgggtgt tgcacagaga tttgaaacct gctaatattt tagttatggg
    1021 tgaaggtcct gagcgaggaa gagtaaaaat tgctgacatg ggctttgccc gattatttaa
    1081 ttcacctttg aagcctttag cagatttgga tccagtggtt gttacattct ggtaccgagc
    1141 ccctgaacta cttcttggag caaggcatta taccaaagct attgatattt gggctatagg
    1201 gtgtatattt gcagaactac taacgtcaga accaatattt cactgtcgac aagaggacat
    1261 caaaactagt aatccttatc accatgacca gctggacaga atattcaatg taatgggatt
    1321 tcctgcagat aaagattggg aagatataaa aaagatgcct gaacattcaa cattaatgaa
    1381 agatttcaga agaaatacgt ataccaactg cagccttatc aagtatatgg aaaaacataa
    1441 agttaaacca gatagtaaag cattccactt gcttcagaag ctgcttacca tggacccaat
    1501 aaagcgaatt acctcagaac aggctatgca ggacccctat ttcttagaag acccacttcc
    1561 tacatcagac gtttttgccg gttgtcaaat cccttaccca aaacgagaat ttttaacgga
    1621 agaagaacct gatgacaaag gagacaaaaa ccagcagcag cagcagggca ataaccacac
    1681 taatggaact ggccacccag ggaatcaaga cagcagtcac acacagggac ccccgttgaa
    1741 gaaagtgaga gttgttcctc ctaccactac ctcaggtgga cttatcatga cctcagacta
    1801 tcagcgttcc aatccacatg ctgcctatcc caaccctgga ccaagcacat cacagccgca
    1861 gagcagcatg ggatactcag ctacctccca gcagcctcca cagtactcac atcagacaca
    1921 tcggtactga gctgcatcgg aatcttgtcc atgcactgtt gcgaatgctg cagggctgac
    1981 tgtgcagctc tctgcgggaa cctggtatgg gccatgagaa tgtactgtac aaccacatct
    2041 tcaaaatgtc cagtagccaa gttccaccac ttttcacaga ttggggtagt ggcttccaag
    2101 ttgtacctat tttggagtta gacttgaaaa gaaagtgcta gcacagtttg tgttgtggat
    2161 ttgctacttc catagtttac ttgacatggt tcagactgac caatgcattt ttttcagtga
    2221 cagtctgtag cagttgaagc tgtgaatgtg ctaggggcaa gcatttgtct ttgtatgtgg
    2281 tgaatttttt cagtgtaaca acattatctg accaatagta cacacacaga cacaaagttt
    2341 aactggtact tgaaacatac agtatatgtt aacgaaataa ccaagactcg aaatgagatt
    2401 attttggtac acctttcttt ttagtgtctt atcagtgggc tgattcattt tctacattaa
    2461 tcagtgtttt ctgaccaaga atattgcttg gatttttttg aaagtacaaa aagccacata
    2521 gtttttccag aaaggtttca aaactcccaa agattaactt ccaacttata agtttgtttt
    2581 tattttcaat ctatgacttg actggtatta aagctgctat ttgatagtaa ttaaatatgt
    2641 tgtcattgat ataaacctgt ttggttcagc aaacaaacta aaatgattgt catagacagt
