WO2020165570A1 - Procédés se rapportant à la rupture de la liaison de protéines partenaires af9 à af9 et/ou enl - Google Patents

Procédés se rapportant à la rupture de la liaison de protéines partenaires af9 à af9 et/ou enl Download PDF

Info

Publication number
WO2020165570A1
WO2020165570A1 PCT/GB2020/050307 GB2020050307W WO2020165570A1 WO 2020165570 A1 WO2020165570 A1 WO 2020165570A1 GB 2020050307 W GB2020050307 W GB 2020050307W WO 2020165570 A1 WO2020165570 A1 WO 2020165570A1
Authority
WO
WIPO (PCT)
Prior art keywords
enl
peptide
dot1l
inhibitor
cancer
Prior art date
Application number
PCT/GB2020/050307
Other languages
English (en)
Inventor
Markus MUELLNER
Grahame James Mckenzie
Alberto Moreno DE LA GÁNDARA
Original Assignee
Phoremost Limited
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 Phoremost Limited filed Critical Phoremost Limited
Publication of WO2020165570A1 publication Critical patent/WO2020165570A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • 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
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5014Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing toxicity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • KRAS has generally been considered to be "undruggable" for many years.
  • the discovery of so-called 'synthetic lethal' cellular targets whose inhibition can selectively kill cancer cells carrying oncogenic KRAS mutations is, therefore, of significant interest in cancer therapy.
  • KRAS K-Ras, K-ras, Ki-ras
  • HRAS Epidermal Growth Factor Receptor
  • NRAS Epidermal Growth Factor Receptor
  • Ras at the conserved codons 12, 13 or 61 corresponding to amino acid residues G12, G13 or Q61
  • GAPs GTPase activating proteins
  • Ras isoforms display distinct codon-specific mutational profiles (Prior et al., 2012).
  • KRAS is typically mutated at codon 12 or codon 13 and whilst mutations at both sites are activating, due to impaired GAP binding, the position of the mutation has functional and clinical relevance.
  • Metastatic colorectal cancer is one of the leading causes of cancer related death world-wide. Overall survival is relatively poor; first-line therapy for advanced colon cancer involves treatment with anti-EGFR monoclonal antibodies such as Cetuximab in combination with standard chemotherapy. Failure to respond to Cetuximab is common and a key determinant of this resistance is the presence of activating mutations in KRAS, which are present in approximately one third of CRC tumours. Consequently, mCRC tumours are routinely genotyped for KRAS status, to predict Cetuximab responsiveness and this therapy is restricted to patients with homozygous wild type KRAS allele.
  • KRAS mutations occur in over 90% of human pancreatic cancers, almost always at codon 12.
  • the KRAS mutations occur relatively early in cancer development: while they are rare in early pancreatic intraepithelial neoplasm (PIN) lesions, they are present in the majority of advanced lesions and are near universal in frank pancreatic cancers. 5 year survival rates for patients with pancreatic cancer diagnoses are very poor: the disease is one of the most lethal of all neoplasms.
  • KRAS mutant KRAS has been largely unsuccessful as a cancer therapy. Significant efforts have been made to develop therapies targeting downstream elements of the RAS signalling pathways, including RAF, MEK, ERK and PI3K inhibitors, but none of these agents have yet been approved to treat KRAS mutant colon cancer, in particular.
  • the difficulties with targeting mutant KRAS directly or via downstream effectors mean that there remains a need to identify new therapeutic approaches to the treatment of cancers that harbour KRAS mutations, in particular colon cancers that harbour KRAS mutations.
  • several other types of cancer are characterised by relatively high frequencies of KRAS mutation, including pancreatic and non-small- cell lung cancer.
  • the present invention is based on identifying KRAS mutated cancers as being suitable for treatment with AF9 and/or ENL inhibitors or DOTH inhibitors.
  • the present inventors have shown that in KRAS mutated cancer cell lines the AF9 and/or ENL gene(s) are important to cell viability.
  • Inhibiting the activity of the AF9/ENL proteins for example by using inhibitors that block the binding of one or more AF9/ENL partner proteins (such as DOT1L and AF4) to AF9/ENL, has enhanced lethality in cells harbouring the KRAS mutation.
  • the invention therefore provides a new therapeutic approach for the treatment of KRAS mutant cancers, inhibitors of AF9 and/or ENL or DOT1L that may be useful in the treatment of KRAS mutant cancer, new screening methods to identify agents that are useful in the treatment of KRAS mutated cancer and new methods of selecting and/or treating patients with cancers that harbour KRAS mutations.
  • the invention provides inhibitors of AF9 and/or ENL, or DOT1L, for use in a method of treating an individual having cancer, wherein the cancer has a mutation in KRAS.
  • the cancer may be a non-haematological cancer, e.g. a solid tumour.
  • Cancers which may be treated include those that have a high incidence of Kras mutations including colon, lung, pancreas, peritoneum, biliary tract, small intestine, ovarian and endometrial cancers.
  • Suitable inhibitors of AF9 and/or ENL, or DOT1L, for use in the method of treatment include nucleic acids, antibodies, small molecules, peptides, or inducers of AF9 and/or ENL or DOT1L degradation.
  • the inhibitor is a peptide that binds to AF9 and/or ENL, preferably the C terminus of AF9 and/or ENL, for example, a peptide comprising or consisting of the Ras002 amino acid sequence (SEQ ID NO: 1), or a functional fragment thereof; or a peptide having 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the Ras-002 peptide; or a peptide comprising or consisting of the DOT1L Sites 2 and 3 peptide amino acid sequence (SEQ ID NO: 2), or a functional fragment thereof such as DOTlL-18-site 3, DOTlL-10-site 2, DOTlL-7-site 2,
  • the inhibitors bind to the C-terminus of AF9 and/or ENL. Binding to this region of the AF9 and/or ENL proteins may be advantageous because binding to this region is believed to block binding of endogenous DOT1L and/or other AF9 and/or ENL partner proteins.
  • AF9 partner protein include DOT1L and AF4, whose binding to AF9/ENL could be blocked. It has been reported by Shen et al., (J Biol Chem 2013 288(42):30585-30596) that the interaction of AF9/ENL is mutually exclusive with AF4 and DOTH. It was demonstrated that both AF4 and DOTIL compete for the same binding site on the C-terminal of AF9. Blocking the binding of one the AF9 partner proteins to AF9 could result in blocking the binding of other AF9 partner proteins.
  • the inhibitor of AF9 and/or ENL is selected from any of the inhibitors provided in WO2014127191.
  • the inhibitor is an RNAi molecule or a siRNA molecule or a shRNA molecule or a sgRNA or gRNA designed to recruit an RNA-guided nuclease.
  • the inhibitor is an agent that inhibits DOTIL by binding to DOTIL and thereby reducing the enzymatic activity of DOTIL, or is an agent that binds to DOTIL and thereby prevents DOTIL from binding to AF9 and/or ENL.
  • the DOTIL inhibitor is selected from any of the inhibitors in W02014100662A1.
  • inhibitors and methods of treatment provided herein may be combined with additional anti-cancer therapies.
  • the method of treatment is utilised to treat cancers that have been identified as comprising a mutation in KRAS.
  • the method may further include the step of determining whether the cancer has a mutation in KRAS.
  • a method of screening for agents useful in the treatment of cancer comprising: (a) contacting an AF9 and/or ENL protein with at least one candidate agent; and (b) selecting a candidate agent that inhibits the activity of the AF9 and/or ENL protein as being useful for the treatment of a cancer having a mutation in KRAS.
  • the method may be performed by adding the candidate agent to a cell line that comprises one or more mutations in KRAS.
  • the method may be performed by contacting AF9 and/or ENL protein with at least one candidate agent and the inhibitor of AF9 and/or ENL, or DOTIL such as Ras002, DOTILsites 1 and 3 or DOTIL sites 1 peptides; and selecting the candidate agent that disrupts the interaction between the inhibitors and AF9 and/or ENL.
  • This method would select for a candidate agent that competes with Ras002 or DOTIL derived peptides to bind to AF9 and/or ENL.
  • a peptide comprising or consisting of the Ras002 amino acid sequence (SEQ ID NO: 1), or a functional fragment thereof, or a peptide having 70%, 75%, 80%, 85%, 90% ,95%, 96%, 97%, 98% or 99% identity to the Ras-002 peptide (SEQ ID NO:l); or a peptide comprising or consisting of the DOTIL Sites 2 and 3 peptide amino acid sequence (SEQ ID NO: 2), or a functional fragment thereof, or a peptide having 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to the DOTIL Sites 2 and 3 peptide; or a peptide comprising or consisting of the DOTIL Sites 1 peptide amino acid sequence (SEQ ID NO: 3), or a functional fragment thereof; or a peptide having 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%
  • the peptide binds to AF9 and/or ENL, preferably the C-terminus of AF9 and/or ENL.
  • nucleic acid molecules comprising a nucleic acid sequence encoding any of the peptides provided herein, vectors comprising the nucleic acid sequences and cells containing the nucleic acids/vectors.
  • binding moieties that bind to the peptides provided herein, for example antibodies or functional fragment thereof.
  • the peptides, nucleic acids, vectors or cells may be for use in medicine, particularly for use in the treatment of cancer.
  • the novel peptides provided herein, particularly those that bind to AF9 and/or ENL, may be useful for treating leukaemia, particularly MLL-fusion protein associated leukaemia.
  • compositions comprising any of the inhibitors (for example peptides), nucleic acid molecules, vectors, or binding moieties together with a pharmaceutically acceptable carrier.
  • Figure 1 Analysis of PROTEINi probes identified in the screen. Each dot represents a PROTEINi sequence and the circle size is 1/CV of replicates. The large circles have less noise. The average number of hits was measured against Score (l/p_value). The hit threshold was p ⁇ 0.01. Ras-002 is circled in the figure.
  • Ras-002 demonstrates a consistent reduction in cell viability with oncogenic KRAS in follow-up studies. Normalised cell viability is shown on the y-axis. Reduced cell numbers indicate cell death. The shaded columns represent cells where oncogenic KRAS is induced with doxycycline.
  • FIG. 3 Ras-002 binds to the crucial, C-terminal region of the proteins encoded by the MLLT3 and MLLT1 genes. Note the very high amino acid identity between MLLT3 and MLLT1 proteins (known as AF9 and ENL respectively) in this region.
  • FIG. 4 Ras-002 binds to the crucial, C-terminal region of the proteins encoded by the MLLT3 and MLLT1 genes. Note the very high amino acid identity between MLLT3 and MLLT1 proteins (known as AF9 and ENL respectively) in this region.
  • FIG. 5 The DOT1L peptide sequence engages C-terminal MLLT3 through a clear peptide binding groove.
  • Ras-002 and DOTIL-peptide both show synthetic lethality in the presence of oncogenic KRAS, and this can be rescued by ectopic AF9 expression.
  • Figure 7 Fluorescence polarisation assay shows the binding of DOTIL-peptides to AF9 C- terminal.
  • DOTlL-18-site 3 and DOTlL-10-site have a higher affinity for AF9 compared to DOTlL-7-site 2 and DOTlL-7-site 3 derived peptides.
  • the data provided herein indicates that the disrupting the activity of AF9 and/or ENL protein(s) may be useful in the treatment of cancers, particularly cancers having a mutation in KRAS.
  • the Ras-002 peptide provided herein binds to AF9 and ENL and kills cancer cells having one or more KRAS mutations.
  • the Ras-002 peptide, and agents that mimic the activity of Ras-002 may be useful in the treatment of cancers having one or more KRAS mutations. It is hypothesised that RAS002 competes for binding with one or more endogenous partner proteins (such as DOT1L) at the C terminal domain of AF9/ENL.
  • endogenous partner proteins such as DOT1L
  • AF9/ENL have a number of partner proteins (see for example those listed in Table 1 below which are known partner proteins of AF9) and preventing the binding of one or more of these proteins to AF9/ENL may function to inhibit the activity of AF9/ENL and thus be useful in the treatment of KRAS mutant cancers.
  • Inhibitors of AF9/ENL activity that may be employed in the methods of treatment, uses or assays provided herein include AF9/ENL binding agents (inhibitors) that bind to AF9 and/or ENL and block the protein-protein interactions between AF9 and/or ENL and one or more endogenous partner proteins e.g. such as DOTIL, thereby inhibiting the activity of the AF9 and/or ENL, for example by inhibiting the formation and/or activity of DOTIL: AF9/ENL complex.
  • Inhibitors of AF9 and/or ENL activity may function by blocking binding of one or more endogenous partner proteins at the C terminal domain of AF9 and/or ENL (amino acids 498 to 568 of AF9).
  • AF9/ENL binding agents include the Ras-002 peptide (SEQ ID NO:l) or analogues thereof, or the DOTIL peptides such as the DOTIL Sites 2 and 3 peptide- see SEQ ID NO:2, and the DOTIL Site 1 peptide SQISEKQRHCLELQISIVELEKSQRQ (SEQ ID NO: 3), which is sometimes referred to as DOTIL 628-653 sequence.
  • DOTIL sites 2 and 3 peptides are listed in Table 2 and Table 3.
  • AF9/ENL binding agents include those described in WO2014127191 (Al),
  • inhibiting the activity of AF9 and/or ENL may be
  • an agent e.g., a peptide, a small molecule, peptidomimetic, a cyclic peptide
  • an agent e.g., a peptide, a small molecule, peptidomimetic, a cyclic peptide
  • such an inhibitor has binding properties similar to the region of DOT1L corresponding to DOTIL 865LPISIPLSTV874.
  • such binding of the region of AF9 and/or ENL that binds with DOTIL furthermore results in prevention of aberrant H3K79 methylation in KRAS mutant cancers.
  • the present invention provides methods for screening drugs through identifying agents capable of, for example, binding to the C terminal domain of AF9 and/or ENL, and/or more specifically the interaction site for DOTIL (e.g., an agent having binding properties similar to the region of DOTIL corresponding to DOTIL 865LPISIPLSTV874).
  • agents capable of, for example, binding to the C terminal domain of AF9 and/or ENL, and/or more specifically the interaction site for DOTIL (e.g., an agent having binding properties similar to the region of DOTIL corresponding to DOTIL 865LPISIPLSTV874).
  • the present invention is not limited to certain types or kinds of agents capable of binding the AF9/ENL interaction site for DOTIL.
  • the agent is a peptide capable of binding the MLL-fusion protein interaction site for DOTH (e.g., the AF9/ENL interaction site for DOTH).
  • the peptide comprises 865LPISIPLSTV874 of DOTH.
  • the agent is a peptidomimetic designed and synthesized to mimic a peptide capable of binding a region of AF9 or ENL known to interact with DOT1L (e.g., 865LPISIPLSTV874 of DOTIL).
  • the agent is a cyclic peptide designed and synthesized to mimic a peptide capable of binding a region of AF9 or ENL known to interact with DOTI1L (e.g., 865LPISIPLSTV874 of DOTIL).
  • DOTI1L e.g., 865LPISIPLSTV874 of DOTIL
  • the agent is a small molecule capable of binding to the AF9 and/or ENL interaction site for DOTIL.
  • DOTIL inhibitors to reduce the amount of endogenous DOTIL available to bind to AF9 and/or ENL and thereby reduce the activity of AF9/ENL:DOTlL inhibitors include agents that bind to the DOTIL enzyme and inhibit the enzymatic activity thereof, or agents that bind to DOTIL and block DOTIL binding to AF9/ENL.
  • DOTIL and/or the DOTIL binding site on AF9/ENL may be identified by the use of high throughput screening strategies.
  • detecting compounds that interact with DOTIL and/or the DOTIL binding site on AF9/ENL those skilled in the art will be familiar with biophysical techniques such as surface plasmon resonance, thermal melt assays, fluorescence polarization assays, homogeneous time resolved fluorescence assays or other binding assays such as those provided on the KinomeScan platform (DiscoverX).
  • DOTIL and/or the DOTIL binding site on AF9/ENL for example the nucleic acid sequence, the amino acid sequence, the secondary, tertiary or quaternary structure of the proteins, e.g. the crystal structure of AF9/ENL, the location of particular functional domain(s) within the nucleic acid sequence, amino acid sequence or protein structure, the location of domain(s) that interact with a receptor or target of AF9/ENL, can be exploited in order to design putative AF9/ENL or DOTIL inhibitors for use in any aspect of the invention.