    2701 gttttatttt tcctgttggt gttgctgatt tgtgagcatg ctttaagatg aaaaaagcat
    2761 gaatgataac ttccttaaaa aggtgcggca tccaattcaa atattttcgt cctgatttta
    2821 aagctggttg gtgtagtgct attaaaattt cgttcagtta attttccttt tgaaaacttg
    2881 ttcgcacgtt gtttagggtg cccttacttc agcaaaggag aaggagtagg agagccttag
    2941 aatttttgag gaaaaaaaaa cctataacat acaatgtact gtatcaaact attttacatg
    3001 aatgacacaa gtattctgaa taaaaaataa ttgaacattg ttaaaaacaa ggtgttatgt
    3061 aataaattta tttttcataa atcaaaaaaa aaaaaaaa
  • Cyclin dependent kinase 8 (CDK8), transcript variant 3 (NM_001346501.1) (SEQ ID NO: 18)
  •    1 gagtgccctc cctcctcctc tctttgagga ggtaccggct gttgtgcggc tctgcccttc
      61 tgtttgagtg tatgggagag tgagtgagtg agtgagtgtg agcgtgtgtg tgagagcgtg
     121 aggcgtgagt gcgcgtgtga gaggacgaga gcccgcctgg ccgccccgcc gctcccgccg
     181 cagcaggagc agaacgcgcg gccggagaga gcggcggagc cggcgcccag ggagcccgcg
     241 gggacaaggg cagagacacc gctccccacc cccagccctc gtccctcggc tctccttcgc
     301 cgggggatcc tccccgttcc tccacccccg gccggcctct gccccgccgt ccccctggat
     361 gtccctggcg ctttcgcggg gcctcctcct gctcttgccg catcagtcgg gctggtgctg
     421 cggccggcgg gcgtagagcg ggcgggttcc cgggggctgc ggctgcccgt gcttccccgg
     481 tccccacccc tgccccccgg ccccccgacc cagctctccg gcctcagagg ctgtgacaat
     541 ggactatgac tttaaagtga agctgagcag cgagcgggag cgggtcgagg acctgtttga
     601 atacgagggc tgcaaagttg gccgaggcac ttatggtcac gtctacaaag ccaagaggaa
     661 agatgggaag gatgataaag actatgcttt aaaacaaata gaaggaactg ggatctctat
     721 gtcggcatgt agagaaatag cattacttcg agagcttaag catccaaacg tcatttctct
     781 tcaaaaggtg tttctgtctc atgctgatag gaaggtgtgg cttctgtttg actatgctga
     841 acatgacctc tggcatataa tcaagtttca cagagcttct aaagcaaaca agaagccagt
     901 tcagttacct cggggaatgg tgaagtcact attatatcag atcctagatg gtattcacta
     961 cctgcatgct aactgggtgt tgcacagaga tttgctgaca tgggctttgc ccgattattt
    1021 aattcacctt tgaagccttt agcagatttg gatccagtgg ttgttacatt ctggtaccga
    1081 gcccctgaac tacttcttgg agcaaggcat tataccaaag ctattgatat ttgggctata
    1141 gggtgtatat ttgcagaact actaacgtca gaaccaatat ttcactgtcg acaagaggac
    1201 atcaaaacta gtaatcctta tcaccatgac cagctggaca gaatattcaa tgtaatggga
    1261 tttcctgcag ataaagattg ggaagatata aaaaagatgc ctgaacattc aacattaatg
    1321 aaagatttca gaagaaatac gtataccaac tgcagcctta tcaagtatat ggaaaaacat
    1381 aaagttaaac cagatagtaa agcattccac ttgcttcaga agctgcttac catggaccca
    1441 ataaagcgaa ttacctcaga acaggctatg caggacccct atttcttaga agacccactt
    1501 cctacatcag acgtttttgc cggttgtcaa atcccttacc caaaacgaga atttttaacg
    1561 gaagaagaac ctgatgacaa aggagacaaa aagaaccagc agcagcagca gggcaataac
    1621 cacactaatg gaactggcca cccagggaat caagacagca gtcacacaca gggacccccg
    1681 ttgaagaaag tgagagttgt tcctcctacc actacctcag gtggacttat catgacctca
    1741 gactatcagc gttccaatcc acatgctgcc tatcccaacc ctggaccaag cacatcacag
    1801 ccgcagagca gcatgggata ctcagctacc tcccagcagc ctccacagta ctcacatcag
    1861 acacatcggt actgagctgc atcggaatct tgtccatgca ctgttgcgaa tgctgcaggg
    1921 ctgactgtgc agctctctgc gggaacctgg tatgggccat gagaatgtac tgtacaacca
    1981 catcttcaaa atgtccagta gccaagttcc accacttttc acagattggg gtagtggctt
    2041 ccaagttgta cctattttgg agttagactt gaaaagaaag tgctagcaca gtttgtgttg
    2101 tggatttgct acttccatag tttacttgac atggttcaga ctgaccaatg catttttttc
    2161 agtgacagtc tgtagcagtt gaagctgtga atgtgctagg ggcaagcatt tgtctttgta
    2221 tgtggtgaat tttttcagtg taacaacatt atctgaccaa tagtacacac acagacacaa
    2281 agtttaactg gtacttgaaa catacagtat atgttaacga aataaccaag actcgaaatg
    2341 agattatttt ggtacacctt tctttttagt gtcttatcag tgggctgatt cattttctac
    2401 attaatcagt gttttctgac caagaatatt gcttggattt ttttgaaagt acaaaaagcc
    2461 acatagtttt tccagaaagg tttcaaaact cccaaagatt aacttccaac ttataagttt
    2521 gtttttattt tcaatctatg acttgactgg tattaaagct gctatttgat agtaattaaa
    2581 tatgttgtca ttgatataaa cctgtttggt tcagcaaaca aactaaaatg attgtcatag
    2641 acagtgtttt atttttcctg ttggtgttgc tgatttgtga gcatgcttta agatgaaaaa
    2701 agcatgaatg ataacttcct taaaaaggtg cggcatccaa ttcaaatatt ttcgtcctga
    2761 ttttaaagct ggttggtgta gtgctattaa aatttcgttc agttaatttt ccttttgaaa
    2821 acttgttcgc acgttgttta gggtgccctt acttcagcaa aggagaagga gtaggagagc
    2881 cttagaattt ttgaggaaaa aaaaacctat aacatacaat gtactgtatc aaactatttt
    2941 acatgaatga cacaagtatt ctgaataaaa aataattgaa cattgttaaa aacaaggtgt
    3001 tatgtaataa atttattttt cataaatcaa aaaaaaaaaa aaa