  • small molecule inhibitors could be identified using a range of assay technologies which monitor Protei Protein interactions. Examples are the NanoBit split reporter system, a Fluorescence Polarisation (FP) Assay, or an Enzyme-Linked Immunosorbent Assay (ELISA). Any small molecule capable of disrupting the interaction between Ras-002 or the DOT1L peptides and AF9 in such an assay might reasonably be expected to block the interaction between AF9 and its endogenous ligands in a cell. By extension from the synthetic lethal data provided herein, it is expected that such small molecules should be able to selectively kill cancer cells carrying oncogenic KRAS mutations.
  • assay technologies which monitor Protei Protein interactions. Examples are the NanoBit split reporter system, a Fluorescence Polarisation (FP) Assay, or an Enzyme-Linked Immunosorbent Assay (ELISA).
  • FP Fluorescence Polarisation
  • ELISA Enzyme-Linked Immunosorbent Assay
  • the present invention extends to the use of agents or molecules found to interact with DOT1L (e.g. DOT1L inhibitors) and/or the DOT1L binding site on AF9/ENL (AF9/ENL inhibitors) including those identified using the screening methods disclosed herein and to derivatives thereof.
  • DOT1L e.g. DOT1L inhibitors
  • AF9/ENL inhibitors e.g. AF9/ENL inhibitors
  • a person skilled in the art will also recognise that it will be possible to take small molecules that interact with DOT1L and/or the DOT1L binding site on AF9/ENL and modify them to include linker groups (e.g. flexible polyethylene glycol linkers) and a second protein-interacting moiety known to interact with the ubiquitin ligase system that is responsible for a large fraction of protein turnover in the cell.
  • linker groups e.g. flexible polyethylene glycol linkers
  • a second protein-interacting moiety known to interact with the ubiquitin ligase system that is responsible for a large fraction of protein turnover in the cell.
  • Suitable targets for the second protein-interacting moiety include VEIL, MDM2, cerebelon and c-IAP.
  • Bifunctional compounds comprise BRD4 antagonists such as JQ1 and cerebelon ligands such as pomalidomide will be known by those skilled in the art to target BRD4 for ubiquitin/proteosome mediated destruction.
  • Agents useful for treating cancer includes the peptides provided herein for example peptides that bind to AF9/ENL and block DOT1L binding to AF9/ENL, e.g. Ras-002, including functional fragments thereof.
  • Peptides may be also be generated that block the catalytic sites of the DOT1L enzyme.
  • Peptide fragments can be readily prepared according to well-established, standard liquid or, preferably, solid-phase peptide synthesis methods, general descriptions of which are broadly available.
  • amino acid residues comprising the peptides described herein may be chemically modified.
  • chemical modifications include those corresponding to post translational modifications for example phosphorylation, acetylation and deamidation.
  • the N or C terminal ends of the peptide may be modified to improve the stability, bioavailability and or affinity of the peptides.
  • the peptides described herein may be modified by amino acid substitution, insertion or deletion. Amino acid substitution means that an amino acid residue is substituted for a replacement amino acid residue at the same position.
  • Inserted amino acid residues may be inserted at any position and may be inserted such that some or all of the inserted amino acid residues are immediately adjacent to one another or may be inserted such that none of the inserted amino acid residues is immediately adjacent another inserted amino acid residue.
  • Inserted amino acids and replacement amino acids may be naturally occurring amino acids or may be non-naturally occurring amino acids and, for example, may contain a non-natural side chain, and/or be linked together via non-native peptide bonds.
  • the peptides of the invention bind to the C terminus of AF9/ENL.
  • the amino acid modifications described above will not impair the ability of the peptide to bind AF9/ENL.
  • the amino acid modifications improve the ability of the peptide to bind AF9/ENL.
  • Inhibitors of the invention may be conjugated to additional moieties such as carrier molecules. Carriers are compounds and substances that improve and/or prolong the delivery of an active ingredient to a subject in the context of a pharmaceutical composition.
  • Carrier may serve to prolong the in vivo activity of a drug or slow the release of the drug in a subject, using controlled-release technologies. Carriers may also decrease drug metabolism in a subject and/or reduce the toxicity of the drug. Carrier can also be used to target the delivery of the drug to particular cells or tissues in a subject.
  • Common carriers include fat emulsions, lipids, PEGylated phospholids, PEGylated liposomes, PEGylated liposomes coated via a PEG spacer with a cyclic RGD peptide c(RGDDYK), liposomes and lipospheres, microspheres (including those made of biodegradable polymers or albumin), polymer matrices, biocompatible polymers, protein-DNA complexes, protein conjugates, erythrocytes, vesicles, nanoparticles, and side-chains for hydro carbon stapling.
  • the aforementioned carriers can also be used to increase cell membrane permeability of the AF9 and/or ENL inhibitors of the invention.
  • carriers may also be used in compositions for other uses, such as research uses in vitro (e.g., for delivery to cultured cells) and/or in vivo.
  • the peptides may also be employed in accordance with the present invention by expression of the peptides in vivo, i.e., via gene therapy approaches known in the art.
  • Another class of inhibitors useful for treatment of KRAS mutated cancer includes nucleic acid inhibitors of AF9/ENL, or the complements thereof, which inhibit activity or function by down regulating production of active polypeptide. This can be monitored using conventional methods well known in the art, for example by screening using real time PCR as described in the examples.
  • RNAi RNAi
  • Xing et al. Int J Mol Med. 2016 Aug; 38(2): 407-416
  • Zhao et al. Mol Med Rep. 2015 Jul; 12(1): 960-966
  • Zheng et al. Acta Pharmacol Sin. 2009 Dec; 30(12): 1625-1633
  • Zhu et al. Genet Mol Biol. 2012;35:538- 544
  • DNA or RNA guided nuclease systems can also be used to inhibit AF9/ENL activity by editing the AF9/ENL gene to knock out AF9/ENL expression or reduce functional AF9/ENL protein.
  • a guide nucleic e.g., a guide RNA that binds to an AF9/ENL nucleic acid is administered to or expressed in the cell, along with a CRISPR/Cas9 nuclease that results in targeted cleavage of the AF9/ENL gene sequence and introduces a mutation in the gene that inhibits the production of functional AF9/ENL, see, e.g., Jinek et al.
  • the CRISPR/Cas system can alternatively be used like RNA interference, turning off the AF9/ENL gene in a reversible fashion.
  • the RNA can guide the Cas protein to the promoter, sterically blocking RNA polymerases.
  • the AF9/ENL inhibitors may also be employed in accordance with the present invention by expression of the inhibitors in vivo, i.e., via gene therapy.
  • the present invention also encompasses: (i) polynucleotide sequences encoding AF9 and/or ENL inhibitors (for example the Ras002 or DOT1L peptides, or functional fragments or variants thereof) or polynucleotide sequences encoding DOT1L inhibitors of the present invention, (ii) vectors into which the polynucleotide sequences are inserted, (iii) host cells genetically engineered (transduced, transformed, or transfected) with the vectors, (iv) methods of culturing the host cells under conditions promoting production of the AF9 and/or ENL inhibitors encoded by the polynucleotide sequences.
  • AF9/ENL inhibitor as used in the context of any aspect of the present invention encompasses a molecule or agent that results in a decrease in the activity or function of AF9/ENL.
  • the molecule or agent could act at the nucleic acid level or at the protein level of AF9/ENL.
  • the decrease in the activity or function of AF9/ENL may be as a result of a reduction in the level of AF9/ENL. This reduction in the level of AF9/ENL may be due to e.g. reduced expression of AF9/ENL or degradation of AF9/ENL.