Claims (22)

What is claimed is:
1. A method of treating a patient diagnosed with triple-negative breast cancer (TNBC), comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19), wherein the agent comprises a small molecule inhibitor of CDK19 activity, and wherein administration of the agent results in at least one of a reduction in cachexia, increase in survival time, elongation in time to tumor progression, reduction in tumor mass, reduction in tumor burden, prolongation in time to tumor metastasis, a prolongation in time to tumor recurrence, tumor response, complete response, partial response, stable disease, progressive disease, or progression free survival.
2. A method of treating a patient diagnosed with triple-negative breast cancer (TNBC), wherein the cancer is characterized by a tumor comprising EpCAMmed/high and CD10-/low epithelial cells, the method comprising administering a therapeutically effective dose of an agent that inhibits cyclin-dependent kinase 19 (CDK19) expression or activity, wherein the agent is a small molecule inhibitor of CDK19 activity, wherein the treatment reduces the number of EpCAMmed/high and CD10-/low cells in the tumor, reduces to number of EpCAMmed/high and CD10-/low cells per unit volume of the tumor, or results in a reduction of the ratio of EpCAMmed/high and CD10-/low epithelial cells to normal cells in the tumor.
3. A method of reducing metastasis of TNBC in a patient, the method comprising administering a therapeutically effective dose of an agent that inhibits expression or activity of CDK19, wherein the agent is a small molecule inhibitor of CDK19 activity.
4. The method of claim 1, wherein the patient is treated with a combination therapy comprising (a) an agent that inhibits expression or activity of CDK19 and (b) radiation therapy and/or chemotherapy.
5. The method of claim 1, comprising detecting EpCAMmed/high/CD10-/low cells in a tissue sample from the patient prior to or after initiating therapy.
6. (canceled)
7. The method of any of claim 1 wherein the agent inhibits expression or activity of CDK19 to a greater extent than it inhibits expression or activity of CDK8.
8. The method of claim 1 claim 1 wherein the agent is a small molecule inhibitor that binds to the ATP binding site of CDK19 to inhibit its activity.
9. The method of claim 1, wherein the agent binds to parts of CDK19 outside of the ATP binding site.
10. The method of claim 1, wherein the agent binds to CDK19 with a higher affinity than to CDK8.
11. The method of claim 1 wherein the agent is a small molecule inhibitor other than one or more compounds selected from the group consisting of Cortistatin A, Sorafenib, Linifanib, Ponatinib, Senexin B, CCT251545, and CCT251921.
12-15. (canceled)
16. The method of claim 1, wherein the agent binds CDK 19 in the cytoplasm of a breast epithelial cell.
17. A method of predicting the likely therapeutic responsiveness of a subject with TNBC to the method of treatment of claim 1 comprising:
(a) quantitating EpCAMmed/high/CD10-/low cells in a tumor sample obtained from the subject;
(b) comparing the quantity of EpCAMmed/high/CD10′/low cells in (a) to a reference value characteristic of tumors responsive to a CDK19 targeting therapy, and
(c) treating the patient with the agent that inhibits expression or activity of cyclin-dependent kinase 19 (CDK19) if the quantity of EpCAMmed/high/CD10-/low cells is equal to or exceeds the reference value.
18. The method of claim 2, wherein the agent inhibits expression or activity of CDK 19 to a greater extent than it inhibits expression or activity of CDK8.
19. The method of claim 2, wherein the agent is a small molecule inhibitor other than one or more compounds selected from the group consisting of Cortistatin A, Sorafenib, Linifanib, Ponatinib, Senexin B, CCT251545, and CCT251921.
20. The method of claim 3, comprising detecting EpCAMmed/high/CD10′/low cells in a tissue sample from the patient prior to the administering.
21. The method of claim 3, wherein the agent inhibits expression or activity of CDK 19 to a greater extent than it inhibits expression or activity of CDK8.
22. The method of claim 3, wherein the agent binds to CDK19 with a higher affinity than to CDK8.
23. The method of claim 3, wherein the agent is a small molecule inhibitor that binds to the ATP binding site of CDK19 to inhibit its activity.
24. The method of claim 3, wherein the agent binds to parts of CDK19 outside of the ATP binding site.
25. The method of claim 3, wherein the agent is a small molecule inhibitor other than one or more compounds selected from the group consisting of Cortistatin A, Sorafenib, Linifanib, Ponatinib, Senexin B, CCT251545, and CCT251921.
US17/823,028 2017-09-18 2022-08-29 Methods for treating triple-negative breast cancer Pending US20230285439A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/823,028 US20230285439A1 (en) 2017-09-18 2022-08-29 Methods for treating triple-negative breast cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762560140P 2017-09-18 2017-09-18
PCT/US2018/051489 WO2019055977A1 (en) 2017-09-18 2018-09-18 Methods for treating triple-negative breast cancer
US202016648088A 2020-03-17 2020-03-17
US17/823,028 US20230285439A1 (en) 2017-09-18 2022-08-29 Methods for treating triple-negative breast cancer

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2018/051489 Continuation WO2019055977A1 (en) 2017-09-18 2018-09-18 Methods for treating triple-negative breast cancer
US16/648,088 Continuation US11471477B2 (en) 2017-09-18 2018-09-18 Methods for treating triple-negative breast cancer

Publications (1)

Publication Number Publication Date
US20230285439A1 true US20230285439A1 (en) 2023-09-14

Family

ID=65723126

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/648,088 Active US11471477B2 (en) 2017-09-18 2018-09-18 Methods for treating triple-negative breast cancer
US17/823,028 Pending US20230285439A1 (en) 2017-09-18 2022-08-29 Methods for treating triple-negative breast cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/648,088 Active US11471477B2 (en) 2017-09-18 2018-09-18 Methods for treating triple-negative breast cancer

Country Status (7)

Country Link
US (2) US11471477B2 (en)
EP (1) EP3684420A4 (en)
JP (1) JP7392954B2 (en)
CN (1) CN111278469A (en)
AU (1) AU2018333072B2 (en)
CA (1) CA3075784A1 (en)
WO (1) WO2019055977A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023044041A1 (en) * 2021-09-16 2023-03-23 University Of South Carolina Cdk8/19 inhibitors for preventing drug resistance
CN114854755B (en) * 2022-05-30 2023-05-23 陕西师范大学 Lentiviral vector for inhibiting VSTM2L expression and application thereof
CN114832110B (en) * 2022-06-21 2023-04-28 中山大学孙逸仙纪念医院 Use of long-chain non-coding RNA CRCAL-3 inhibitor in preparation of product for treating radiotherapy resisting breast cancer
CN116773815B (en) * 2023-08-14 2023-11-10 四川省医学科学院·四川省人民医院 Application of SLC2A6 as treatment target and screening target and breast cancer screening kit
CN117144008B (en) * 2023-08-31 2024-03-08 武汉科技大学 Triple negative breast cancer biomarker and application thereof

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4704692A (en) 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US20080293142A1 (en) 2007-04-19 2008-11-27 The Board Of Regents For Oklahoma State University Multiple shRNA Expression Vectors and Methods of Construction
PT2419144T (en) 2009-04-17 2019-09-16 Univ Oxford Innovation Ltd Composition for delivery of genetic material
US8598344B2 (en) 2009-11-30 2013-12-03 Senex Biotechnology CDKI pathway inhibitors and uses thereof
SG11201401343YA (en) * 2011-09-13 2014-08-28 Igor Roninson TREATMENT OF DISEASE OR DISORDERS CAUSED BY INDUCED NFkB TRANSCRIPTIONAL ACTIVITY
KR20220045091A (en) 2011-11-18 2022-04-12 알닐람 파마슈티칼스 인코포레이티드 RNAi AGENTS, COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING TRANSTHYRETIN (TTR) ASSOCIATED DISEASES
GB201121069D0 (en) 2011-12-07 2012-01-18 Isis Innovation Delivery system
SG11201404596UA (en) * 2012-02-02 2014-10-30 Senex Biotechnology Inc Cdk8/cdk19 selective inhibitors and their use in anti-metastatic and chemopreventative methods for cancer
EP2914266B1 (en) 2012-11-01 2019-06-19 University of South Carolina Cdk8/cdk19 selective inhibitors for use in a method for treating prostate cancer
WO2015002956A1 (en) 2013-07-01 2015-01-08 Ohio State Innovation Foundation Exosome delivery system
US20170115308A1 (en) 2014-11-14 2017-04-27 Senex Biotechnology, Inc. Assay to measure efficacy of cdk8/19 inhibitors
CN114921468A (en) * 2017-01-30 2022-08-19 国立大学法人京都大学 Novel compound and method for producing regulatory T cell

Also Published As

Publication number Publication date
WO2019055977A9 (en) 2019-04-25
JP2020534354A (en) 2020-11-26
US20210121495A1 (en) 2021-04-29
CA3075784A1 (en) 2019-03-21
AU2018333072B2 (en) 2022-09-29
EP3684420A1 (en) 2020-07-29
US11471477B2 (en) 2022-10-18
EP3684420A4 (en) 2021-06-16
AU2018333072A1 (en) 2020-03-26
CN111278469A (en) 2020-06-12
WO2019055977A1 (en) 2019-03-21
JP7392954B2 (en) 2023-12-06