  • the present invention is concerned with methods of screening candidate compounds to determine whether one or more candidate agents (putative AF9/ENL inhibitors) are likely to be useful for the treatment of KRAS mutated cancer, including solid tumours having one or more KRAS mutations.
  • candidate agents putative AF9/ENL inhibitors
  • approaches that may be used for these methods of screening, either alone or in any combination or order.
  • a method of screening may involve using cell lines to profile candidate agents to identify and select compounds that are synthetically lethal to cells with KRAS mutations: namely compounds that are cytotoxic to a high fraction of cells or cell lines that are KRAS mutant, but less toxic to cells or cell lines with homozygous wild-type KRAS alleles including normal cells. For example, a higher level of cytotoxicity is observed in the KRAS mutant cells than compared to the wildtype cells. Suitable methods of determining cytotoxicity are described herein.
  • the candidate agents show a greater lethal effect in KRAS mutant cells as compared to wildtype cells.
  • the present invention also includes methods of screening that employ AF9/ENL as a protein target for the screening of candidate compounds to find AF9/ENL inhibitors. Accordingly, methods of screening may be carried out for identifying candidate agents that are capable of inhibiting AF9/ENL for subsequent use of development as agents for the treatment of KRAS mutated cancer. Conveniently, this may be done in an assay buffer to help the components of the assay interact, and in a multiple well format to test a plurality of candidate agents. The activity of AF9/ENL can then be determined in the presence and absence of the one or more candidate compounds to determine whether a given candidate is an inhibitor of AF9/ENL.
  • the candidate agent may be a known or putative AF9/ENL inhibitor, an antibody, a peptide, a nucleic acid molecule or an organic or inorganic compound, e.g. a small molecule with molecular weight of less than 100 Da.
  • the agent in question may be tested to determine whether it is not lethal to normal cells or otherwise is suited to therapeutic use. Following these studies, the agent may be manufactured and/or used in the preparation of a medicament, pharmaceutical composition or dosage form.
  • the present invention provides methods and medical uses for the treatment of cancer, particularly KRAS mutated cancer.
  • a cancer may be identified as KRAS mutated by testing a sample of cancer cells from an individual, for example to determine whether the KRAS protein contains one or more mutations or to determine the expression of the KRAS gene to evaluate whether expression is increased compared to normal. It is known that KRAS mutations are present at high frequency in colon, lung, pancreas, peritoneum, biliary tract, small intestine, ovarian and endometrial cancers. Cancer types that may be treatable in accordance with the methods of the invention include colon, lung, pancreas, peritoneum, biliary tract, small intestine, ovarian and endometrial cancers.
  • Cancer types that may be treatable in accordance with the methods of the invention include colon, lung, peritoneum, biliary tract, small intestine, ovarian and endometrial cancers.
  • KRAS mutated cancers that are treatable in accordance with the present invention are likely to be found as further work is done to identify the prevalence of KRAS mutations, and to confirm that the presence of the KRAS mutation(s) is associated with AF9/ENL expression.
  • cancer types that may be treatable in accordance with the methods of the invention include but are not limited to: adrenal gland, autonomic ganglia, biliary tract, bladder, bone, breast, CNS, cervix, endometrial, eye, fallopian tube, gastrointestinal tract, genital tract, haematopoietic and lymphoid tissue, kidney, large intestine, liver, lung, meninges, nervous system, oesophagus, ovary, pancreas, penis, peritoneum, pleura, prostate, salivary gland, skin, small intestine, soft tissue, stomach, testis, thymus, thyroid, upper aerodigestive tract, urinary tract, or vulva.
  • databases such as the COSMIC database include further information on the cancer types that have been identified as containing KRAS activating mutations, including G12 and G13 mutations.
  • composition comprising an agent for use in the in the treatment of cancer combined with any pharmaceutically acceptable carrier, adjuvant or vehicle wherein the cancer is a KRAS mutated cancer.
  • compositions may be in the form of, for example, tablets, capsules, powders, granules, elixirs, lozenges, suppositories, syrups and liquid preparations including suspensions and solutions.
  • composition in the context of this invention means a composition comprising an active agent and comprising additionally one or more pharmaceutically acceptable carriers.
  • the composition may further contain ingredients selected from, for example, diluents, adjuvants, excipients, vehicles, preserving agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavouring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispersing agents, depending on the nature of the mode of administration and dosage forms.
  • compositions of the invention may be administered orally in any orally acceptable dosage form including, but not limited to tablets, dragees, powders, elixirs, syrups, liquid preparations including suspensions, sprays, inhalants, tablets, lozenges, emulsions, solutions, cachets, granules and capsules.
  • dosage forms are prepared according to techniques known in the art of pharmaceutical formulation.
  • the pharmaceutical compositions may be administered nasally. Suitable formulations for this purpose are known to those skilled in the art.
  • compositions of the invention may be administered by injection and may be in the form of a sterile liquid preparation for injection, including liposome preparations.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing the active compound into association with a carrier, which may constitute one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • compositions comprising agents disclosed herein for the treatment of cancer, particularly the treatment of KRAS mutated cancer may be used in the methods described herein in combination with standard chemotherapeutic regimes or in conjunction with radiotherapy.
  • chemotherapeutic agents include Amsacrine (Amsidine), Bleomycin, Busulfan, Capecitabine (Xeloda), Carboplatin, Carmustine (BCNU), Chlorambucil (Leukeran), Cisplatin, Cladribine (Leustat), Clofarabine (Evoltra), Crisantaspase (Erwinase), Cyclophosphamide, Cytarabine (ARA-C), dacarbazine (DTIC), Dactinomycin (Actinomycin D), Daunorubicin, Docetaxel (Taxotere), Doxorubicin, Epirubicin, Etoposide (Vepesid, VP-16), Fludarabine (Fludara), Fluorouracil
  • Administration in-vivo can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals) throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • a suitable dose of the active compound is in the range of about 100 pg to about 250 mg per kilogram body weight of the subject per day.
  • the active compound is a salt, an ester, prodrug, or the like
  • the amount administered is calculated on the basis of the parent compound, and so the actual weight to be used is increased proportionately.
  • the active agents disclosed herein for the treatment of KRAS mutated cancer according to the present invention are preferably for administration to an individual in a "prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual.
  • a prophylaxis may be considered therapy
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc., is within the responsibility of general practitioners and other medical doctors, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners.
  • compositions may be administered alone or in combination with other treatments, either simultaneously or sequentially, dependent upon the condition to be treated.
  • the present invention also encompasses: (i) polynucleotide sequences encoding AF9 and/or ENL inhibitors (for example polynucleotides encoding the Ras002 or DOT1L peptides, or functional fragments or variants thereof) or DOT1L inhibitors of the present invention, (ii) vectors into which the polynucleotide sequences are inserted, (iii) host cells genetically engineered (transduced, transformed, or transfected) with the vectors, (iv) methods of culturing the host cells under conditions promoting production of the AF9 and/or ENL inhibitors encoded by the polynucleotide sequences.
  • polynucleotide sequences encoding AF9 and/or ENL inhibitors (for example polynucleotides encoding the Ras002 or DOT1L peptides, or functional fragments or variants thereof) or DOT1L inhibitors of the present invention
  • targeting the DOT1L binding site on AF9/ENL may be useful in the treatment of non-haematological malignancies, e.g. solid tumors.
  • these solid tumours may comprise one or more KRAS mutations.
  • Cancer-driving KRAS mutations are colloquially termed activating mutations and increase the fraction of protein present in its signalling-promoting GTP-bound form. These mutations typically occur at codons 12 or 13 in human KRAS (but at lower frequency elsewhere in the protein, including codons 59 or 61) and impair the GTP-ase activity of the molecule.
  • the K-Ras mutation may comprise one or more activating mutations in codon 12, 13, 59 and/or 61.
  • the presence of a K-Ras mutation in a cancer can be determined by molecular diagnostic testing methods that include, but are not restricted to, mutation-specific PCR-amplification methods, tandem PCR amplification and DNA-sequencing methods (including Sanger-based, pyrosequencing- based and mass-spectrometry-based methods), whole genome sequencing, or proteomic-based technologies (including Western blotting, immuno-histochemistry and protein mass-spectrometry). Examples of such cancers are diverse, with K-Ras mutations being present in a significant proportion of many cancer types, including but not limited to colon, lung, pancreas, prostate, endometrium, ovarian, liver, thyroid, biliary tract, stomach and ovary.
  • the present invention may be useful in the treatment of these cancers or any other cancer that is identified as KRAS mutated.
  • the present invention may be useful in the treatment of KRAS mutated cancers in humans, or in other mammalian species.
  • PROTEINi ® probes Three million PROTEINi ® probes were used to scan for all targets that can selectively kill mutant KRAS cells. These probes are derived from DNA-encoded, protein-fragment expression libraries that can be screened in high throughput in phenotypic assays (such as described in WO 2013/116903); often dubbed 'Protein-interference' (PROTEINi ® ).
  • a cell line with Doxycycline-inducible KRAS G12V was constructed using the Flp-lnTM T-RExTM Core Kit (Invitrogen Cat#650001) and cells were expanded and infected with lentivirus encoding for the PROTEINi probes. Cells were infected at a multiplicity of infection of 0.3 in order to obtain cells carrying only one PROTEINi insert on average. Cells were subsequently selected on puromycin to remove uninfected cells and were propagated and expanded for an additional 5 days to remove all PROTEINi's that are toxic to HeLa cells without active Ras.
  • Ras-002 demonstrated an increased ability to kill cancer cells carrying an activating KRASG12V mutation compared to WT KRAS FleLa cells. It was associated with approximately 50% depletion of KRAS G12V cells (against control cells) in 72 hours ( Figure 2).
  • a yeast two hybrid system In order to discover the cellular target of Ras-002, and thus the mechanism through which it shows synthetic lethality with oncogenic KRAS, a yeast two hybrid system was employed. This system conceptually relies on the activation of a downstream reporter gene by binding of a transcription factor to an upstream activating sequence. The transcription factor is split into two fragments: one called the DNA binding domain and the other known as the activating domain. When the two fragments of the transcription factor are physically proximal, the transcription factor is functional and therefore can activate transcription of the reporter gene.
  • yeast two hybrid screening A third party contractor, Flybrigenics Services of Paris, France, was used to conduct the yeast two hybrid screening.
  • the most common screening approach is to carry out a survival screen.
  • the yeast is genetically engineered to lack the biosynthesis of a specific nutrient, on which the yeast depends on entirely for its survival.
  • the yeast is then transformed with the "bait", which in our case is the PROTEINi fused to the binding domain of GAL4 and a human cDNA library "prey" fused to the activating domain of GAL4.
  • the "bait" which in our case is the PROTEINi fused to the binding domain of GAL4 and a human cDNA library "prey" fused to the activating domain of GAL4.
  • MLLT3 A short-list of 5 high confidence interactions, based on sequences found to be in frame, in the sense orientation and not occurring frequently across multiple Y2H screens, was identified.
  • the prey genes encoded by these clones included MLLT3 and MLLT1. Of specific interest were the 48 clones encoding 10 different regions of MLLT3, and the 2 clones encoding MLLT1.
  • MLLT3 and MLLT1 are closely-related genes, both components of the super elongation complex.
  • MLLT3 encodes for the AF9 protein while MLLT1 encodes for the ENL protein.
  • Ras-002 interacts with the C-terminal region of MLLT3 and MLLT1.
  • Ras-002 and AF9/ENL are known to play a key role as oncogenic drivers in leukemia, this paved the path for a leukemia model.
  • Both proteins are common fusion partners of MLL in acute leukemias.
  • the C- terminal region of the proteins to which Ras-002 binds to is the same region which is translocated in pro-leukemic fusion transgenes. This adds significant weight to the hypothesis that Ras-002 binds to the crucial, oncogenic region of a fusion transgene that is commonly present in lymphoid and myeloid leukemias. Therefore, the Ras-002 and AF9/ENL interaction in leukemias is a second model being explored. This is shown in Figure 3.
  • Ras-002 shows significant amino acid identity to DOT1L, a histone methyltransferase, which has been implicated in the development of MLL-rearranged leukemias, and which is a known binding partner for the C-terminal region of AF9.
  • Mistargeting of DOT1L in the MLL-MLLT3 (also known as MLL-AF9) fusion transgene causes aberrant FI3K79 methylation at homeobox genes, and subsequent transcriptional dysregulation.
  • DOTIL's essential role in leukemic transformation has made it an attractive therapeutic target.
  • Ras-002 its mechanism of action is that it competes for M LLT3 binding with endogenous client proteins, such as DOT1L, and that oncogenic KRAS signalling relies upon some aspect of MLLT3::DOTlL-dependent histone methylation for its function. Further this data suggests that peptides derived from the AF9 binding domain of DOT1L should phenocopy the effects of Ras-002. This is shown in a simplified cartoon form in Figure 4.
  • the NMR structure is available for the MLLT3::DOTlL peptide complex (PDB: 2mv7) (Kuntimaddi et al., Cell Reports 11, 808-820, May 5, 2015).
  • the C-terminal of AF9 is partly unfolded in the absence of peptide ligand. There is a clear peptide groove, and within this grove, two residues (D544, D546) are essential for peptide binding. Additional binding energy is from hydrophobic interactions with the groove floor ( Figure 5).
  • Target validation experiments were carried out to a) confirm the Ras002 phenotype and validate the phenotype of DOT1L derived peptides in the presence of KRAS and b) identify if this expression changes upon overexpression of AF9. These experiments were carried out as stated below:
  • the PROTEINi ® was expressed in FleLa Flpln-TRex cells at a MOI of 0.3 to generate a stable cell line.
  • a PROTEINi ® control which contains the same nucleotide sequence as the PROTEINi ® but randomized so it is non-functional, was expressed in the same manner to generate a scramble control cell line.
  • the FleLa Flpln-TRex cells can be induced to express KRAS mutant upon addition of doxycycline. Cells not induced with doxycycline are referred to as wild type (WT) cells.
  • each of the two cell populations were plated into 96 well at 5000 cells/well and a 1:2 serial dilution of cell number carried out to 156 cells/well. Each condition was plated in triplicate. Following six hours of plating to allow the cells to adhere, 200 ng/ml doxcycyline was added to half the population of each of the two cell lines to induce KRASG12V mutant expression - the second half did not express KRAS mutant and therefore was used as a control. Following five days of treatment, cell proliferation was measured in the two populations using a Sulforhodamine B (SRB) assay.
  • SRB Sulforhodamine B
  • Nanobit split reporter assay system was employed. These experiments were carried out detailed below.
  • AF9 C-terminal region (498 to 568 aa) was PCR amplified from a gene synthesised full length AF9 ORF, and cloned into pBiTl.l-C[TK/LgBiT] (Promega) to generate the pBiT-LgC-AF9 using the Bglll and EcoRI sites.
  • AF9_DN_F_gra 5'- ACGTGGATCCATGGGCGAGTGCGACAAGGCCTACCTGG-3' (SEQ ID NO: 4) and AF9_DN_R_gra: 5'- ACGTCTCGAGTCAGCTGGTGCCGCTTGTTTCC-3' (SEQ ID NO:5)), and ligations were carried out using T4 DNA ligase (NEB) following manufacturer's recommendations.
  • NEB T4 DNA ligase
  • Wild type DOT1L region encompassing amino acids 862 to 890 and indicated mutants (in Table 2) were cloned into pBiT2.1-N[TK/SmBiT] (Promega) using Xhol and Bglll sites.
  • oligonucleotides corresponding to DOT1L sense and anti-sense region were synthesised (SIGMA), 5' ends phosphorylated using T4 PNK (NEB) according to manufacturer's instructions (1 uM oligonucleotide, 0.5 mM ATP, 10U of PNK enzyme in a total volume of 50 uL with T4 PNK buffer) for 60 minutes at 37°C, followed by inactivation of enzyme at 65°C for 20 minutes.
  • Double strand DNA were formed by mixing 10 uL of each corresponding DNA oligonucleotide phosphorylation reactions followed by boiling at 95°C for 3 minutes and allowed to slowly cool to ambient temperature. Ligations were carried out using T4 DNA ligase (NEB) following manufacturer's recommendations.
  • FIEK-293FT cells (Invitrogen) were seeded onto six well plates (Corning) at a total of 5E+05 per well in 2 mL total of DMEM supplemented with 10% FBS (Flyclone). Cells were transfected 24 hours after plating using jetPRIME reagent (Polyplus-transfection). In brief, 1 ug of each plasmid DNA expressing indicated proteins was mixed with 200 uL of jetPRIM E buffer, followed by 8uL of jetPRIM E reagent. The mixture was mixed by 10 second vortexing followed by brief centrifugation to bring contents down. Transfection mixture was added to corresponding well after a 10 minute incubation at ambient temperature. Cells were returned to incubator for further 24 hours.
  • NanoBit activity was carried out using the NanoGlo reagent (Promega). Media was removed from each well and replaced by 250 uL of fresh media equilibrated at ambient temperature. To each well 250 uL of NanoGlo reagent of ambient temperature equilibrated was added, prepared according to manufacturer's instructions. Plates were mixed vigorously in a plate shaker for 10 minutes, then 100 uL of each well was transferred into a white opaque 96 well assay plate in triplicate (Corning). Luminescence was read on a ClarioStar plate reader (BMG LABTECFI), and data analysed using Prism 7 (GraphPad).
  • a synthetic plasmid containing a DNA sequence encoding polyhistidine tag, GB1 solubility domain (B1 domain of Streptococcal protein G), TEV (Tobacco Etch virus) protease cleavage site and the C- terminal region of AF9 (residues 470 to 568) was synthesised by Thermo GeneArt services. This construct was further subcloned into Ncol and Xhol sites in pEt28a protein expression plasmid (Novagen).
  • E. coli BL21 (DE3) (NEB) bacteria were transformed with the pMPl-AF9 plasmid and grown overnight on LB agar containing 30 ug/mL of kanamycin antibiotic for selection. A single colony was then selected and used to inoculate 50 mL LB media supplemented with 30 ug/ mL kanamycin. It was then incubated overnight at 37°C with shaking (200 rpm). Overnight bacterial culture was then added (1:100, v/v) to fresh LB media supplemented with 30 ug/ mL kanamycin.
  • Cell pellet was resuspended (3:1, v/w) in lysis buffer (20 mM HEPES pH 7.5, 500 mM NaCI, 5% glycerol, 20 mM imidazole, 5 mM beta-mercaptoethanol, ImM PMSF, lx protease inhibitor cocktail EDTA-free (Roche)). Lysis of cells was carried out using a refrigerated FPG12800 homogeniser (Homogenising Systems Ltd) at 15,000 Psi. Cell lysate was then supplemented with Triton X-100 up to the final concentration of 0.5% and incubated at 4°C for lh. Extract was cleared by centrifugation at 25000 x g at 4C for 30 minutes, and soluble fraction pooled for further purification.
  • lysis buffer (20 mM HEPES pH 7.5, 500 mM NaCI, 5% glycerol, 20 mM imidazole, 5 mM beta-mercaptoethanol, ImM
  • the protein was initially purified using Immobilised Metal Affinity Chromatography (IMAC) in an AKTA Start (GE Healthcare). Cleared cell lysate was loaded onto a 5 mL HisTrap HP column (GE Healthcare) previously equilibrated with 10 column volumes of the buffer consisting of 20 mM HEPES pH 7.5, 500 mM NaCI, 5% glycerol, 5 mM beta-mercaptoethanol and 20 mM imidazole. The column was then washed with 150 mL of 20 mM HEPES pH 7.5, 500 mM NaCI, 5% glycerol, 5 mM beta-mercaptoethanol and 100 mM imidazole.
  • IMAC Immobilised Metal Affinity Chromatography
  • AF9 was eluted from the column by increasing the imidazole concentration to 300 mM. Fractions of the eluent were analysed by SDS-PAGE, and fractions containing AF9 were pooled and concentrated using a 5 kDa MWCO centrifugal concentrator (GE Healthcare) and quantified.
  • the protein was further purified using Size Exclusion Chromatography. HiLoad 16/600 Superdex 200 pg column (GE Healthcare) was equilibrated with 1 column volume of 20 mM HEPES pH 7.5, 250 mM NaCI and 1 mM TCEP. Samples containing 10 mg of AF9 were loaded onto the column and resolved using 1.5 column volume of the same buffer, with corresponding fractions collected and analysed by SDS-PAGE. Fractions containing AF9 were pooled, concentrated using a 5 kDa MWCO centrifugal concentrator (GE Healthcare), quantified and stored at -80°C.
  • peptides used for the development of the FP assay were designed by the inventors and purchased from Designer Biosciences (Cambridge, UK). Seven to eighteen amino-acid long DOT1L derived tracer peptides were singly labelled with tetramethylrhodamine (TAMRA) either through their amino end or side chain of their N-terminal lysine residue. An unrelated fifteen amino-acid long control peptide was singly labelled with tetramethylrhodamine (TAMRA) via its amino terminal. Peptides were analysed by HPLC and MS for quality assessment prior to fluorescence polarisation experiments. AF9 C-terminal protein (490-568) was purified as described in previous paragraph.
  • Fluorescence polarisation experiment demonstrated specific interaction between the C-terminal (490-568) domain of AF9 and peptides that mimic the AF9-binding motif of DOT1L.
  • AF9 binds DOT1L- derived peptides with affinity positively correlated to their size.
  • DOTlL-18-site 3 and DOTlL-10-site 2 have a higher binding affinity for AF9 compared to DOTlL-7-site 2 and DOTlL-7-site 3.
  • a set of probe peptides mimicking AF9-binding motifs of DOT1L and AF4 was designed to build a HTRF assay with a suitable assay window. All peptides used for the development of the HTRF assay were purchased from GenScript (Leiden, Netherlands). Probe peptides were N-terminally labelled with biotin. Peptides were analysed by HPLC and MS for quality check prior to HTRF experiments.
  • AF9 C-terminal protein (490-568) was purified as described in previous paragraph.
  • HTRF donor and acceptor were Streptavidin-Tb cryptate (CisBio, #610SATLF) and MAb Anti 6HIS-d2 (CisBio, #61HISDLF), respectively.
  • Working solutions of HTRF labelling reagents were prepared in a dedicated Terbium detection buffer (CisBio).
  • Optimal assay conditions were identified by matrix cross-titration of peptides (0 to 500 nM) and AF9 protein (0 to 250 nM), in a buffer consisting of 20 mM HEPES pH 7.5, 150 mM NaCI, 1 mM DTT and 5% glycerol. Components of reaction were mixed in low volume 384-well microplates and incubated for 15 min at room temperature. HTRF labelling reagents were added at lx and 2x concentration, as recommended by the manufacturer. Samples were incubated for lh at room temperature prior to data collection, in a total sample volume of 20 uL. All HTRF measurements were performed on a Clariostar plate reader (BMG Labtech). HTRF ratio values were calculated according to the following equation using MARS software (BMG Labtech):

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Organic Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des inhibiteurs d'AF9 et/ou d'ENL, ou de DOT1L. L'invention concerne l'utilisation d'inhibiteurs d'AF9 et/ou d'ENL pour une utilisation dans une méthode de traitement d'un individu atteint d'un cancer. En particulier, l'invention concerne une méthode de traitement d'un individu atteint d'un cancer, le cancer ayant une mutation KRAS ; et le cancer étant un cancer non hématopoïétique.
PCT/GB2020/050307 2019-02-11 2020-02-11 Procédés se rapportant à la rupture de la liaison de protéines partenaires af9 à af9 et/ou enl WO2020165570A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1901817.5A GB201901817D0 (en) 2019-02-11 2019-02-11 Methods
GB1901817.5 2019-02-11

Publications (1)

Publication Number Publication Date
WO2020165570A1 true WO2020165570A1 (fr) 2020-08-20

Family

ID=65998559

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2020/050307 WO2020165570A1 (fr) 2019-02-11 2020-02-11 Procédés se rapportant à la rupture de la liaison de protéines partenaires af9 à af9 et/ou enl

Country Status (2)

Country Link
GB (1) GB201901817D0 (fr)
WO (1) WO2020165570A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023183412A1 (fr) * 2022-03-23 2023-09-28 Baylor College Of Medicine Molécule chimère (protac) ciblant la protéolyse pour la dégradation de enl et thérapie anticancéreuse

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026930A1 (fr) * 1993-05-14 1994-11-24 Thomas Jefferson University Procedes pour depister et traiter des leucemies resultant d'anomalies chromosomiques de la region all-1
EP1716227A2 (fr) * 2004-01-27 2006-11-02 Compugen Ltd. Procede d'identification de produits genetiques putatifs par comparaison de sequences inter-especes et de sequences de biologie moleculaire exposees par celles-ci
KR20120136234A (ko) * 2011-06-08 2012-12-18 한국과학기술원 인간 히스톤 H3 라이신 79 메틸화 효소 Dot1L을 이용한 줄기세포 분화 조절 방법
WO2013116903A1 (fr) 2012-02-10 2013-08-15 Phylogica Limited Procédé de caractérisation de sites d'interaction sur des protéines cibles
WO2014100662A1 (fr) 2012-12-21 2014-06-26 Epizyme, Inc. Inhibiteurs de dot1 l à utiliser dans le traitement de la leucémie
WO2014127191A1 (fr) 2013-02-15 2014-08-21 The Regents Of The University Of Michigan Compositions et méthodes empêchant le recrutement de dot1l par les protéines hybrides mll
US20170232030A1 (en) * 2014-08-13 2017-08-17 Epizyme, Inc. Combination therapy for treating cancer

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994026930A1 (fr) * 1993-05-14 1994-11-24 Thomas Jefferson University Procedes pour depister et traiter des leucemies resultant d'anomalies chromosomiques de la region all-1
EP1716227A2 (fr) * 2004-01-27 2006-11-02 Compugen Ltd. Procede d'identification de produits genetiques putatifs par comparaison de sequences inter-especes et de sequences de biologie moleculaire exposees par celles-ci
KR20120136234A (ko) * 2011-06-08 2012-12-18 한국과학기술원 인간 히스톤 H3 라이신 79 메틸화 효소 Dot1L을 이용한 줄기세포 분화 조절 방법
WO2013116903A1 (fr) 2012-02-10 2013-08-15 Phylogica Limited Procédé de caractérisation de sites d'interaction sur des protéines cibles
WO2014100662A1 (fr) 2012-12-21 2014-06-26 Epizyme, Inc. Inhibiteurs de dot1 l à utiliser dans le traitement de la leucémie
WO2014127191A1 (fr) 2013-02-15 2014-08-21 The Regents Of The University Of Michigan Compositions et méthodes empêchant le recrutement de dot1l par les protéines hybrides mll
US20170232030A1 (en) * 2014-08-13 2017-08-17 Epizyme, Inc. Combination therapy for treating cancer

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Cancer Treatment Using Peptides: Current Therapies and Future Prospects", JOURNAL OF AMINO ACIDS, vol. 2012, pages 13
"Remington's Pharmaceutical Sciences", 2000, LIPPINCOTT, WILLIAMS & WAF9/ENLINS
DAIGLE SR ET AL., ANCER CELL, vol. 20, 2011, pages 53 - 65
DATABASE UniProt [online] 20 June 2018 (2018-06-20), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:PKY51417.1};", XP002798614, retrieved from EBI accession no. UNIPROT:A0A2I1GXQ5 Database accession no. A0A2I1GXQ5 *
DATABASE UniProt [online] 28 March 2018 (2018-03-28), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:LAA85973.1}; Flags: Fragment;", XP002798613, retrieved from EBI accession no. UNIPROT:A0A2D4IP03 Database accession no. A0A2D4IP03 *
HOU, Z. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 110, 2013, pages 15644 - 15649
JIANG ET AL., NAT. BIOTECHNOL., vol. 31, 2013, pages 233 - 239
JINEK ET AL., SCIENCE, vol. 337, 2012, pages 816 - 821
KUNTIMADDI ET AL., CELL REPORTS, vol. 11, 5 May 2015 (2015-05-05), pages 808 - 820
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
MARQUS S ET AL., JOURNAL OF BIOMEDICAL SCIENCE, vol. 24, 2017, pages 21
PRIOR ET AL., CANCER RES., vol. 72, 2012, pages 2457 - 67
QUINLANSETTLEMAN, FUTURE ONCOL, vol. 5, 2009, pages 105 - 16
SHEN ET AL., J BIOL CHEM, vol. 288, no. 42, 2013, pages 30585 - 30596
SIENA ET AL., JNCI, vol. 101, 2009, pages 1308 - 1324
XING ET AL., INT J MOL MED., vol. 38, no. 2, August 2016 (2016-08-01), pages 407 - 416
ZHAO ET AL., MOL MED REP., vol. 12, no. 1, July 2015 (2015-07-01), pages 960 - 966
ZHENG ET AL., ACTA PHARMACOL SIN., vol. 30, no. 12, December 2009 (2009-12-01), pages 1625 - 1633
ZHU ET AL., GENET MOL BIOL., vol. 35, 2012, pages 538 - 544

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023183412A1 (fr) * 2022-03-23 2023-09-28 Baylor College Of Medicine Molécule chimère (protac) ciblant la protéolyse pour la dégradation de enl et thérapie anticancéreuse

Also Published As

Publication number Publication date
GB201901817D0 (en) 2019-04-03

Similar Documents

Publication Publication Date Title
JP7219791B2 (ja) Rtk突然変異細胞を有する患者を処置するための組成物及び方法
Garnham et al. The chemical complexity of cellular microtubules: tubulin post‐translational modification enzymes and their roles in tuning microtubule functions
Kuo et al. NSD2 links dimethylation of histone H3 at lysine 36 to oncogenic programming
JP4351896B2 (ja) p38/JTV−1を有効成分とする癌治療用薬学的組成物及び癌治療用薬学的組成物のスクリーニング方法
Yan et al. Structural insights into the SPRED1-neurofibromin-KRAS complex and disruption of SPRED1-neurofibromin interaction by oncogenic EGFR
KR20120051258A (ko) Gkn 1을 함유하는 항암용 조성물
CA3025423A1 (fr) Interfaces de proteines
Woodhouse et al. The genetic basis of resistance to cancer chemotherapy
WO2020165570A1 (fr) Procédés se rapportant à la rupture de la liaison de protéines partenaires af9 à af9 et/ou enl
US9023343B2 (en) Phosphorylation-inhibiting agent or dephosphorylating agent for PTEN
ES2373986T3 (es) Actividad relacionada con ubiquitinación y/o degradación de siva y moduladores de la misma.
KR102658844B1 (ko) 펩티드 유도체 및 약학적 조성물
WO2019117188A1 (fr) Inhibiteur de la prolifération cellulaire et composition pharmaceutique pour le traitement ou la prévention du cancer comprenant l'inhibiteur de la prolifération cellulaire
US8846360B2 (en) Activation and transfer cascade for ubiquitin
EP3288963B1 (fr) Polypeptide à domaine c-terminal pink1 et leurs méthodes d'utilisation dans le traitement du cancer
EP4212171A1 (fr) Application de protéine rdr dans une thérapie antitumorale
US9243234B2 (en) Sequence-specific MRNA interferase and uses thereof
Li et al. Mutations at the dimer interface and surface residues of Nm23-H1 metastasis suppressor affect its expression and function
Baruti Genetic and Chemical Genetic Tools for Examining Context-Dependent Protein-Protein Interactions
WO2012068232A1 (fr) Effecteurs de l'aurora a kinase
US9145554B2 (en) Cancer therapy method
Sgro An investigation into the interdomain region of Caenorhabditis elegans fatty acid synthase and its implications as a drug target
US20090130760A1 (en) Method for the ubiquitination of common subunit of rna polymerases

Legal Events

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

Ref document number: 20707758

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20707758

Country of ref document: EP

Kind code of ref document: A1