Similar Documents

Publication Publication Date Title
Chen et al. Circular RNA circRHOBTB3 represses metastasis by regulating the HuR-mediated mRNA stability of PTBP1 in colorectal cancer
US20230285439A1 (en) Methods for treating triple-negative breast cancer
Baluapuri et al. MYC recruits SPT5 to RNA polymerase II to promote processive transcription elongation
Li et al. Epigenetic and transcriptional control of the epidermal growth factor receptor regulates the tumor immune microenvironment in pancreatic cancer
Zhao et al. The IncRNA SNHG5/miR‐32 axis regulates gastric cancer cell proliferation and migration by targeting KLF4
Chen et al. MicroRNA-211 enhances the oncogenicity of carcinogen-induced oral carcinoma by repressing TCF12 and increasing antioxidant activity
Yeh et al. MicroRNA‐138 suppresses ovarian cancer cell invasion and metastasis by targeting SOX4 and HIF‐1α
Pekow et al. miR-193a-3p is a key tumor suppressor in ulcerative colitis–associated colon cancer and promotes carcinogenesis through upregulation of IL17RD
Duan et al. HOXA13 is a potential GBM diagnostic marker and promotes glioma invasion by activating the Wnt and TGF-β pathways
Shi et al. Circular RNA ANAPC7 inhibits tumor growth and muscle wasting via PHLPP2–AKT–TGF-β signaling Axis in pancreatic cancer
Rossi et al. Circular RNA ZNF609/CKAP5 mRNA interaction regulates microtubule dynamics and tumorigenicity
Matsuda et al. Extracellular vesicle‐based therapeutic targeting of β‐catenin to modulate anticancer immune responses in hepatocellular cancer
Li et al. LncRNA PRADX-mediated recruitment of PRC2/DDX5 complex suppresses UBXN1 expression and activates NF-κB activity, promoting tumorigenesis
US20100305188A1 (en) Nucleic acid capable of regulating the proliferation of cell
Andolfo et al. The micro-RNA 199b-5p regulatory circuit involves Hes1, CD15, and epigenetic modifications in medulloblastoma
US20190284554A1 (en) Micrornas and methods of their use
WO2012087983A1 (en) Polycomb-associated non-coding rnas
Chen et al. Non-coding RNAs, guardians of the p53 galaxy
Wang et al. Altered p53 regulation of miR-148b and p55PIK contributes to tumor progression in colorectal cancer
Bhattacharjya et al. Inhibition of nucleoporin member Nup214 expression by miR-133b perturbs mitotic timing and leads to cell death
Liu et al. MiR-144 inhibits tumor growth and metastasis in osteosarcoma via dual-suppressing RhoA/ROCK1 signaling pathway
Xu et al. STON2 negatively modulates stem-like properties in ovarian cancer cells via DNMT1/MUC1 pathway
Chang et al. Micro RNA‐551b‐3p inhibits tumour growth of human cholangiocarcinoma by targeting Cyclin D1
Li et al. MiR-1-3p inhibits lung adenocarcinoma cell tumorigenesis via targeting protein regulator of cytokinesis 1
Zheng et al. uc. 77-downregulation promotes colorectal cancer cell proliferation by inhibiting FBXW8-mediated CDK4 protein degradation

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CLARK, MICHAEL F.;HSIEH, ROBERT W.;REEL/FRAME:060968/0506

Effective date: 20190206

AS Assignment

Owner name: THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY, CALIFORNIA

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE LAST NAME OF ASSIGNOR MICHAEL F. CLARKE ON THE COVERSHEET PREVIOUSLY RECORDED AT REEL: 060968 FRAME: 0506. ASSIGNOR(S) HEREBY CONFIRMS THE ASSIGNMENT;ASSIGNORS:CLARKE, MICHAEL F.;HSIEH, ROBERT W.;REEL/FRAME:061495/0364

Effective date: 20190206

AS Assignment

Owner name: THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY;REEL/FRAME:061838/0154

Effective date: 20221103

Owner name: CHAN ZUCKERBERG BIOHUB, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE BOARD OF TRUSTEES OF THE LELAND STANFORD JUNIOR UNIVERSITY;REEL/FRAME:061838/0154

Effective date: 20221103

AS Assignment

Owner name: CZ BIOHUB SF, LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHAN ZUCKERBERG BIOHUB, INC.;REEL/FRAME:062700/0478

Effective date: 20230127

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION