WO1998048013A1 - Brca2 transcriptional activator domain and uses thereof - Google Patents

Brca2 transcriptional activator domain and uses thereof Download PDF

Info

Publication number
WO1998048013A1
WO1998048013A1 PCT/GB1998/001181 GB9801181W WO9848013A1 WO 1998048013 A1 WO1998048013 A1 WO 1998048013A1 GB 9801181 W GB9801181 W GB 9801181W WO 9848013 A1 WO9848013 A1 WO 9848013A1
Authority
WO
WIPO (PCT)
Prior art keywords
brca2
fragment
sequence
nucleic acid
variant
Prior art date
Application number
PCT/GB1998/001181
Other languages
French (fr)
Inventor
Tony Kouzarides
Original Assignee
Cancer Research Campaign Technology 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 Cancer Research Campaign Technology Limited filed Critical Cancer Research Campaign Technology Limited
Priority to CA002288335A priority Critical patent/CA2288335A1/en
Priority to EP98917445A priority patent/EP0977847A1/en
Priority to AU70674/98A priority patent/AU7067498A/en
Publication of WO1998048013A1 publication Critical patent/WO1998048013A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4705Regulators; Modulating activity stimulating, promoting or activating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention provides polypeptides, nucleic acid encoding the polypeptides, substances which interact
  • activator domain severely reduces activation of transcription. Furthermore, molecules (polypeptide domains) which interact with this region of the BRCA2 polypeptide have been identified experimentally and may be used to modulate
  • FIG. 1 shows that sequences within exon 3 of BRCA2
  • FIG. 2 shows the protein and DNA sequence of residues
  • FIG 3 shows the amino acid and DNA sequence of a portion of the protein named "BBPl” (BRCA2 Binding Protein-1) found to interact with the BRCA2 TAD and to modulate its transcriptional activation.
  • BBPl BRCA2 Binding Protein-1
  • polypeptide which has the amino acid sequence of a fragment of BRCA2 protein and which is able to
  • acids less than about 120 amino acids, less than about 100
  • amino acids or less than about 70 amino acids.
  • polypeptides according to this aspect of the present invention may include or be fused or otherwise operably linked to a DNA binding domain, which may be heterologous or
  • BRCA2 foreign to BRCA2 , e.g. being of a polypeptide such as GAL4 , or LexA or any suitable example, of which many are known and
  • inhibitory domains within the fusion protein.
  • a polypeptide according to the present invention may be any polypeptide according to the present invention.
  • Figure 2 or an amino acid sequence which is a fragment, mutant, variant, allele or derivative thereof.
  • particular embodiments of the present invention may make individual use of the two fragments of the exon 3 activation
  • fragments within the fragment of BRCA2 residues 18-105 may be
  • polypeptide may be used to identify regions and/or residues
  • polypeptide may include an amino acid sequence which differs
  • variants derivatives, alleles, mutants and homologues, e.g.
  • amino acid sequence shares homology with amino acid sequence
  • sequence or Figure 2 preferably at least about 30%, or 40%, or 50%, or 60%, or 70%, or 75%, or 80%, or 85% homology, or at least about 90% or 95% homology.
  • homology at the amino acid level is generally in terms of amino acid similarity or identity. Similarity allows for "conservative variation”, i.e.
  • valine leucine or methionine for another, or the
  • Similarity may be as defined and determined
  • GAP uses the Needleman and unsch
  • Homology may be over the full-length of the relevant polypeptide or may more preferably be over a contiguous sequence of about 15, 20, 25, 30, 40, 50 or more amino acids, compared with the relevant wild-type amino acid sequence.
  • Preferred polypeptides may include a transcriptional
  • activation domain with at least about 50%, 60%, 70%, 80%,
  • the present invention extends to
  • nucleic acid that hybridizes with any one or more of the
  • Suitable conditions include, e.g. for detection
  • the residues 18-105 fragment of BRCA2 polypeptide is referred to as the BRCA2 transcription
  • TAD activation domain
  • PAR primary activating region
  • One such embodiment includes the portion from residues 126 to 197, or a fragment, mutant, allele, variant or derivative thereof.
  • polypeptide includes or consists essentially of the fragment
  • the TAD including the PAR and the AAR is flanked by two "inhibitory regions" which are bound by molecules which inhibit activation of transcription by the TAD, as demonstrated experimentally and described below. The position of the two inhibitory regions is shown
  • fragments may be used instead of the wild-type sequence
  • a polypeptide which includes IRl and/or IR2 with the BRCA2 TAD may be used in the
  • peptides including or
  • Inhibitor domains have been shown to be present in a number of transcription factors (references 4, 5, 6 below) .
  • throughput screen may be used to find molecules which
  • BRCA2 transcriptional activation which may do this by binding the inhibitor domain and preventing an intramolecular interaction.
  • 0 may include or consist essentially of the IRl and/or IR2 sequences shown in Figure 2. Where additional amino acids are included, which amino acids may be from BRCA2 (so that the peptide is a larger fragment of BRCA2) or may be heterologous or foreign to BRCA2 , the peptide may be about
  • a peptide according to this aspect may be included within a larger fusion
  • BRCA2 i.e. heterologous or foreign sequence, such as a
  • nucleic acid in an expression system.
  • the present invention also provides in
  • BBPl BRCA2 Binding Protein 1
  • nucleic acid according to the present invention is provided as an isolate, in isolated and/or purified form, or free or substantially free of material with which it is naturally associated, such as free or
  • Nucleic acid may be wholly or
  • genomic DNA partially synthetic and may include genomic DNA, cDNA or RNA.
  • nucleic acid according to the invention includes RNA
  • samples of such nucleic acid e.g. from genomic sources, (ii) chemical synthesis, or (iii) preparing cDNA sequences.
  • Modifications to the BRCA2 sequences can be made, e.g. using site directed mutagenesis, to lead to the expression of
  • sequences can be incorporated in a vector
  • the vectors may include other sequences such as promoters or enhancers to control its expression.
  • the vectors may include other sequences such as promoters or enhancers to control its expression.
  • Polypeptide can then be obtained by
  • polypeptide is produced and recovering the polypeptide from
  • Prokaryotic and eukaryotic cells are used for this purpose in the art,
  • E. coli including strains of E. coli, yeast, and eukaryotic cells such as COS or CHO cells.
  • yeast including strains of E. coli, yeast, and eukaryotic cells such as COS or CHO cells.
  • eukaryotic cells such as COS or CHO cells.
  • the present invention also encompasses a method of making a polypeptide or peptide (as disclosed) , the method
  • polypeptide or peptide generally nucleic acid according to
  • Polypeptides and peptides may also be expressed in in vitro systems, such as reticulocyte lysate.
  • host cells include bacteria, eukaryotic cells such as mammalian and yeast, and baculovirus systems.
  • polypeptide include Chinese hamster ovary cells, HeLa cells,
  • E. coli common, preferred bacterial host is E. coli.
  • Suitable vectors can be chosen or constructed,
  • Vectors may be plasmids, viral e.g. 'phage, or phagemid, as appropriate.
  • plasmids viral e.g. 'phage, or phagemid, as appropriate.
  • nucleic acid for manipulation of nucleic acid, for example in preparation
  • nucleic acid constructs mutagenesis, sequencing,
  • the nucleic acid of the invention may be integrated into
  • the nucleic acid may be on an extra-chromosomal vector within the cell, or otherwise identifiably
  • a still further aspect provides a method which includes introducing the nucleic acid into a host cell.
  • the introduction which may (particularly for in vi tro
  • transformation may employ any available technique.
  • suitable techniques may include calcium
  • retrovirus or other virus e.g. vaccinia or, for insect cells, baculovirus.
  • retrovirus or other virus e.g. vaccinia or, for insect cells, baculovirus.
  • vaccinia e.g. vaccinia
  • insect cells e.g. vaccinia
  • baculovirus e.g. vaccinia
  • suitable techniques for bacterial cells, suitable techniques
  • sensitivity genes may be used in identifying clones
  • nucleic acid of interest containing nucleic acid of interest, as is well known in the
  • the introduction may be followed by causing or allowing
  • the encoded polypeptide (or peptide) is produced. If the
  • polypeptide is expressed coupled to an appropriate signal leader peptide it may be secreted from the cell into the
  • a polypeptide or peptide may be isolated and/or purified from the host cell and/or culture medium, as the case may be, and subsequently used as desired, e.g. in the formulation of a
  • composition which may include one or more additional ingredients
  • nucleic acid may take place in vivo by
  • a host cell containing nucleic acid according to the
  • nucleic acid into the cell or into an ancestor of the cell
  • cell or ancestor (which introduction or alteration may take place in vivo or ex vivo) , may be comprised (e.g. in the
  • soma within an organism which is an animal, particularly a
  • mammal which may be human or non-human, such as rabbit,
  • guinea pig, rat, mouse or other rodent cat, dog, pig, sheep, goat, cattle or horse, or which is a bird, such as a chicken.
  • _ sequence may allow the organism to be used as a model in
  • a polypeptide according to the present invention may be any polypeptide according to the present invention.
  • Such molecules may be useful in a therapeutic (possibly including prophylactic) context.
  • the present invention relates to screening and assay methods and means, and substances
  • the invention includes providing a polypeptide or peptide of
  • Binding may be determined by
  • Further assays are for substances which interact with or bind the TAD and/or modulate its ability to activate transcription.
  • BBPl BRCA2 Binding Protein-1
  • Assays of the invention may therefore use the BRCA2 TAD, or
  • transcriptional activation may stimulate and/or enhance
  • Jnk Jun N-terminal kinase
  • phosphorylase removes the phosphate to deactivate the transcriptional activator function.
  • a molecule that inhibits or prevents the dephosphorylation may be used to enhance transcriptional activation.
  • Such a molecule may include a
  • the BRCA2 TAD has significant sequence homology with the TAD of Jun and a kinase is able to bind it.
  • a kinase is able to bind it.
  • Phosphorylation may be determined for example by
  • Such antibodies may be
  • Phosphorylation may be determined by immobilisation of BRCA2 or a fragment, mutant, variant or
  • a suitable substrate such as a bead or
  • Another aspect of the present invention provides an assay (A) which includes :
  • polypeptide or peptide and the test substance.
  • TAD, IRl and/or IR2 may be isolated and/or purified
  • the present invention provides an
  • the present invention including a BRCA2 TAD and optionally a
  • IRl and/or IR2 and a DNA binding domain capable of binding a
  • nucleotide sequence within a promoter and a putative
  • polypeptide is capable of binding to activate transcription of a sequence operably linked to the promoter
  • a compound which binds IRl and/or IR2 may inhibit transcriptional activation by the polypeptide.
  • a molecule which binds IRl and/or IR2 in vivo e.g. a molecule naturally present in a mammalian, e.g. human, tumour
  • non-tumour cell e.g. breast tumour
  • a breast tumour e.g. breast tumour
  • non-tumour cell e.g. breast tumour
  • molecule which inhibits transcriptional activation by the BRCA2 TAD obtainable using assay (A) or assay (B) may be isolated and may be manufactured, and may be subsequently used to assay for substances which interfere with its binding _ to IRl and/or IR2 within BRCA2 polypeptide and/or ability to inhibit BRCA2 TAD transcriptional activation.
  • Such an assay may include bringing into contact a
  • peptide or polypeptide including a IRl or IR2 sequence and a
  • binding molecule for the IRl or IR2 sequence (such as obtainable by means of assay (A) or assay (B) ) in the
  • test conditions are
  • binding molecule such binding occurs, and determining binding between the IRl and/or IR2. This may be followed by
  • Fragments may be generated and used in any combination
  • fragments may be generated by taking encoding DNA, identifying suitable restriction enzyme recognition sites
  • the portion from the DNA.
  • the portion may then be operably
  • primers Small fragments (up to about 20 or 30 amino acids)
  • a polypeptide may be fused to a heterologous DNA binding
  • the GAL 4 transcription factor includes two functional domains. These domains are the DNA binding domain (GAL4DBD)
  • GAL4TAD GAL4 transcriptional activation domain
  • LexA DNA binding domain and the VP60
  • the interaction between the polypeptides may be studied in vi tro by labelling one with a
  • Suitable detectable labels include 35 S-methionine which may be incorporated into recombinantly produced peptides and
  • polypeptides may also be expressed as a fusion protein
  • the protein which is immobilized on a solid support may
  • a preferred in vi tro interaction may utilise a
  • GST glutathione-S-transferase
  • This may be immobilized on glutathione agarose beads.
  • beads may be rinsed to remove unbound protein and the amount
  • An assay according to the present invention may also take the form of an in vivo assay.
  • the in vivo assay may be performed in a cell line such as a yeast strain in which the relevant polypeptides or peptides are expressed from one or
  • a further assay according to the present invention tests
  • BRCA2 TAD activation by BRCA2 TAD, e.g. by inhibiting interaction of
  • IRl and/or IR2 with a binding molecule or respective binding
  • Such an assay may involve :
  • the present invention including a BRCA2 TAD and a IRl and/or
  • IR2 and a DNA binding domain capable of binding a nucleotide
  • IRl and/or IR2 inhibit (s) transcriptional activation of the
  • polypeptide is capable of binding to activate transcription of a sequence operably linked to the promoter when the
  • polypeptide is not bound by the IRl and/or IR2 binding
  • a reporter gene construct including a promoter which
  • Two or more binding sites (for example 3, 4 or 5)
  • nucleic acid construct may be present in the nucleic acid construct and this may
  • operably linked in the context of a sequence of interest and a promoter means joined as part of the same nucleic acid molecule, suitably positioned and oriented for transcription to be initiated from the promoter.
  • DNA operably linked to a promoter is "under transcriptional
  • the gene may be transcribed into rnRNA which may be translated into a peptide or polypeptide product which
  • reporter gene i.e. a gene
  • a reporter gene preferably encodes an enzyme which catalyses a reaction which produces a detectable signal, preferably a visually detectable signal, such as a coloured
  • ⁇ -galactosidase activity may be assayed by production of blue colour on substrate, the assay being by
  • Fluorescence for example that produced as a result of
  • luciferase activity may be quantitated using a
  • Radioactive assays may be used, for
  • the binding molecule may be labelled directly or indirectly using
  • a method of screening for ability of a substance to modulate activity of a promoter may include contacting an expression system, such as a host cell, containing assay
  • the presence of the test substance indicates ability of the test substance
  • a promoter construct may be introduced into a cell line
  • the cell line containing the reporter construct integrated into the genome may be grown and incubated with test
  • the cells may be grown in 96
  • the cells may then be washed and the reporter
  • the substance may be investigated further. Furthermore, it may be manufactured
  • compositions such as a medicament, pharmaceutical
  • composition or drug may be administered to a subject.
  • composition or drug may be administered to a subject.
  • concentrations of putative inhibitor compound may be used,
  • Compounds which may be used may be natural or synthetic
  • putative inhibitor compounds can be derived from the BRCA2
  • Peptides can also be generated wholly or partly by
  • binding regions determined to provide single chain antibodies and fragments thereof which are responsible for disrupting
  • Antibodies may be obtained using techniques which are
  • a mammal e.g. mouse, rat, rabbit, horse, goat,
  • Antibodies may be obtained from immunised animals using any of a variety of techniques known in the art, and screened,
  • producing cells from an animal may be accompanied by a step of sacrificing the animal .
  • an antibody specific for a protein may be
  • the library may be naive, that
  • fragments may be one constructed using sequences obtained from an organism which has been exposed to the
  • Antibodies according to the present invention may be any suitable antibodies according to the present invention.
  • invention covers antibody fragments, derivatives, functional equivalents and homologues of antibodies, including synthetic
  • Example antibody fragments, capable of binding an antigen or other binding partner are the Fab fragment consisting of the VL, VH, Cl and CHI domains; the Fd fragment consisting of the VH and CHI domains; the Fv fragment consisting of the NL and VH domains of a single arm of an antigen or other binding partner.
  • the dAb fragment which consists of a VH domain
  • the present invention may be subject to genetic mutation or
  • a monoclonal antibody can be any monoclonal antibody.
  • Such techniques may involve introducing D ⁇ A encoding the immunoglobulin variable region, or the complementarity determining regions
  • binding characteristics are within the scope of the present invention, as are host cells, eukaryotic or prokaryotic, containing nucleic acid encoding antibodies (including
  • the invention also provides methods of production of the
  • antibodies including growing a cell capable of producing the
  • the reactivities of antibodies on a sample e.g. the
  • the reporter molecules may be any suitable reporter molecules.
  • the reporter molecules may be any suitable reporter molecules.
  • reporter molecules may be any linkage of reporter molecules.
  • Linkage via a peptide bond may be as
  • Suitable fluorochromes include fluorescein, rhodamine, phycoerythrin and Texas Red.
  • Suitable chromogenic dyes include diaminobenzidine .
  • Other reporters include macromolecular colloidal
  • - particles or particulate material such as latex beads that are coloured, magnetic or paramagnetic, and biologically or
  • molecules may be enzymes which catalyse reactions that
  • properties for example. They may be molecularly excitable,
  • Biosensors Biotin/avidin or biotin/streptavidin and
  • alkaline phosphatase detection systems may be employed.
  • the mode of determining binding is not a feature of the present invention and those skilled in the art are able to
  • Antibodies in accordance with the present invention may be used in screening for the presence of a particular polypeptide, for example in a test sample containing cells or
  • cell lysate as discussed, such as a BRCA2 polypeptide including a mutation in the TAD, IRl and/or IR2, where such _ mutation is reflected in an alteration in one or more
  • Antibodies may also be used in purifying and/or
  • an individual may be useful in a therapeutic context (which
  • prophylaxis may include prophylaxis
  • An antibody may be provided in a kit, which may include instructions for use of the antibody, e.g. in determining the
  • One or more other reagents may be included, such as labelling
  • Reagents may be provided within containers which protect them from the external environment , such as a sealed vial .
  • candidate inhibitor compounds may be based on modelling the 3 -dimensional structure of a polypeptide or
  • the invention provides compounds
  • the assay of the invention when conducted in vivo,
  • composition to a patient, e.g. for anti-tumour or other anti-
  • proliferative treatment which may include preventative
  • composition including admixing such a
  • substance with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients.
  • BRCA2 polypeptide TAD IRl or IR2
  • binding ligand or ligands for IRl and/or IR2 which interfere with interaction between these polypeptides, and/or transcriptional activation by BRCA2 TAD.
  • functional mimetic means a substance which may not contain an active portion of the
  • a substance identified using the present invention may be any substance identified using the present invention.
  • Non-peptide small
  • peptides are not well suited as active agents for oral compositions as they tend to be 0 quickly degraded by proteases in the alimentary canal.
  • Mimetic design, synthesis and testing may be used to avoid randomly screening large number of molecules for a target property. There are several steps commonly taken in the design of
  • a range of sources e.g. spectroscopic techniques, X-ray
  • mapping (which models the charge and/or volume of a
  • a template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted.
  • the mimetic is easy to synthesise, is likely to be pharmacologically acceptable
  • administration is preferably in a "prophylactically effective
  • a composition may be administered alone or in
  • compositions according to the present invention are provided.
  • inventions and for use in accordance with the present invention, may include, in addition to active ingredient, a
  • injection e.g. cutaneous, subcutaneous or intravenous.
  • compositions for oral administration may include
  • a tablet may include a solid carrier such as gelatin or an adjuvant.
  • Liquid pharmaceutical compositions generally include a liquid
  • Physiological saline solution dextrose or other saccharide solution or glycols such as ethylene glycol , propylene glycol or polyethylene glycol may be included.
  • isotonic vehicles such as Sodium Chloride Injection
  • peptide or other substance as disclosed herein may be provided in a kit, e.g. sealed in a suitable container which
  • kit may include instructions for use.
  • IR2 may be used in diagnostic and prognostic screening.
  • Oligonucleotide probes or primers as well as the full- length BRCA2 TAD (optionally including IRl and/or IR2)
  • nucleic acid useful in screening a test sample containing nucleic acid for
  • the probes include cancers, the probes
  • hybridisation can be controlled to minimise non-specific
  • binding and preferably stringent to moderately stringent
  • Nucleic acid isolated and/or purified from one or more cells e.g. human
  • a nucleic acid library derived from nucleic acid isolated and/or purified from cells e.g. a cDNA library derived from mRNA isolated from the cells
  • PCR polymerase chain reaction
  • a method may include hybridisation of one or more (e.g.
  • nucleic acid is double-stranded DNA, hybridisation will be
  • the hybridisation may be as part of a PCR
  • PCR An example procedure would be a combination of PCR and
  • binding of a probe to target nucleic acid may be any suitable nucleic acid (e.g. DNA).
  • target nucleic acid e.g. DNA
  • probes may be radioactively, fluorescently or enzymatically labelled. Other methods not employing labelling of probe
  • Probing may employ the standard Southern blotting technique . For instance DNA may be extracted from cells and
  • fragments may then be separated by electrophoresis on an
  • Labelled probe may be hybridised to
  • RNA preparations from cells may be prepared from RNA preparations from cells.
  • the target sequence is known to allow suitable forward and reverse oligonucleotide primers to be designed to be identical or similar to the polynucleotide sequence that is
  • PCR includes steps of denaturation of template nucleic acid (if double-stranded) ,
  • the nucleic acid probed or used as template in the amplification reaction may be genomic DNA, cDNA or RNA.
  • PCR can be used to amplify specific sequences from genomic DNA, specific RNA
  • oligonucleotide probes or primers may be designed, taking
  • nucleic acid is derived.
  • nucleic acid amplification may have about 10 or fewer codons
  • primers for use processes such as PCR.
  • a further aspect of the present invention provides an oligonucleotide or polynucleotide fragment of the nucleotide sequence shown for the BRCA2 TAD, IRl and IR2 regions in
  • nucleotides but preferably without abolition of ability to hybridise selectively with nucleic acid with the sequence
  • oligonucleotides In some preferred embodiments, oligonucleotides
  • disorder of cell proliferation are at least about 10
  • nucleotides in length more preferably at least about 15 nucleotides in length, more preferably at least about 20
  • Fragments and other oligonucleotides may be used as primers or probes as discussed but may also be generated (e.g. by
  • PCR in methods concerned with determining the presence in a test sample of a sequence indicative of susceptibility to cancer or other disorder of cell -cycle regulation.
  • susceptibility in the coding sequence shown in Figure 2 may be about 20 nucleotides in length, or may include a
  • oligonucleotide sequence may be designed so that it
  • Figure 2 thus include :
  • oligonucleotides will anneal without mismatch to
  • TAT at codon 42 is mutated to TGT.
  • wild-type sequence with mismatch include 5 ' -TGTAATTCTGAACCTGCAGA-3 '
  • oligonucleotides may be employed for any mutation or other sequence alteration within a BRCA2 fragment according to the present invention.
  • fragments include: 5'-ATGCCTATTGGATCCAAAGA-3 '
  • oligonucleotides may be used in various ways. These and other oligonucleotides may be used in various ways.
  • the methods may be used to detect alleles
  • cancer or other proliferative disorder in the future, e.g.
  • the methods divide into those screening for the
  • nucleic acid sequences or polypeptide are suspected of contain the nucleic acid sequences or polypeptide.
  • biological samples include blood,
  • Exemplary approaches for detecting nucleic acid or polypeptides include:
  • IRl and/or IR2 IRl and/or IR2 ) , and/or is mutated in such a region; or,
  • nucleic acid sequence either a normal sequence or a nucleic acid sequence
  • binding member including nucleic acid hybridisable with the
  • a "specific binding pair” includes a specific binding
  • binding partner (bm) and a binding partner (bp) which have a particular specificity for each other and which in normal
  • binding member and the binding partner include a part of a
  • the specific binding pair are nucleic acid sequences, they will be of a length to hybridise to each other under the conditions of the assay, preferably greater than 10 nucleotides long, more preferably greater than 15 or 20 nucleotides long. In most embodiments for screening for susceptibility
  • analyte as compared to other sequences present in the sample .
  • This initial step may be avoided by using highly sensitive array techniques that are becoming increasingly important in
  • the gene may be for diagnosing cancer of a patient with the disease as being associated with the gene.
  • a variant form of the gene may contain one or more
  • nucleic acid level are not necessarily reflected by a

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Exon 3 of BRCA2 encodes a transcriptional activator domain ('TAD'), including a primary activating region ('PAR') and an auxiliary activating region ('AAR'), flanked by two inhibitory regions ('IR1' and 'IR2') which are bound by molecules which inhibit activation of transcription by the TAD. Appropriate fragments of BRCA2 polypeptide are useful in activating transcription and in assays for substances able to modulate transcriptional activation by BRCA2.

Description

BRCA2 TRANSCRIPTIONAL ACTIVATOR
DOMAIN AND USES THEREOF
The present invention provides polypeptides, nucleic acid encoding the polypeptides, substances which interact
with the polypeptides, oligonucleotide probes and primers,
and various methods and uses thereof. In particular it
relates to transcriptional activator polypeptides and
substances which modulate their activity. It is founded on
the surprising discovery, supported by clear experimental data, that portions of the polypeptide encoded by the tumour
suppressor gene, BRCA2 (Tavtigian, S.V., Simard, J. , Rommens,
J., Couch, F. Shattuck-Eidens, D., et al . (1996), Nat. Genet.
12, 333-337), have transcriptional activation capacity, and
that these portions are flanked in full-length polypeptide by
regions which interact with inhibitor molecules which inhibit
transcriptional activation. Experimental evidence is also
provided showing that a mutation that is associated with
familial breast cancer and is within the transcriptional
activator domain severely reduces activation of transcription. Furthermore, molecules (polypeptide domains) which interact with this region of the BRCA2 polypeptide have been identified experimentally and may be used to modulate
transcriptional activation. Figure 1 shows that sequences within exon 3 of BRCA2
show sequence similarity to the activation domain of c-jun.
Below the alignment is the exon structure of the BRCA2
protein. Various portions of BRCA2 are shown below, along
with relative figures for transcription activation when fused
to the GA 4 DNA binding domain (1-147) , compared with the
activity of the GAL4 DNA binding domain alone. The results
represent an average of several independent experiments, of which details are provided below.
Figure 2 shows the protein and DNA sequence of residues
1-197 of BRCA2 , including the various domains and regions
employed in aspects and embodiments of the present invention.
A mutation of tyr to cys at residue 42 is indicated, such
mutation being associated with familial breast cancers (3) .
Nordling et al . (10) have also reported (after the priority
date of the present invention) that a large deletion which
disrupts the exon 3 transcription activator domain of BRCA2 is the disease-causing mutation in a Swedish breast/ovarian
cancer family.
Figure 3 shows the amino acid and DNA sequence of a portion of the protein named "BBPl" (BRCA2 Binding Protein-1) found to interact with the BRCA2 TAD and to modulate its transcriptional activation. Figure 4 shows a portion of the BBPl protein and
encoding DNA expressed and demonstrated to interact with
BRCA2 TAD, modulating its transcriptional activation.
According to a first aspect of the present invention
there is provided a polypeptide which has the amino acid sequence of a fragment of BRCA2 protein and which is able to
act as transcriptional activator. Further aspects provide
use of such a polypeptide in activating transcription and
methods of activating transcription which employ such a
polypeptide.
A fragment of BRCA2 according to the present invention
may in some embodiments have less than about 300 amino acids,
less than about 200 amino acids, less than about 150 amino
acids, less than about 120 amino acids, less than about 100
amino acids, or less than about 70 amino acids.
Generally in order for a polypeptide to have
transcriptional activator function requires a DNA binding domain which recognises a site within a promoter sequence. Thus, polypeptides according to this aspect of the present invention may include or be fused or otherwise operably linked to a DNA binding domain, which may be heterologous or
foreign to BRCA2 , e.g. being of a polypeptide such as GAL4 , or LexA or any suitable example, of which many are known and
in standard use in the art. See e.g. "Gene regulation" by
David Latchman, published by Unwin Hyman Ltd (1990) .
There are many examples of transcription activation
domains being fused to DNA binding domains of polypeptides
such as GAL4 , in order to study and/or manipulate transcriptional activation. For instance, Martin et al .
(Nature (1995) 375: 691-694) fused activation domain portions
of E2F1 to the DNA binding domain of GAL4 , to study interaction with MDM2 , while Brown et al . { The EMBO J. (1995)
14 (1) : 124-131) fused portions of c-Fos protein to the DNA
binding domain of GAL4, to investigate transcriptional
activation and particularly silencing of such activation on
inclusion of inhibitory domains within the fusion protein.
A polypeptide according to the present invention may
include or consist essentially of amino acids 18-105 or 23-
105 of the human BRCA2 polypeptide (residue 23 is the
boundary of exon 3 ) , the sequences of which are shown in
Figure 2, or an amino acid sequence which is a fragment, mutant, variant, allele or derivative thereof. For instance, particular embodiments of the present invention may make individual use of the two fragments of the exon 3 activation
domain demonstrated experimentally as described below to have the ability to activate transcription, i.e. the fragment which is amino acids 18 to 60 (shown to be a primary
activating region) and the fragment which is amino acids 60
to 105 (shown to be an auxiliary activating region) . Other
fragments within the fragment of BRCA2 residues 18-105 may be
employed as transcription activators provided they retain the
ability to so function. Alanine scanning and other
techniques of systematic alteration and/or fragmentation of a
polypeptide may be used to identify regions and/or residues
functionally involved or required.
Instead of using a wild-type BRCA2 fragment, the
polypeptide may include an amino acid sequence which differs
by one or more amino acid residues from the wild-type amino acid sequence, by one or more of addition, insertion,
deletion and substitution of one or more amino acids. Thus,
variants, derivatives, alleles, mutants and homologues, e.g.
from other organisms, are included.
Preferably, the amino acid sequence shares homology with
the sequence or Figure 2, preferably at least about 30%, or 40%, or 50%, or 60%, or 70%, or 75%, or 80%, or 85% homology, or at least about 90% or 95% homology.
As is well-understood, homology at the amino acid level is generally in terms of amino acid similarity or identity. Similarity allows for "conservative variation", i.e.
substitution of one hydrophobic residue such as isoleucine,
valine, leucine or methionine for another, or the
substitution of one polar residue for another, such as
arginine for lysine, glutamic for aspartic acid, or glutamine
for asparagine. Similarity may be as defined and determined
by the TBLASTN program, of Altschul et al . (1990) J". Mol .
Biol . 215: 403-10, which is in standard use in the art, or more preferably using the algorithm GAP (Genetics Computer
Group, Madison, I) . GAP uses the Needleman and unsch
algorithm to align two complete sequences that maximizes the
number of matches and minimizes the number of gaps.
Generally, the default parameters are used, with a gap
creation penalty = 12 and gap extension penalty = 4. Use of
either of the terms "homology" and "homologous" herein does
not imply any necessary evolutionary relationship between compared sequences, in keeping for example with standard use
of terms such as "homologous recombination" which merely
requires that two nucleotide sequences are sufficiently
similar to recombine under the appropriate conditions. Homology may be over the full-length of the relevant polypeptide or may more preferably be over a contiguous sequence of about 15, 20, 25, 30, 40, 50 or more amino acids, compared with the relevant wild-type amino acid sequence.
Preferred polypeptides may include a transcriptional
activation domain with at least about 50%, 60%, 70%, 80%,
85%, 90% or 95% identity with that of BRCA2 exon 3, as shown
in Figure 2, or the fragment which is amino acids 18 to 60,
or the fragment which is amino acids 60 to 105.
At the nucleic acid level sequence identity may be assessed by means of hybridization of molecules under
stringent conditions. The present invention extends to
nucleic acid that hybridizes with any one or more of the
specific sequences disclosed herein under stringent
conditions. Suitable conditions include, e.g. for detection
of sequences that are about 80-90% identical, hybridization
overnight at 42 °C in 0.25M Na2HP04, pH 7.2, 6.5% SDS, 10%
dextran sulfate and a final wash at 55°C in 0. IX SSC, 0.1%
SDS. For detection of sequences that are greater than about
90% identical, suitable conditions include hybridization
overnight at 65°C in 0.25M Na2HP04, pH 7.2 , 6.5% SDS, 10% dextran sulfate and a final wash at 60°C in 0. IX SSC, 0.1% SDS.
For convenience herein, the residues 18-105 fragment of BRCA2 polypeptide is referred to as the BRCA2 transcription
activation domain ("TAD"), the residues 18-60 fragment as the BRCA2 primary activating region or "PAR", and the residues
_60-105 as the BRCA2 auxiliary activating region or "AAR",
bearing in mind that mutants, alleles, variants, derivatives
and smaller fragments may be employed in the present
invention and are therefore generally encompassed by use of
such terms in the description unless e.g. context requires
otherwise .
One or more additional portions of the BRCA2 polypeptide
may be included, if desired. One such embodiment includes the portion from residues 126 to 197, or a fragment, mutant, allele, variant or derivative thereof. In further
embodiments of various aspects of the present invention, a
polypeptide includes or consists essentially of the fragment
of BRCA2 at residues 126 to 197, which may be used for instance in looking for and/or obtaining substances which
interact with it and which may have an effect on a BRCA2
function.
Further experimental evidence included below indicates
that within the BRCA2 polypeptide the transcription activation domain of the invention, the TAD including the PAR and the AAR, is flanked by two "inhibitory regions" which are bound by molecules which inhibit activation of transcription by the TAD, as demonstrated experimentally and described below. The position of the two inhibitory regions is shown
_ in Figure 1 ("IRl" and "IR2"), with sequence information
being given in Figure 2.
Further polypeptides according to the present invention
include IRl and/or IR2 in addition to the TAD. Of course,
mutant, allele, derivative or variant sequences, or
fragments, may be used instead of the wild-type sequence
shown. As discussed below, a polypeptide which includes IRl and/or IR2 with the BRCA2 TAD may be used in the
identification and isolation of molecules which interact with
or bind the inhibitory regions to inhibit transcriptional
activation by the TAD, and substances which interfere with
such interaction or binding and/or inhibition of
transcriptional activation. Also, peptides including or
consisting essentially of all or part of the IRl and IR2
sequences may be used similarly in the identification and
isolation of molecules which interact or bind, preferably which inhibit transcriptional activation by the TAD upon interaction or binding, and in the identification and
isolation of molecules which interfere with such interaction or binding to modulate inhibition of activation of transcription by the TAD.
Inhibitor domains have been shown to be present in a number of transcription factors (references 4, 5, 6 below) .
_ In c-Fos and others there is evidence that the inhibitor is
functioning by binding a protein or proteins to mediate
repression of transcriptional activation. This evidence
comes from "squelching" experiments in which the inhibitor
domain was added in excess to compete away any potential
repressor protein (4) . Similar experiments may be performed
using polypeptides and peptides of the present invention.
Addition of BRCA2 IRl and/or IR2 peptides in excess may
increase the activation capacity of inactive BRCA2 fragment residues 1-125. There are other instances where an inhibitor domain functions via intra-molecular interaction which
"masks" the activation domain. As discussed more fully
below, assay methods and means, for example using a high
throughput screen, may be used to find molecules which
activate BRCA2 transcriptional activation, which may do this by binding the inhibitor domain and preventing an intramolecular interaction.
A peptide according to a further aspect of the invention
0 may include or consist essentially of the IRl and/or IR2 sequences shown in Figure 2. Where additional amino acids are included, which amino acids may be from BRCA2 (so that the peptide is a larger fragment of BRCA2) or may be heterologous or foreign to BRCA2 , the peptide may be about
-20, 25, 30 or 35 amino acids in length. A peptide according to this aspect may be included within a larger fusion
protein, particularly where the peptide is fused to a non-
BRCA2 (i.e. heterologous or foreign) sequence, such as a
polypeptide or protein domain.
Further similar aspects relate in the same or similar
terms to peptides including or consisting essentially of
fragments of BBPl (see below) .
Various screening and assay formats employing polypeptides and peptides according to the present invention
are discussed below.
A convenient way of producing a polypeptide or peptide
according to the present invention is to express nucleic acid
encoding it, by use of nucleic acid in an expression system.
Accordingly the present invention also provides in
various aspects nucleic acid encoding the polypeptides and
peptides of the invention. (This includes "BBPl" (BRCA2 Binding Protein 1) discussed further below.)
Generally, nucleic acid according to the present invention is provided as an isolate, in isolated and/or purified form, or free or substantially free of material with which it is naturally associated, such as free or
_ substantially free of nucleic acid flanking the gene in the
human genome, except possibly one or more regulatory
sequence (s) for expression. Nucleic acid may be wholly or
partially synthetic and may include genomic DNA, cDNA or RNA.
Where nucleic acid according to the invention includes RNA,
reference to the sequence shown should be construed as
encompassing the RNA equivalent, with U substituted for T.
Nucleic acid sequences encoding a polypeptide in
accordance with the present invention can be readily prepared by the skilled person using the information and references contained herein and techniques known in the art (for
example, see Sambrook, Fritsch and Maniatis, "Molecular
Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory
Press, 1989, and Ausubel et al , Short Protocols in Molecular
Biology, John Wiley and Sons, 1992) , given the nucleic acid
sequence and clones available (e.g. Tavtigian, S.V., Simard,
J. , Rommens, J. , Couch, F. Shattuck-Eidens, D., et al .
(1996), Nat. Genet. 12, 333-337). These techniques include (i) the use of the polymerase chain reaction (PCR) to amplify
samples of such nucleic acid, e.g. from genomic sources, (ii) chemical synthesis, or (iii) preparing cDNA sequences. Modifications to the BRCA2 sequences can be made, e.g. using site directed mutagenesis, to lead to the expression of
_ modified BRCA2 polypeptide or to take account of codon preference in the host cells used to express the nucleic
acid. In order to obtain expression of the nucleic acid
sequences, the sequences can be incorporated in a vector
having one or more control sequences operably linked to the
nucleic acid to control its expression. The vectors may include other sequences such as promoters or enhancers to
drive the expression of the inserted nucleic acid, nucleic
acid sequences so that the polypeptide or peptide is produced
as a fusion and/or nucleic acid encoding secretion signals so that the polypeptide produced in the host cell is secreted from the cell. Polypeptide can then be obtained by
transforming the vectors into host cells in which the vector
is functional, culturing the host cells so that the
polypeptide is produced and recovering the polypeptide from
the host cells or the surrounding medium. Prokaryotic and eukaryotic cells are used for this purpose in the art,
including strains of E. coli, yeast, and eukaryotic cells such as COS or CHO cells.
Thus, the present invention also encompasses a method of making a polypeptide or peptide (as disclosed) , the method
-including expression from nucleic acid encoding the
polypeptide or peptide (generally nucleic acid according to
the invention) . This may conveniently be achieved by growing
a host cell in culture, containing such a vector, under
appropriate conditions which cause or allow expression of the
polypeptide. Polypeptides and peptides may also be expressed in in vitro systems, such as reticulocyte lysate.
Systems for cloning and expression of a polypeptide in a variety of different host cells are well known. Suitable
host cells include bacteria, eukaryotic cells such as mammalian and yeast, and baculovirus systems. Mammalian cell
lines "available in the art for expression of a heterologous
polypeptide include Chinese hamster ovary cells, HeLa cells,
baby hamster kidney cells, COS cells and many others. A
common, preferred bacterial host is E. coli.
Suitable vectors can be chosen or constructed,
containing appropriate regulatory sequences, including
promoter sequences, terminator fragments, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate. Vectors may be plasmids, viral e.g. 'phage, or phagemid, as appropriate. For further details see, for example, Molecular Cloning: a Laboratory Manual: 2nd edition, Sambrook et al . , 1989, Cold Spring
JHarbor Laboratory Press. Many known techniques and protocols
for manipulation of nucleic acid, for example in preparation
of nucleic acid constructs, mutagenesis, sequencing,
introduction of DNA into cells and gene expression, and
analysis of proteins, are described in detail in Current
Protocols in Molecular Biology, Ausubel et al . eds . , John
Wiley & Sons, 1992.
Thus, a further aspect of the present invention provides
a host cell containing heterologous nucleic acid as disclosed
herein.
The nucleic acid of the invention may be integrated into
the genome (e.g. chromosome) of the host cell. Integration
may be promoted by inclusion of sequences which promote
recombination with the genome, in accordance with standard
techniques. The nucleic acid may be on an extra-chromosomal vector within the cell, or otherwise identifiably
heterologous or foreign to the cell.
A still further aspect provides a method which includes introducing the nucleic acid into a host cell. The introduction, which may (particularly for in vi tro
introduction) be generally referred to without limitation as "transformation", may employ any available technique. For eukaryotic cells, suitable techniques may include calcium
phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using
retrovirus or other virus, e.g. vaccinia or, for insect cells, baculovirus. For bacterial cells, suitable techniques
may include calcium chloride transformation, electroporation
and transfection using bacteriophage . As an alternative,
direct injection of the nucleic acid could be employed.
Marker genes such as antibiotic resistance or
sensitivity genes may be used in identifying clones
containing nucleic acid of interest, as is well known in the
art .
The introduction may be followed by causing or allowing
expression from the nucleic acid, e.g. by culturing host
cells (which may include cells actually transformed although
more likely the cells will be descendants of the transformed
cells) under conditions for expression of the gene, so that
the encoded polypeptide (or peptide) is produced. If the
polypeptide is expressed coupled to an appropriate signal leader peptide it may be secreted from the cell into the
culture medium. Following production by expression, a polypeptide or peptide may be isolated and/or purified from the host cell and/or culture medium, as the case may be, and subsequently used as desired, e.g. in the formulation of a
composition which may include one or more additional
components, such as a pharmaceutical composition which
includes one or more pharmaceutically acceptable excipients,
vehicles or carriers (e.g. see below).
Introduction of nucleic acid may take place in vivo by
way of gene therapy, as discussed below.
A host cell containing nucleic acid according to the
present invention, e.g. as a result of introduction of the
nucleic acid into the cell or into an ancestor of the cell
and/or genetic alteration of the sequence endogenous to the
cell or ancestor (which introduction or alteration may take place in vivo or ex vivo) , may be comprised (e.g. in the
soma) within an organism which is an animal, particularly a
mammal, which may be human or non-human, such as rabbit,
guinea pig, rat, mouse or other rodent, cat, dog, pig, sheep, goat, cattle or horse, or which is a bird, such as a chicken.
Genetically modified or transgenic animals or birds
comprising such a cell are also provided as further aspects of the present invention.
This may have a therapeutic aim. (Gene therapy is discussed below.) Also, the presence of a mutant, allele, derivative or variant sequence within cells of an organism, particularly when in place of a homologous endogenous
_ sequence, may allow the organism to be used as a model in
testing and/or studying substances which modulate activity of
the encoded polypeptide in vitro or are otherwise indicated
to be of therapeutic potential. Conveniently, however,
assays for such substances may be carried out in vi tro,
within host cells or in cell-free systems.
A polypeptide according to the present invention may be
used in screening for molecules which affect or modulate its activity or function. Such molecules may be useful in a therapeutic (possibly including prophylactic) context.
Combinatorial library technology provides an efficient way of
testing a potentially vast number of different substances for
ability to modulate bind to and/or activity of a polypeptide. Such libraries and their use are known in the art. The use
of peptide libraries may be preferred in certain
circumstances .
In various further aspects the present invention relates to screening and assay methods and means, and substances
identified thereby.
Thus, further aspects of the present invention provide the use of a polypeptide or peptide of the invention as
_disclosed, and/or encoding nucleic acid therefor, in
screening or searching for and/or obtaining/identifying a
substance, e.g. peptide or chemical compound, which interacts
and/or binds with the polypeptide or peptide and/or
interferes with its function or activity or that of another
substance, e.g. polypeptide or peptide, which interacts
and/or binds with the polypeptide or peptide of the
invention. For instance, a method according to one aspect of
the invention includes providing a polypeptide or peptide of
the invention and bringing it into contact with a substance,
which contact may result in binding between the polypeptide
or peptide and the substance. Binding may be determined by
any of a number of techniques available in the art, both qualitative and quantitative.
In various aspects the present invention is concerned
with provision of assays for substances which inhibit
interaction between the BRCA2 IRl and IR2 peptide motifs
(discussed above) and the inhibitory molecule or molecules which bind these regions to inhibit transcriptional activation by the TAD.
Further assays are for substances which interact with or bind the TAD and/or modulate its ability to activate transcription.
A protein termed "BBPl" (BRCA2 Binding Protein-1) has
been found to interact with the BRCA2 TAD, specifically the
PAR. A domain of BBPl was expressed (see experimental
section) and shown to repress activity of the BRCA2 TAD.
Assays of the invention may therefore use the BRCA2 TAD, or
suitable fragment thereof, and BBPl, or suitable fragment
thereof, in obtaining substances able to interfere with their
interaction and/or modulate the effect of BBPl on BRCA2
transcription activation.
As noted, the IRl and IR2 peptides of the invention and
polypeptides including one or more of these may be used in
screening for and obtaining the inhibitory molecules which interact or bind IRl and/or IR2 within BRCA2 in vivo to
inhibit transcriptional activation.
A substance which binds the TAD may inhibit
transcriptional activation, or may stimulate and/or enhance
it. For instance, it is known that c-Jun transcriptional activator function is stimulated by phosphorylation of the
active site by a kinase (Jun N-terminal kinase, Jnk) . A
phosphorylase removes the phosphate to deactivate the transcriptional activator function. A molecule that inhibits or prevents the dephosphorylation may be used to enhance transcriptional activation. Such a molecule may include a
binding portion of the kinase. As discussed elsewhere
herein, the BRCA2 TAD has significant sequence homology with the TAD of Jun and a kinase is able to bind it. Thus, a
kinase which operates on the BRCA2 TAD, or a BRCA2 TAD
binding portion of the kinase, has been obtained and used to
modulate transcriptional activation.
Phosphorylation may be determined for example by
immobilising a BRCA2 fragment, mutant, variant or derivative
thereof, e.g. on a bead or plate, and detecting
phosphorylation using an antibody or other binding molecule
which binds the relevant site of phosphorylation with a
different affinity when the site is phosphorylated from when the site is not phosphorylated. Such antibodies may be
obtained by means of any standard technique as discussed
elsewhere herein, e.g. using a phosphorylated peptide (such
as a fragment of BRCA2) . Binding of a binding molecule which
discriminates between the phosphorylated and non- phosphorylated form of BRCA2 or relevant fragment, mutant,
variant or derivative thereof may be assessed using any
technique available to those skilled in the art, which may involve determination of the presence of a suitable label, such as fluorescence. Phosphorylation may be determined by immobilisation of BRCA2 or a fragment, mutant, variant or
derivative thereof, on a suitable substrate such as a bead or
plate, wherein the substrate is impregnated with scintillant,
such as in a standard scintillation proximetry assay, with phosphorylation being determined via measurement of the
incorporation of radioactive phosphate. Phosphate
incorporation into BRCA2 or a fragment, mutant, variant or
derivative thereof, may be determined by precipitation with
acid, such as trichloroacetic acid, and collection of the
precipitate on a suitable material such as nitrocellulose
filter paper, followed by measurement of incorporation of radiolabeled phosphate. SDS-PAGE separation of substrate may
be employed followed by detection of radiolabel .
As described below, the present inventors have obtained in their experimental work substances, in particular
polypeptide domains, which interact with BRCA2 polypeptide
fragments in accordance with the present invention.
Another aspect of the present invention provides an assay (A) which includes :
(a) bringing into contact a polypeptide or peptide according to the invention including BRCA2 IRl and/or IR2 as disclosed, and a putative binding molecule or other test
substance; and
(b) determining interaction or binding between the
polypeptide or peptide and the test substance.
A substance which interacts with or binds to the BRCA2
TAD, IRl and/or IR2 may be isolated and/or purified,
manufactured and/or used to modulate transcriptional
activation as discussed.
In another aspect, the present invention provides an
assay (B) which includes:
(a) bringing into contact a polypeptide according to
the present invention, including a BRCA2 TAD and optionally a
IRl and/or IR2 and a DNA binding domain capable of binding a
nucleotide sequence within a promoter, and a putative
inhibitor compound under conditions where the polypeptide, in
the absence of inhibitor, is capable of binding the
nucleotide sequence within the promoter to activate
transcription;
(b) providing a nucleic acid molecule which includes a
promoter which includes the nucleotide sequence to which the
polypeptide is capable of binding to activate transcription of a sequence operably linked to the promoter; and
(c) measuring the degree of modulation or alteration of transcriptional activation caused by said inhibitor compound.
A compound which increases the level of transcriptional
activation, particularly, when IRl and IR2 are not included
may be useful in enhancing transcriptional activation by
BRCA2 TAD, which has therapeutic potential given BRCA2 ' s role
as a tumour suppressor and the experimental evidence
described below on the effect of a mutation within the BRCA2 TAD which is associated with familial breast cancer on
decreasing BRCA2 TAD transcriptional activation.
A compound which binds IRl and/or IR2 may inhibit transcriptional activation by the polypeptide. Thus,
inhibition of transcriptional activation allows for
identification of molecules which bind IRl and/or IR2 ,
including a natural ligand.
A molecule which binds IRl and/or IR2 in vivo, e.g. a molecule naturally present in a mammalian, e.g. human, tumour
(e.g. breast tumour) or non-tumour cell, and preferably a
molecule which inhibits transcriptional activation by the BRCA2 TAD, obtainable using assay (A) or assay (B) may be isolated and may be manufactured, and may be subsequently used to assay for substances which interfere with its binding _ to IRl and/or IR2 within BRCA2 polypeptide and/or ability to inhibit BRCA2 TAD transcriptional activation.
Thus, a further aspect of the invention employs a
peptide or polypeptide which includes BRCA2 IRl and/or IR2
(bearing in mind these terms always, unless context requires
otherwise, allow for the use of a mutant, variant,
derivative, allele or fragment) and a molecule obtainable by
assay (A) or assay (B) which binds IRl or IR2 in an assay for
substances which interfere with such binding and may
therefore inhibit inhibition of the transcriptional activation capacity of the BRCA2 TAD.
Such an assay may include bringing into contact a
peptide or polypeptide including a IRl or IR2 sequence and a
binding molecule for the IRl or IR2 sequence (such as obtainable by means of assay (A) or assay (B) ) in the
presence of a test substance, wherein the test conditions are
such that in the absence of a substance able to interfere
with binding between the IRl and/or IR2 sequence and the
binding molecule such binding occurs, and determining binding between the IRl and/or IR2. This may be followed by
isolation and/or manufacture and/or use of a substance which tests positive for ability to interefere with the binding of interest .
It is not necessary to use the entire proteins for
assays of the invention which test for binding between two
molecules. Fragments may be generated and used in any
suitable way known to those of skill in the art. Suitable
ways of generating fragments include, but are not limited to,
recombinant expression of a fragment from encoding DNA. Such
fragments may be generated by taking encoding DNA, identifying suitable restriction enzyme recognition sites
either side of the portion to be expressed, and cutting out
said portion from the DNA. The portion may then be operably
linked to a suitable promoter in a standard commercially available expression system. Another recombinant approach is
to amplify the relevant portion of the DNA with suitable PCR
primers. Small fragments (up to about 20 or 30 amino acids)
may also be generated using peptide synthesis methods which are well known in the art .
For example, in a preferred embodiment of the invention a polypeptide may be fused to a heterologous DNA binding
domain such as that of the yeast transcription factor GAL 4. The GAL 4 transcription factor includes two functional domains. These domains are the DNA binding domain (GAL4DBD)
_ and the GAL4 transcriptional activation domain (GAL4TAD) . By fusing one polypeptide or peptide to one of those domains and
another polypeptide or peptide to the respective counterpart,
a functional GAL 4 transcription factor is restored only when
two polypeptides or peptides of interest interact. Thus,
interaction of the polypeptides or peptides may be measured
by the use of a reporter gene probably linked to a GAL 4 DNA
binding site which is capable of activating transcription of
said reporter gene. This assay format is described by Fields
and Song, 1989, Nature 340: 245-246. This type of assay
format can be used in both mammalian cells and in yeast. Other combinations of DNA binding domain and transcriptional activation domain are available in the art and may be
preferred, such as the LexA DNA binding domain and the VP60
transcriptional activation domain.
The precise format of the assay of the invention may be
varied by those of skill in the art using routine skill and
knowledge. For example, the interaction between the polypeptides may be studied in vi tro by labelling one with a
detectable label and bringing it into contact with the other
which has been immobilised on a solid support. Suitable detectable labels include 35S-methionine which may be incorporated into recombinantly produced peptides and
_ polypeptides . Recombinantly produced peptides and
polypeptides may also be expressed as a fusion protein
containing an epitope which can be labelled with an antibody. The protein which is immobilized on a solid support may
be immobilized using an antibody against that protein bound
to a solid support or via other technologies which are known
per se . A preferred in vi tro interaction may utilise a
fusion protein including glutathione-S-transferase (GST) .
This may be immobilized on glutathione agarose beads. In an
in vi tro assay format of the type described above a test
compound can be assayed by determining its ability to
diminish the amount of labelled peptide or polypeptide which
binds to the immobilized GST-fusion polypeptide. This may be determined by fractionating the glutathione-agarose beads by
SDS-polyacrylamide gel electrophoresis . Alternatively, the
beads may be rinsed to remove unbound protein and the amount
of protein which has bound can be determined by counting the
amount of label present in, for example, a suitable scintillation counter.
An assay according to the present invention may also take the form of an in vivo assay. The in vivo assay may be performed in a cell line such as a yeast strain in which the relevant polypeptides or peptides are expressed from one or
more vectors introduced into the cell.
A further assay according to the present invention tests
for ability of a substance to modulate transcriptional
activation by BRCA2 TAD, e.g. by inhibiting interaction of
IRl and/or IR2 with a binding molecule or respective binding
molecules (such as one or more ligands obtainable by assay
(A) or assay (B) ) which inhibit such transcriptional
activation.
Such an assay may involve :
(a) bringing into contact a polypeptide according to
the present invention, including a BRCA2 TAD and a IRl and/or
IR2 and a DNA binding domain capable of binding a nucleotide
sequence within a promoter, a binding molecule or molecules
for IRl and/or IR2 which inhibit (s) transcriptional
activation by the BRCA2 TAD, and a test compound, under
conditions in which the binding molecule or molecule (s) for
IRl and/or IR2 inhibit (s) transcriptional activation of the
promoter by the BRCA2 TAD, ;
(b) providing a nucleic acid molecule which includes a
promoter which includes the nucleotide sequence to which the
polypeptide is capable of binding to activate transcription of a sequence operably linked to the promoter when the
polypeptide is not bound by the IRl and/or IR2 binding
molecule or molecules; and
(c) measuring the degree of modulation or alteration of transcriptional activation caused by said test compound.
A reporter gene construct including a promoter which
includes a nucleotide sequence to which the DNA binding
domain binds, operably linked to a reporter gene, may be
introduced into an expression system such as a cell together
with one or more expression vectors encoding polypeptide or
peptide components of an assay according to the present
invention. Two or more binding sites (for example 3, 4 or 5)
may be present in the nucleic acid construct and this may
enhance sensitivity of the assay.
By "promoter" is meant a sequence of nucleotides from which transcription may be initiated of DNA operably linked
downstream (i.e. in the 3' direction on the sense strand of
double-stranded DNA) .
"Operably linked" in the context of a sequence of interest and a promoter means joined as part of the same nucleic acid molecule, suitably positioned and oriented for transcription to be initiated from the promoter. DNA operably linked to a promoter is "under transcriptional
-initiation regulation" of the promoter.
"Promoter activity" is used to refer to ability to
initiate transcription. The level of promoter activity is
quantifiable for instance by assessment of the amount of mRNA
produced by transcription from the promoter or by assessment
of the amount of protein product produced by translation of
mRNA produced by transcription from the promoter. The amount
of a specific mRNA present in an expression system may be
determined for example using specific oligonucleotides which
are able to hybridise with the mRNA and which are labelled or
may be used in a specific amplification reaction such as the polymerase chain reaction. Use of a reporter gene facilitates determination of promoter activity by reference
to protein production.
Generally, the gene may be transcribed into rnRNA which may be translated into a peptide or polypeptide product which
may be detected and preferably quantitated following expression. A gene whose encoded product may be assayed
following expression is termed a "reporter gene", i.e. a gene
which "reports" on promoter activity.
A reporter gene preferably encodes an enzyme which catalyses a reaction which produces a detectable signal, preferably a visually detectable signal, such as a coloured
-product. Many examples are known, including β-galactosidase and luciferase. β-galactosidase activity may be assayed by production of blue colour on substrate, the assay being by
eye or by use of a spectrophotometer to measure absorbance .
Fluorescence, for example that produced as a result of
luciferase activity, may be quantitated using a
spectrophotometer. Radioactive assays may be used, for
instance using chloramphenicol acetyltransferase, which may
also be used in non-radioactive assays. The presence and/or
amount of gene product resulting from expression from the
reporter gene may be determined using a molecule able to bind
the product, such as an antibody or fragment thereof. The binding molecule may be labelled directly or indirectly using
any standard technique.
Those skilled in the art are well aware of a multitude
of possible reporter genes and assay techniques which may be
used to determine gene activity. Any suitable reporter/assay
may be used and it should be appreciated that no particular choice is essential to or a limitation of the present
invention.
A method of screening for ability of a substance to modulate activity of a promoter may include contacting an expression system, such as a host cell, containing assay
_ components as herein disclosed with a test or candidate substance and determining expression of the heterologous
gene. The level of expression in the presence of the test
substance may be compared with the level of expression in the
absence of the test substance. A difference in expression in
the presence of the test substance indicates ability of the
substance to modulate gene expression. An increase in
expression of the gene compared with expression of another
gene not linked to a promoter as disclosed herein indicates
specificity of the substance for modulation of the promoter.
A promoter construct may be introduced into a cell line
using any technique previously described to produce a stable
cell line containing the reporter construct integrated into the genome. The cells may be grown and incubated with test
compounds for varying times. The cells may be grown in 96
well plates to facilitate the analysis of large numbers of compounds. The cells may then be washed and the reporter
gene expression analysed. For some reporters, such as luciferase the cells will be lysed then analysed.
Following identification of a substance which modulates or affects promoter activity, the substance may be investigated further. Furthermore, it may be manufactured
and/or used in preparation, i.e. manufacture or formulation,
of a composition such as a medicament, pharmaceutical
composition or drug. These may be administered to
individuals.
The amount of test substance or compound which may be
added to an assay of the invention will normally be
determined by trial and error depending upon the type of compound used. Typically, from about 0.01 to 100 nM
concentrations of putative inhibitor compound may be used,
for example from 0.1 to 10 nM. Greater concentrations may be
used when a peptide is the test substance.
Compounds which may be used may be natural or synthetic
chemical compounds used in drug screening programmes.
Extracts of plants which contain several characterised or
uncharacterised components may also be used. A further class
of putative inhibitor compounds can be derived from the BRCA2
polypeptide and/or a ligand which binds IRl and/or IR2. Peptide fragments of from 5 to 40 amino acids, for example
from 6 to 10 amino acids from the region of the relevant polypeptide responsible for interaction, may be tested for their ability to disrupt such interaction. The skilled person can use the techniques described herein and others well known in the art to produce large
amounts of peptides, for instance by expression from encoding
nucleic acid. Peptides can also be generated wholly or partly by
chemical synthesis. The compounds of the present invention
can be readily prepared according to well-established,
standard liquid or, preferably, solid-phase peptide synthesis
methods, general descriptions of which are broadly available
(see, for example, in J.M. Stewart and J.D. Young, Solid
Phase Peptide Synthesis, 2nd edition, Pierce Chemical
Company, Rockford, Illinois (1984) , in M. Bodanzsky and A.
Bodanzsky, The Practice of Peptide Synthesis, Springer Verlag, New York (1984) ; and Applied Biosystems 430A Users Manual, ABI Inc., Foster City, California), or they may be
prepared in solution, by the liquid phase method or by any combination of solid-phase, liquid phase and solution
chemistry, e.g. by first completing the respective peptide
portion and then, if desired and appropriate, after removal of any protecting groups being present, by introduction of
the residue X by reaction of the respective carbonic or sulfonic acid or a reactive derivative thereof. Antibodies .directed to the site of interaction in either
protein form a further class of putative inhibitor compounds .
Candidate inhibitor antibodies may be characterised and their
binding regions determined to provide single chain antibodies and fragments thereof which are responsible for disrupting
the interaction. Antibodies in accordance with the present
invention which discriminate between BRCA2 which is wild-type
and BRCA2 which is mutated in the region of interest, are
useful in screening procedures as discussed further below.
Antibodies may be obtained using techniques which are
standard in the art. Methods of producing antibodies include
immunising a mammal (e.g. mouse, rat, rabbit, horse, goat,
sheep or monkey) with the protein or a fragment thereof. Antibodies may be obtained from immunised animals using any of a variety of techniques known in the art, and screened,
preferably using binding of antibody to antigen of interest.
For instance, Western blotting techniques or
immunoprecipitation may be used (Armitage et al . , 1992,
Nature 357: 80-82) . Isolation of antibodies and/or antibody-
producing cells from an animal may be accompanied by a step of sacrificing the animal .
As an alternative or supplement to immunising a mammal with a peptide, an antibody specific for a protein may be
- obtained from a recombinantly produced library of expressed immunoglobulin variable domains, e.g. using lambda
bacteriophage or filamentous bacteriophage which display
functional immunoglobulin binding domains on their surfaces;
for instance see WO92/01047. The library may be naive, that
is constructed from sequences obtained from an organism which
has not been immunised with any of the proteins (or
fragments) , or may be one constructed using sequences obtained from an organism which has been exposed to the
antigen of interest.
Antibodies according to the present invention may be
modified in a number of ways. Indeed the term "antibody"
should be construed as covering any binding substance having
a binding domain with the required specificity. Thus the
invention covers antibody fragments, derivatives, functional equivalents and homologues of antibodies, including synthetic
molecules and molecules whose shape mimics that of an antibody enabling it to bind an antigen or epitope.
0 Example antibody fragments, capable of binding an antigen or other binding partner are the Fab fragment consisting of the VL, VH, Cl and CHI domains; the Fd fragment consisting of the VH and CHI domains; the Fv fragment consisting of the NL and VH domains of a single arm of an
- antibody; the dAb fragment which consists of a VH domain;
isolated CDR regions and F(ab')2 fragments, a bivalent fragment including two Fab fragments linked by a disulphide bridge at the hinge region. Single chain Fv fragments are
also included.
A hybridoma producing a monoclonal antibody according to
the present invention may be subject to genetic mutation or
other changes. It will further be understood by those
skilled in the art that a monoclonal antibody can be
subjected to the techniques of recombinant D A technology to
produce other antibodies or chimeric molecules which retain
the specificity of the original antibody. Such techniques may involve introducing DΝA encoding the immunoglobulin variable region, or the complementarity determining regions
(CDRs) , of an antibody to the constant regions, or constant
regions plus framework regions, of a different
immunoglobulin. See, for instance, EP184187A, GB 2188638A or EP-A-0239400. Cloning and expression of chimeric antibodies are described in EP-A-0120694 and EP-A-0125023.
Hybridomas capable of producing antibody with desired
binding characteristics are within the scope of the present invention, as are host cells, eukaryotic or prokaryotic, containing nucleic acid encoding antibodies (including
antibody fragments) and capable of their expression. The invention also provides methods of production of the
antibodies including growing a cell capable of producing the
antibody under conditions in which the antibody is produced,
and preferably secreted.
The reactivities of antibodies on a sample (e.g. the
subject of a diagnostic test) may be determined by any
appropriate means. Tagging with individual reporter
molecules is one possibility. The reporter molecules may
directly or indirectly generate detectable, and preferably
measurable, signals. The linkage of reporter molecules may
be directly or indirectly, covalently, e.g. via a peptide
bond or non-covalently. Linkage via a peptide bond may be as
a result of recombinant expression of a gene fusion encoding antibody and reporter molecule.
One favoured mode is by covalent linkage of each
antibody with an individual fluorochrome, phosphor or laser
dye with spectrally isolated absorption or emission
characteristics. Suitable fluorochromes include fluorescein, rhodamine, phycoerythrin and Texas Red. Suitable chromogenic dyes include diaminobenzidine . Other reporters include macromolecular colloidal
- particles or particulate material such as latex beads that are coloured, magnetic or paramagnetic, and biologically or
chemically active agents that can directly or indirectly
cause detectable signals to be visually observed,
electronically detected or otherwise recorded. These
molecules may be enzymes which catalyse reactions that
develop or change colours or cause changes in electrical
properties, for example. They may be molecularly excitable,
such that electronic transitions between energy states result
in characteristic spectral absorptions or emissions. They
may include chemical entities used in conjunction with
biosensors. Biotin/avidin or biotin/streptavidin and
alkaline phosphatase detection systems may be employed.
The mode of determining binding is not a feature of the present invention and those skilled in the art are able to
choose a suitable mode according to their preference and general knowledge.
Antibodies in accordance with the present invention may be used in screening for the presence of a particular polypeptide, for example in a test sample containing cells or
cell lysate as discussed, such as a BRCA2 polypeptide including a mutation in the TAD, IRl and/or IR2, where such _ mutation is reflected in an alteration in one or more
epitopes allowing discrimination between the mutant and wild-
type regions of BRCA2. Screening methods using antibodies
are discussed further below.
Antibodies may also be used in purifying and/or
isolating a polypeptide according to the present invention,
for instance following production of the polypeptide by
expression from encoding nucleic acid therefor. Antibodies
may modulate the activity of the polypeptide to which they
bind and so, if that polypeptide has a deleterious effect in
an individual, may be useful in a therapeutic context (which
may include prophylaxis) .
An antibody may be provided in a kit, which may include instructions for use of the antibody, e.g. in determining the
presence of a particular substance in a test sample. One or more other reagents may be included, such as labelling
molecules, buffer solutions, elutants and so on. Reagents may be provided within containers which protect them from the external environment , such as a sealed vial .
Other candidate inhibitor compounds may be based on modelling the 3 -dimensional structure of a polypeptide or
_ peptide fragment and using rational drug design to provide potential inhibitor compounds with particular molecular
shape, size and charge characteristics.
In a further aspect, the invention provides compounds
obtainable by an assay according to the present invention,
for example peptide compounds based on portions of the BRCA2
polypeptide .
A compound found to have the ability to modulate
transcriptional activity of BRCA2 TAD, with therapeutic
potential in anti-tumour treatment, may be used in
combination with any other anti-tumour compound. In such a case, the assay of the invention, when conducted in vivo,
need not measure the degree of inhibition of binding or
transcriptional activation caused by the inhibitor compound
being tested. Instead the effect on tumorigenicity may be measured. It may be that such a modified assay is run in 0 parallel or subsequent to the main assay of the invention in
order to confirm that any effect on tumorigenicity is as a
result of the inhibition of binding or transcriptional activation caused by said inhibitor compound and not merely a general toxic effect.
The present invention extends in various aspects not
only to a substance identified as a modulator of BRCA2
transcriptional activity, in accordance with what is
disclosed herein, but also a pharmaceutical composition,
medicament, drug or other composition including such a
substance, a method including administration of such a
composition to a patient, e.g. for anti-tumour or other anti-
proliferative treatment, which may include preventative
treatment , use of such a substance in manufacture of a composition for administration, e.g. for anti-tumour or other
anti-proliferative treatment, and a method of making a
pharmaceutical composition including admixing such a
substance with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients.
Also encompassed within the scope of the present
invention are functional mimetics of peptide fragments of
BRCA2 polypeptide TAD, IRl or IR2 , or the binding ligand or ligands for IRl and/or IR2 , which interfere with interaction between these polypeptides, and/or transcriptional activation by BRCA2 TAD. The term "functional mimetic" means a substance which may not contain an active portion of the
- relevant amino acid sequence, and probably is not a peptide
at all, but which retains the relevant interfering activity. The design and screening of candidate mimetics is described
in detail below.
A substance identified using the present invention may
be peptide or non-peptide in nature. Non-peptide "small
molecules" are often preferred for many in vivo
pharmaceutical uses. Accordingly, a mimetic or mimick of the
substance (particularly if a peptide) may be designed for
pharmaceutical use.
The designing of mimetics to a known pharmaceutically
active compound is a known approach to the development of
pharmaceuticals based on a "lead" compound. This might be
desirable where the active compound is difficult or expensive to synthesise or where it is unsuitable for a particular
method of administration, e.g. peptides are not well suited as active agents for oral compositions as they tend to be 0 quickly degraded by proteases in the alimentary canal.
Mimetic design, synthesis and testing may be used to avoid randomly screening large number of molecules for a target property. There are several steps commonly taken in the design of
- a mimetic from a compound having a given target property. Firstly, the particular parts of the compound that are
critical and/or important in determining the target property
are determined. In the case of a peptide, this can be done
by systematically varying the amino acid residues in the
peptide, e.g. by substituting each residue in turn. These parts or residues constituting the active region of the
compound are known as its "pharmacophore" . Once the pharmacophore has been found, its structure is
modelled to according its physical properties, e.g.
stereochemistry, bonding, size and/or charge, using data from
a range of sources, e.g. spectroscopic techniques, X-ray
diffraction data and NMR. Computational analysis, similarity
mapping (which models the charge and/or volume of a
pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modelling process.
In a variant of this approach, the three-dimensional
structure of the ligand and its binding partner are modelled. This can be especially useful where the ligand and/or binding
partner change conformation on binding, allowing the model to take account of this the design of the mimetic.
A template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted. The
- template molecule and the chemical groups grafted on to it
can conveniently be selected so that the mimetic is easy to synthesise, is likely to be pharmacologically acceptable, and
does not degrade in vivo, while retaining the biological
activity of the lead compound. The mimetic or mimetics found
by this approach can then be screened to see whether they
have the target property, or to what extent they exhibit it.
Further optimisation or modification can then be carried out
to arrive at one or more final mimetics for in vivo or
clinical testing.
Whether it is a polypeptide, antibody, peptide, nucleic
acid molecule, small molecule, mimetic or other
pharmaceutically useful compound according to the present
invention that is to be given to an individual,
administration is preferably 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. 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.
A composition may be administered alone or in
combination with other treatments, either simultaneously or
sequentially dependent upon the condition to be treated.
Pharmaceutical compositions according to the present
invention, and for use in accordance with the present invention, may include, in addition to active ingredient, a
pharmaceutically acceptable excipient, carrier, buffer,
stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not
interfere with the efficacy of the active ingredient. The
precise nature of the carrier or other material will depend
on the route of administration, which may be oral, or by
injection, e.g. cutaneous, subcutaneous or intravenous.
Pharmaceutical compositions for oral administration may
be in tablet, capsule, powder or liquid form. A tablet may include a solid carrier such as gelatin or an adjuvant.
Liquid pharmaceutical compositions generally include a liquid
carrier such as water, petroleum, animal or vegetable oils,
mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol , propylene glycol or polyethylene glycol may be included.
For intravenous, cutaneous or subcutaneous injection, or
injection at the site of affliction, the active ingredient
will be in the form of a parenterally acceptable aqueous
solution which is pyrogen-free and has suitable pH,
isotonicity and stability. Those of relevant skill in the
art are well able to prepare suitable solutions using, for
example, isotonic vehicles such as Sodium Chloride Injection,
Ringer's Injection, Lactated Ringer's Injection.
Preservatives, stabilisers, buffers, antioxidants and/or
other additives may be included, as required.
A polypeptide, peptide or other substance able to
interfere with the interaction of the relevant polypeptide,
peptide or other substance as disclosed herein may be provided in a kit, e.g. sealed in a suitable container which
protects its contents from the external environment . Such a
kit may include instructions for use.
The experimental evidence included below demonstrates that a mutation within the BRCA2 TAD which is associated with
familial breast cancer severely reduces the ability of the TAD to activate transcription. Given such experimental evidence, oligonucleotides
-designed to hybridise to a region within the TAD or IRl or
IR2 may be used in diagnostic and prognostic screening.
Oligonucleotide probes or primers, as well as the full- length BRCA2 TAD (optionally including IRl and/or IR2)
sequence (and mutants, alleles, variants and derivatives) are
useful in screening a test sample containing nucleic acid for
the presence of alleles, mutants and variants, especially
those that confer susceptibility or predisposition to
proliferative disorders, including cancers, the probes
hybridising with a target sequence from a sample obtained
from the individual being tested. The conditions of the
hybridisation can be controlled to minimise non-specific
binding, and preferably stringent to moderately stringent
hybridisation conditions are preferred. The skilled person
is readily able to design such probes, label them and devise suitable conditions for the hybridisation reactions, assisted
by textbooks such as Sambrook et al (1989) and Ausubel et al
(1992) . Nucleic acid isolated and/or purified from one or more cells (e.g. human) or a nucleic acid library derived from nucleic acid isolated and/or purified from cells (e.g. a cDNA library derived from mRNA isolated from the cells) , may be probed under conditions for selective hybridisation and/or
-subjected to a specific nucleic acid amplification reaction
such as the polymerase chain reaction (PCR) .
A method may include hybridisation of one or more (e.g.
two) probes or primers to target nucleic acid. Where the
nucleic acid is double-stranded DNA, hybridisation will
generally be preceded by denaturation to produce single- stranded DNA. The hybridisation may be as part of a PCR
procedure, or as part of a probing procedure not involving
PCR. An example procedure would be a combination of PCR and
low stringency hybridisation. A screening procedure, chosen
from the many available to those skilled in the art, is used
to identify successful hybridisation events and isolated
hybridised nucleic acid.
Binding of a probe to target nucleic acid (e.g. DNA) may
be measured using any of a variety of techniques at the disposal of those skilled in the art. For instance, probes may be radioactively, fluorescently or enzymatically labelled. Other methods not employing labelling of probe
include examination of restriction fragment length
polymorphisms, amplification using PCR, RNAase cleavage and allele specific oligonucleotide probing.
Probing may employ the standard Southern blotting technique . For instance DNA may be extracted from cells and
-digested with different restriction enzymes. Restriction
fragments may then be separated by electrophoresis on an
agarose gel, before denaturation and transfer to a
nitrocellulose filter. Labelled probe may be hybridised to
the DNA fragments on the filter and binding determined. DNA
for probing may be prepared from RNA preparations from cells.
Those skilled in the art are well able to employ suitable conditions of the desired stringency for selective
hybridisation, taking into account factors such as
oligonucleotide length and base composition, temperature and
so on.
PCR techniques for the amplification of nucleic acid are
described in US Patent No. 4,683,195. In general, such
techniques require that sequence information from the ends of
the target sequence is known to allow suitable forward and reverse oligonucleotide primers to be designed to be identical or similar to the polynucleotide sequence that is
the target for the amplification. PCR includes steps of denaturation of template nucleic acid (if double-stranded) ,
annealing of primer to target, and polymerisation. The nucleic acid probed or used as template in the amplification reaction may be genomic DNA, cDNA or RNA. PCR can be used to amplify specific sequences from genomic DNA, specific RNA
-sequences and cDNA transcribed from mRNA, bacteriophage or plasmid sequences. References for the general use of PCR
techniques include Mullis et al , Cold Spring Harbor Symp .
Quant. Biol . , 51:263, (1987), Ehrlich (ed) , PCR technology, Stockton Press, NY, 1989, Ehrlich et al , Science, 252:1643-
1650, (1991) , "PCR protocols; A Guide to Methods and Applications", Eds. Innis et al , Academic Press, New York,
(1990) . On the basis of amino acid sequence information,
oligonucleotide probes or primers may be designed, taking
into account the degeneracy of the genetic code, and, where
appropriate, codon usage of the organism from the candidate
nucleic acid is derived. An oligonucleotide for use in
nucleic acid amplification may have about 10 or fewer codons
(e.g. 6, 7 or 8) , i.e. be about 30 or fewer nucleotides in
length (e.g. 18, 21 or 24) . Generally specific primers are
upwards of 14 nucleotides in length, but generally need not
be more than 18-20. Those skilled in the art are well versed in the design of primers for use processes such as PCR.
A further aspect of the present invention provides an oligonucleotide or polynucleotide fragment of the nucleotide sequence shown for the BRCA2 TAD, IRl and IR2 regions in
Figure 2 or a complementary sequence, in particular for use
in a method of obtaining and/or screening nucleic acid. The
sequences referred to above may be modified by addition,
substitution, insertion or deletion of one or more
nucleotides, but preferably without abolition of ability to hybridise selectively with nucleic acid with the sequence
shown for the BRCA2 region in Figure 2, that is wherein the
degree of homology of the oligonucleotide or polynucleotide
with the sequence given is sufficiently high.
In some preferred embodiments, oligonucleotides
according to the present invention that are fragments of the
sequences shown for regions of BRCA2 in Figure 2 , or any
allele associated with susceptibility to cancer or other
disorder of cell proliferation, are at least about 10
nucleotides in length, more preferably at least about 15 nucleotides in length, more preferably at least about 20
nucleotides in length. Such fragments themselves individually represent aspects of the present invention.
Fragments and other oligonucleotides may be used as primers or probes as discussed but may also be generated (e.g. by
PCR) in methods concerned with determining the presence in a test sample of a sequence indicative of susceptibility to cancer or other disorder of cell -cycle regulation.
Preferred probes or primers according to certain
embodiments of this aspect of the present invention are
designed to hybridise with and/or amplify a fragment of the
BRCA2 TAD, shown in Figure 2, including the codon for residue
42, mutation at which is associated with cancer
susceptibility and is demonstrated herein to severely reduce
transcriptional activation by the BRCA2 TAD. This is as a
result of a change in the coding nucleotide sequence from TAT
to TGT.
Thus, suitable oligonucleotides for probing or priming
around codon 42 or other codon associated with disease
susceptibility in the coding sequence shown in Figure 2 may be about 20 nucleotides in length, or may include a
contiguous sequence of about 20 nucleotides of the coding
sequence shown (either sense strand or anti-sense strand,
also known as coding and non-coding strands respectively) . The oligonucleotide sequence may be designed so that it
anneals to the sense or anti-sense strand with the codon of interest (e.g. 42) towards the middle of the oligonucleotide,
or at or adjacent the 3' or 5 ' end, different possibilities being preferred for different purposes.
Exemplary sequences of oligonucleotides designed to anneal around codon .42 of the wild-type sequence shown in
Figure 2 thus include :
5 ' -TATAATTCTGAACCTGCAGA-3 '
(corresponding to a portion of the sense strand, and thus
complementary to the anti-sense strand with codon 42 at the
5 ' end)
5 * -ATAATTCTGAACCTGCAGAT-3
(corresponding to a portion of the sense strand, and thus
complementary to the anti-sense strand with the nucleotide
(A) that is changed (to T) in the mutation discussed herein
at the 5 ' end)
5 ' -ATAGGGTGGAGCTTCTGAAG-3 "
(corresponding to a portion of the anti -sense strand, and
thus complementary to the sense strand with the anti-codon
for codon 42 at the 5' end of the oligonucleotide) 5 * -TAGGGTGGAGCTTCTGAAGA-3 '
(corresponding to a portion of the anti-sense strand, and thus complementary to the sense strand with the nucleotide
complementary to the A that is changed in the mutation
discussed herein at the 5' end of the oligonucleotide) 5 ' -GCTCCACCCTATAATTCTG-3 5 ' -CAGAATTATAGGGTGGAGC-3 (respectively complementary to the anti-sense and sense strands having codon 42 (or its complement) towards the _middle of the oligonucleotides) .
These oligonucleotides will anneal without mismatch to
the wild-type sequence but will include a mismatch where the
TAT at codon 42 is mutated to TGT.
Oligonucleotides that will anneal to the mutated
sequence including TGT at codon 42 without mismatch, and the
wild-type sequence with mismatch, include 5 ' -TGTAATTCTGAACCTGCAGA-3 '
5 ' -ACAATTCTGAACCTGCAGAT-3
5 ' -GTAGGGTGGAGCTTCTGAAG-3 '
5 ' -CAGGGTGGAGCTTCTGAAGA-3 '
5 ' -GCTCCACCCTGTAATTCTG-3 '
5 • -CAGAATTACAGGGTGGAGC-3
(corresponding to the oligonucleotides given above which
anneal without mismatch to the wild-type sequence, but with the requisite change at the position corresponding to codon 42) .
Similarly designed oligonucleotides may be employed for any mutation or other sequence alteration within a BRCA2 fragment according to the present invention.
Detection of mismatches and other techniques useful for screening of mutations are discussed further below. Other oligonucleotides useful in accordance with various
" aspects of the present invention, such as methods involving
amplification of sequences encoding particular BRCA2
fragments, include: 5'-ATGCCTATTGGATCCAAAGA-3 '
(complementary to the 3 ' end of the antisense strand of the
coding sequence shown in Figure 2)
5 ' -GAACTTGACCAAGACATATC-3 '
(complementary to the 3 ' end of the sense strand of the coding sequence shown in Figure 2)
5 ' -ATCCATTTTAGTTTTCACTG-3 '
(complementary to the sense strand of the coding sequence at
the 3' end of the region encoding IR2 , ending with codon 125
in Figure 2)
5 ' -TAAGTCTAATTTGAATTTAT-3 '
(complementary to the sense strand of the coding sequence at the 3 ' end of the region encoding the TAD AAR, ending with codon 105)
5 -AACCTATTTAAACTCCACAA-3 ' (complementary to the anti-sense strand of the coding
sequence at its 3 ' end corresponding to the 5 ' end of the region of the sense strand encoding the TAD AAR, beginning with codon 60) 5 ' -GTTTGGTTCGTAATTGTTGT-3 '
(complementary to the sense strand of the coding sequence at the 3 ' end of the region encoding the TAD PAR, ending with
codon 60)
5 ' -CGCTGCAACAAAGCAGATTT-3 '
(complementary to the anti -sense strand of the coding
sequence at its 3 ' end corresponding to the 5 ' end of the
region of the sense strand encoding the TAD PAR, beginning at codon 18) .
These and other oligonucleotides may be used in various
combinations to amplify particular regions of the BRCA2
fragment, including the whole fragment for which the sequence
is shown in Figure 2, the TAD, the TAD AAR, the TAD PAR, and
the TAD plus IRl and/or IR2. Methods involving use of nucleic acid in diagnostic
and/or prognostic contexts, for instance in determining
susceptibility to, e.g., cancer, and other methods concerned with determining the presence of sequences indicative of,
e.g., cancer susceptibility
A number of methods are known in the art for analysing
biological samples from individuals to determine whether the individual carries an BRCA2 allele with a mutation within the region for which the sequence is shown in Figure 2
predisposing them to disease. The purpose of such analysis
may be used for diagnosis or prognosis, and serve to detect
the presence of, e.g., an existing cancer, to help identify
the type of cancer, to assist a physician in determining the
severity or likely course of the cancer and/or to optimise
treatment of it . The methods may be used to detect alleles
that are statistically associated with a susceptibility to
cancer or other proliferative disorder in the future, e.g.
early onset cancer, identifying individuals who would benefit from regular screening to provide early diagnosis of cancer.
Broadly, the methods divide into those screening for the
presence of nucleic acid sequences and those that rely on
detecting the presence or absence of polypeptide. The
methods make use of biological samples from individuals that
are suspected of contain the nucleic acid sequences or polypeptide. Examples of biological samples include blood,
plasma, serum, tissue samples, tumour samples, saliva and
urine .
Exemplary approaches for detecting nucleic acid or polypeptides include:
(a) comparing the sequence of nucleic acid in the sample with an BRCA2 nucleic acid sequence to determine whether the
_ sample from the patient contains one or more mutations in a
region for which the sequence is shown in Figure 2; or,
(b) determining the presence in a sample from a patient
of the polypeptide encoded by BRCA2 and, if present,
determining whether the polypeptide is includes a region
corresponding to that shown in Figure 2 (e.g. including TAD,
IRl and/or IR2 ) , and/or is mutated in such a region; or,
(c) using DNA fingerprinting to compare the restriction
pattern produced when a restriction enzyme cuts a sample of
nucleic acid from the patient with the restriction pattern
obtained from a region corresponding to that shown in Figure
2 for normal gene or from known mutations thereof; or,
(d) using a specific binding member capable of binding
to a nucleic acid sequence (either a normal sequence or a
known mutated sequence) encoding a fragment of BRCA2
corresponding to a region for which the sequence is shown in
Figure 2 (e.g. including TAD, IRl and/or IR2) , the specific
binding member including nucleic acid hybridisable with the
0 BRCA2 sequence, or substances including an antibody domain
with specificity for a native or mutated BRCA2 fragment
nucleic acid sequence or the polypeptide encoded by it, the
specific binding member being labelled so that binding of the specific binding member to its binding partner is detectable;
-or,
(e) using PCR involving one or more primers based on
normal or mutated BRCA2 gene sequence to screen for normal or
mutant sequences within the region of BRCA2 corresponding to
a region for which the sequence is shown in Figure 2 (e.g.
including TAD, IRl and/or IR2) in a sample from a patient.
A "specific binding pair" includes a specific binding
member (sbm) and a binding partner (bp) which have a particular specificity for each other and which in normal
conditions bind to each other in preference to other
molecules. Examples of specific binding pairs are antigens
and antibodies (see above) , molecules and receptors and
complementary nucleotide sequences. The skilled person will
be able to think of many other examples and they do not need
to be listed here. Further, the term "specific binding pair"
is also applicable where either or both of the specific
binding member and the binding partner include a part of a
larger molecule. In embodiments in which the specific binding pair are nucleic acid sequences, they will be of a length to hybridise to each other under the conditions of the assay, preferably greater than 10 nucleotides long, more preferably greater than 15 or 20 nucleotides long. In most embodiments for screening for susceptibility
alleles, the BRCA2 nucleic acid in the sample will initially
be amplified, e.g. using PCR, to increase the amount of the
analyte as compared to other sequences present in the sample .
This allows the target sequences to be detected with a high
degree of sensitivity if they are present in the sample.
This initial step may be avoided by using highly sensitive array techniques that are becoming increasingly important in
the art .
To reiterate in further detail, the identification of a
significant region of the BRCA2 gene and its implication with
disorders of cell proliferation paves the way for aspects of
the present invention to provide the use of materials and
methods, such as are disclosed and discussed above, for
establishing the presence or absence in a test sample of an variant form of the gene, in particular an allele or variant
specifically associated with cancer. This may be for diagnosing a predisposition of an individual to cancer. It
may be for diagnosing cancer of a patient with the disease as being associated with the gene.
This allows for planning of appropriate therapeutic and/or prophylactic treatment, permitting stream-lining of treatment by targeting those most likely to benefit.
A variant form of the gene may contain one or more
insertions, deletions, substitutions and/or additions of one
or more nucleotides compared with the wild-type sequence
which may or may not disrupt the transcriptional activation
function of the region examined herein. Differences at the
nucleic acid level are not necessarily reflected by a
difference in the amino acid sequence of the encoded
polypeptide. However, a mutation or other difference in a
gene may result in a frame-shift or stop codon, which could
seriously affect the nature of the polypeptide produced, or a
point mutation or gross mutational change to the encoded
polypeptide, including insertion, deletion, substitution
and/or addition of one or more amino acids or regions in the
polypeptide, which may affect transcriptional activation.
There are various methods for determining the presence
or absence in a test sample of a particular nucleic acid
sequence, such as the sequence shown for a BRCA2 fragment in Figure 2 or a mutant, variant or allele thereof.
Tests may be carried out on preparations containing genomic DNA, cDNA and/or mRNA. Testing cDNA or mRNA has the
advantage of the complexity of the nucleic acid being reduced by the absence of intron sequences, but the possible
disadvantage of extra time and effort being required in
making the preparations. RNA is more difficult to manipulate
than DNA because of the wide-spread occurrence of RN'ases.
Nucleic acid in a test sample may be sequenced and the
sequence compared with the sequence shown in Figure 2 to
determine whether or not a difference is present. If so, the
difference can be compared with known susceptibility alleles
to determine whether the test nucleic acid contains one or more of the variations indicated, or the difference can be
investigated for association with cancer.
Since it will not generally be time- or labour-efficient
to sequence all nucleic acid in a test sample or even the
whole BRCA2 gene fragment corresponding to the region shown
in Figure 2, a specific amplification reaction such as PCR
using one or more pairs of primers may be employed to amplify
the region of interest in the nucleic acid. The amplified
nucleic acid may then be sequenced as above, and/or tested in any other way to determine the presence or absence of a particular feature. Nucleic acid for testing may be prepared
from nucleic acid removed from cells or in a library using a
variety of other techniques such as restriction enzyme digest and electrophoresis . Nucleic acid may be screened using a variant- or allele-
specific probe. Such a probe corresponds in sequence to a
region of the BRCA2 gene, or its complement, containing a
sequence alteration known to be associated with
susceptibility to cancer or other proliferative disorder.
Under suitably stringent conditions, specific hybridisation
of such a probe to test nucleic acid is indicative of the
presence of the sequence alteration in the test nucleic acid.
For efficient screening purposes, more than one probe may be
used on the same test sample.
Allele- or variant-specific oligonucleotides may
similarly be used in PCR to specifically amplify particular
sequences if present in a test sample. Assessment of whether
a PCR band contains a gene variant may be carried out in a
number of ways familiar to those skilled in the art. The PCR
product may for instance be treated in a way that enables one
to display the mutation or polymorphism on a denaturing
polyacrylamide DNA sequencing gel, with specific bands that are linked to the gene variants being selected. An alternative or supplement to looking for the presence
of variant sequences in a test sample is to look for the presence of the normal sequence, e.g. using a suitably specific oligonucleotide probe or primer. Use of oligonucleotide probes and primers has been
discussed in more detail above.
Approaches which rely on hybridisation between a probe
and test nucleic acid and subsequent detection of a mismatch
may be employed. Under appropriate conditions (temperature,
pH etc.), an oligonucleotide probe will hybridise with a
sequence which is not entirely complementary. The degree of
base-pairing between the two molecules will be sufficient for
them to anneal despite a mis-match. Various approaches are
well known in the art for detecting the presence of a mis¬
match between two annealing nucleic acid molecules.
For instance, RN'ase A cleaves at the site of a mis¬
match. Cleavage can be detected by electrophoresing test
nucleic acid to which the relevant probe or probe has
annealed and looking for smaller molecules (i.e. molecules
with higher electrophoretic mobility) than the full length
probe/test hybrid. Other approaches rely on the use of enzymes such as resolvases or endonucleases .
Thus, an oligonucleotide probe that has the sequence of a region of the normal BRCA2 gene (either sense or anti-sense strand) corresponding to the fragment shown in Figure 2 in which at least one mutation associated with, e.g., cancer susceptibility is known to occur, may be annealed to test nucleic acid and the presence or absence of a mis-match
-determined. Detection of the presence of a mis-match may
indicate the presence in the test nucleic acid of a mutation
associated with, e.g., cancer susceptibility. On the other
hand, an oligonucleotide probe that has the sequence of a
region of the BRCA2 gene including a mutation associated
with, e.g., cancer susceptibility may be annealed to test
nucleic acid and the presence or absence of a mis-match
determined. The presence of a mis-match may indicate that
the nucleic acid in the test sample has the normal sequence. In either case, a battery of probes to different regions of
the gene may be employed.
The presence of differences in sequence of nucleic acid
molecules may be detected by means of restriction enzyme digestion, such as in a method of DNA fingerprinting where
the restriction pattern produced when one or more restriction
enzymes are used to cut a sample of nucleic acid is compared
with the pattern obtained when a sample containing the normal
gene or a variant or allele is digested with the same enzyme
or enzymes.
A test sample of nucleic acid may be provided for example by extracting nucleic acid from cells, e.g. in saliva or preferably blood, or for pre-natal testing from the amnion, placenta or foetus itself.
Nucleic acid according to the present invention, such as a full-length coding sequence or oligonucleotide probe or
primer, may be provided as part of a kit, e.g. in a suitable
container such as a vial in which the contents are protected
from the external environment. The kit may include
instructions for use of the nucleic acid, e.g. in PCR and/or
a method for determining the presence of nucleic acid of
interest in a test sample. A kit wherein the nucleic acid is intended for use in PCR may include one or more other
reagents required for the reaction, such as polymerase,
nucleosides, buffer solution etc. The nucleic acid may be labelled. A kit for use in determining the presence or
absence of nucleic acid of interest may include one or more
articles and/or reagents for performance of the method, such
as means for providing the test sample itself, e.g. a swab for removing cells from the buccal cavity or a syringe for
removing a blood sample (such components generally being sterile) . In a further aspect, the present invention provides an apparatus for screening for BRCA2 TAD, IRl and/or IR2 nucleic acid, the apparatus including storage means including the BRCA2 nucleic acid sequence as set out in Figure 2, or a fragment thereof, the stored sequence being
_used to compare the sequence of the test nucleic acid to
determine the presence of mutations.
There are various methods for determining the presence
or absence in a test sample of a particular polypeptide, such
as a polypeptide including a fragment of BRCA2 corresponding
to a region for which the amino acid sequence is shown in
Figure 2 or an amino acid sequence mutant, variant or allele
thereof.
A sample may be tested for the presence of a binding
partner for a specific binding member such as an antibody (or
mixture of antibodies) , specific for one or more particular
variants of the BRCA2 polypeptide shown in the figures, or a
mutant, variant or allele thereof.
A sample may be tested for the presence of a binding partner for a specific binding member such as an antibody (or
mixture of antibodies) , specific for the BRCA2 polypeptide
shown in the figures. In such cases, the sample may be tested by being contacted with a specific binding member such as an antibody under appropriate conditions for specific binding, before binding is determined, for instance using a reporter system as discussed. Where a panel of antibodies is used, different
_reporting labels may be employed for each antibody so that binding of each can be determined.
A specific binding member such as an antibody may be
used to isolate and/or purify its binding partner polypeptide
from a test sample, to allow for sequence and/or biochemical
analysis of the polypeptide to determine whether it has the
sequence and/or properties of the BRCA2 polypeptide whose
sequence is shown in the figures, or if it is a mutant or
variant form. Amino acid sequence is routine in the art
using automated sequencing machines.
Nucleic acid according to the present invention,
encoding a polypeptide functional as a transcriptional
activator, may be used in methods of gene therapy, for
instance in treatment of individuals with the aim of
preventing or curing (wholly or partially) cancer or other disorder involving loss of proper regulation of the cell-
cycle and/or cell growth.
Nucleic acid encoding an authentic biologically active BRCA2 fragment polypeptide, i.e. with ability to activate transcription, may be used in a method of gene therapy, to treat a patient who is unable to synthesize the active polypeptide or unable to synthesize it at the normal level,
thereby providing the effect provided by wild-type and
suppressing the occurrence of cancer and/or reduce the size
or extent of existing defects in cell -cycle and/or growth
regulation in the target cells.
Vectors such as viral vectors have been used in the
prior art to introduce genes into a wide variety of different
target cells. Typically the vectors are exposed to the
target cells so that transfection can take place in a
sufficient proportion of the cells to provide a useful
therapeutic or prophylactic effect from the expression of the
desired polypeptide. The transfected nucleic acid may be
permanently incorporated into the genome of each of the
targeted tumour cells, providing long lasting effect, or
alternatively the treatment may have to be repeated
periodically.
A variety of vectors, both viral vectors and plasmid
vectors, are known in the art, see US Patent No. 5,252,479 and WO 93/07282. In particular, a number of viruses have been used as gene transfer vectors, including papovaviruses, such as SV40, vaccinia virus, herpesviruses , including HSV and EBV, and retroviruses . Many gene therapy protocols in the prior art have used disabled murine retroviruses. As an alternative to the use of viral vectors other
_ known methods of introducing nucleic acid into cells includes electroporation, calcium phosphate co-precipitation,
mechanical techniques such as microinjection, transfer
mediated by liposomes and direct DNA uptake and receptor-
mediated DNA transfer.
As mentioned above, the aim of gene therapy using
nucleic acid encoding an BRCA2 polypeptide, or an active
portion thereof, is to increase the amount of the expression
product of the nucleic acid in cells in which the level of
the wild-type polypeptide is absent or present only at reduced levels. Such treatment may be therapeutic in the
treatment of cells which are already cancerous or pre- cancerous or prophylactic in the treatment of individuals
known through screening to have an BRCA2 susceptibility
allele and hence a predisposition to cancer.
Receptor-mediated gene transfer, in which the nucleic
acid is linked to a protein ligand via polylysine, with the ligand being specific for a receptor present on the surface
of the target cells, is an example of a technique for
specifically targeting nucleic acid to particular cells.
As noted, a polypeptide termed "BBPl" (BRCA2 Binding Protein 1) has been found to interact with the PAR of BRCA2
_and a portion of it able to repress transcriptional
activation by BRCA2 TAD.
The isolated polypeptide, isolated nucleic acid encoding
it, vectors and host cells including the nucleic acid, and
methods of making the polypeptide all represent further
aspects of the present invention.
Thus in one aspect the present invention provides a
polypeptide including the amino acid sequence shown in Figure
3 or the amino acid sequence shown in Figure 4. In one
embodiment a polypeptide according to the present invention
may be approximately up to about 2000 amino acids, or as encoded by the approximately 7.5kb mRNA discussed below. In
one embodiment a polypeptide according to the invention
includes the amino acid sequence of Figure 3 or Figure 4, is
able to bind BRCA2 TAD and is obtainable from human ovary or
testis cells. Antibodies directed against the amino acid
sequence of Figure 3 or Figure 4 , or a suitable fragment
thereof, may be used in isolation and/or purification of the
polypeptide from ovary or testis cells. The polypeptide may be provided in isolated form and may be formulated into a composition containing at least one additional component, such as a pharmaceutically acceptable excipient. Nucleic acid encoding the BBPl polypeptide is also
provided as an aspect of the present invention. The invention provides a nucleic acid molecule including the
encoding nucleotide sequence shown in Figure 3 or Figure 4.
The nucleic acid molecule may be RNA of approximately 7.5 kb
including said encoding sequence (the RNA equivalent) or a
cDNA copy of such RNA and obtainable by probing a cDNA
library or RNA generated from human ovary or testes .
Nucleic acid according to this aspect of the present
invention is obtainable using one or more oligonucleotide probes or primers designed to hybridise with one or more
fragments of the nucleic acid sequence shown in Figure 3 or Figure 4, particularly fragments of relatively rare sequence,
based on codon usage or statistical analysis. A primer
designed to hybridise with a fragment of the nucleic acid
sequence shown in Figure 3 or Figure 4 may be used in conjunction with one or more oligonucleotides designed to
hybridise to a sequence in a cloning vector within which
target nucleic acid has been cloned, or in so-called "RACE"
(rapid amplification of cDNA ends) in which cDNA's in a
library are ligated to an oligonucleotide linker and PCR is performed using a primer which hybridises with the sequence shown in Figure 3 or Figure 4 and a primer which hybridises to the oligonucleotide linker.
In the context of cloning, it may be necessary for one
or more gene fragments to be ligated to generate a full-
length coding sequence. Also, where a full-length encoding
nucleic acid molecule has not been obtained, a smaller
molecule representing part of the full molecule, may be used
to obtain full-length clones. Inserts may be prepared from
partial cDNA clones and used to screen cDNA libraries. The
full-length clones isolated may be subcloned into expression
vectors and activity assayed by transfection into suitable
host cells, e.g. with a reporter plasmid.
A method may include hybridisation of one or more (e.g.
two) probes or primers to target nucleic acid. Where the nucleic acid is double-stranded DNA, hybridisation will
generally be preceded by denaturation to produce single-
stranded DNA. The hybridisation may be as part of a PCR
procedure, or as part of a probing procedure not involving PCR. An example procedure would be a combination of PCR and
low stringency hybridisation. A screening procedure, chosen
from the many available to those skilled in the art, is used to identify successful hybridisation events and isolated hybridised nucleic acid.
Binding of a probe to target nucleic acid (e.g. DNA) may be measured using any of a variety of techniques at the
disposal of those skilled in the art. For instance, probes may be radioactively, fluorescently or enzymatically
labelled. Other methods not employing labelling of probe
include examination of restriction fragment length
polymorphisms, amplification using PCR, RNAase cleavage and
allele specific oligonucleotide probing.
Probing may employ the standard Southern blotting
technique. For instance DNA may be extracted from cells and
digested with different restriction enzymes. Restriction
fragments may then be separated by electrophoresis on an
agarose gel, before denaturation and transfer to a nitrocellulose filter. Labelled probe may be hybridised to
the DNA fragments on the filter and binding determined. DNA
for probing may be prepared from RNA preparations from cells.
Preliminary experiments may be performed by hybridising
under low stringency conditions various probes to Southern
blots of DNA digested with restriction enzymes. Suitable conditions would be achieved when a large number of
hybridising fragments were obtained while the background
hybridisation was low. Using these conditions nucleic acid libraries, e.g. cDNA libraries representative of expressed sequences, may be searched. Those skilled in the art are well able to employ
-suitable conditions of the desired stringency for selective hybridisation, taking into account factors such as
oligonucleotide length and base composition, temperature and
so on .
The BBPl polypeptide, fragments, mutants, variants and
derivatives thereof, and encoding nucleic acid may be used in
the same or similar terms as fragments of BRCA2 as discussed
above, e.g. in assays. Peptide fragments may be used to
modulate transcriptional activation by BRCA2.
Further aspects and embodiments will be apparent to
those of ordinary skill in the art upon consideration of the
above disclosure and the following experimental report,
presented by way of illustration of embodiments of the present invention and without limitation.
All documents mentioned in this specification are hereby
incorporated herein by reference.
EXPERIMENTAL EXEMPLIFICATION
The BRCA2 gene encodes a large 3418 residue protein of unknown function which is found mutated in 45% of familial
breast cancers (1) . The present inventors have scrutinised the BRCA2 protein sequence for any similarity to proteins of
known function. It has surprisingly been found that exon 3
sequences at the N-terminus of BRCA2 (within a region highly
conserved between human and mouse) show sequence similarity
to the activation domain of c-jun (Figure 1) . The homology
overlaps the delta region of c-jun which contains the binding
site for the Jun N-terminal Kinase (JNK, (2)) . In view of
the similarity of exon 3 sequences to a transcriptional activation domain, the inventors investigated whether this
region of BRCA2 has transcriptional activation capacity.
Surprisingly, it has been found that BRCA2 sequences
spanning exon 3 (23-105) are able to activate transcription
in yeast, when linked to the lexA DNA binding domain. In
contrast, all the other highly conserved domains of BRCA2 (919-1171, 1500-1589, 2034-2223 and 3200-3326) do not show
any activation potential in this assay. These various domains were fused to the lexA DNA binding domain and used to
drive the activity of a promoter including a lexA binding site, which was linked to the B-gal gene. The results are
shown in Table 1.
BRCA2 exon 3 sequences (18-105) also have potent activation potential in two different mammalian cell lines,
TJ20S (Figure 1) and NMuMG (data not shown) when these
sequences are linked to the GAL4 DNA binding domain.
Fusions of portions of the BRCA2 protein and the DNA
binding domain of GAL4 (1-147) were co-transfected into U20S
cells along with a target promoter 5GAL4EIBCAT and CAT
activity was then measured 24 h following transfection. The
activity shown represents the relative value compared to the
activity of the GAL4 DNA binding domain alone. The results represent an average of several independent experiments.- The
expression levels of the different constructs were
established by western blotting using a GAL4 specific
antibody.
The c-jun homology region (60-105) contributes to this activation potential but has relatively little independent
activity, whereas the adjacent region (18-60) remains
significant, although reduced activation capacity. Residues
18-60 therefore represent a primary activating region (PAR)
whereas residues (60-105) represent an auxiliary activating region (AAR) .
Within the PAR lies a tyr residue at position 42 which is found mutated to cys in familial breast cancers (3) (shown in Figure 2) . Introduction of this tyr to cys mutation into PAR severely compromises its activation potential.
Further characterisation of this region reveals that the activation potential within exon 3 is under negative control
of inhibitory regions (IRl and IR2) present directly N- and
C-terminal to exon 3. Together these small regions
completely mask the activation potential of BRCA2. This type
of regulation by inhibitor regions has been shown to be
operational in a number of transcription factors (4,5,6) and
in the case of c-Fos, inhibitor function has been
demonstrated in the context of the intact protein assayed on
its natural binding site (4) .
These results provide the first insight into the
potential function and regulation of the BRCA2 protein. They provide indication that (a) BRCA2 has the ability to
stimulate transcription, (b) its activation potential is
under negative control by inhibitory sequences and (c) signal transduction pathways culminating in the stimulation of JNK-
like kinases may regulate BRCA2 activity.
To date two genes, BRACl and BRCA2 , have been implicated
in the generation of familial breast cancers. Although these two proteins have no apparent sequence similarity,
circumstantial evidence relating mainly to their size and overlapping expression patterns, has raised the possibility that their functions may be related (7) . Our finding that
-BRCA2 , like BRACl (8,9) has transcriptional activation potential provides the first functional evidence to support
this. Indeed, the fact that the mutations found naturally in
breast cancers disrupt the activation potential of both BRACl
or BRCA2 , argues that compromising this activity may be an
important step in the generation of familial breast cancers.
This is further supported by the Nordling et al . finding
noted above, i.e. that a large deletion disrupting the exon 3
transcription activator domain of BRCA2 is the disease-
causing mutation in a Swedish breast/ovarian cancer family.
Using two hybrid screens, polypeptide regions which
interact with BRCA2 fragments according to the present
invention are identified. Polypeptides or peptides including
such regions may be included in screens and assays as
discussed above and may be used to modulate BRCA2
transcriptional activation. Furthermore, they may themselves
be built into screens and assays such as two hybrid assays to obtain and/or identify molecules which modulate or interfere with their interaction with BRCA2 and/or their action on BRCA2 transcriptional activation. Identification of a protein which binds the BRCA2 PAR and ^ modulates i ts transcriptional activi ty, and cloning of
encoding nucleic acid
In two hybrid screens (see below) using the BRCA2 PAR a
protein was identified which binds the PAR.
The protein has been termed "BBPl" - BRCA2 Binding
Protein 1.
An initial partial encoding sequence was obtained and is
shown in Figure 4. More encoding sequence of 2.2kb was obtained. Northerns
showed that the BBPl message is about 7.5kb, and expressed highly in ovary and testes, where BRCA2 is also highly
expressed (comparisons with other tissues) .
De-Tions ra ion that BBPl modulates transcription activi ty of
the BRCA2 TAD
A portion of the BBPl protein as shown in Figure 4 was
expressed in mammalian cells as above and shown to repress activity of the BRCA2 activation domain.
The BRCA2 TAD was linked to the GAL4 DNA binding domain
and a reporter gene under control of a promoter including GAL4 sites included in the cells.
Titration in of increasing concentrations of the BBPl expression vector gave increasing repression of expression of
-the reporter construct.
Mapping of interaction si te for BBPl in BRCA2 Overlapping fragments of BRCA2 TAD were used to map the
BBPl binding site to within amino acids 18-46 of BRCA2. The
overlap is a highly conserved region in BRCA2 between human
and mouse and is where the noted mutation associated with
breast cancer lies (residue 42) .
This provides ample basis for screens and assays for
substances which interfere with interaction between BBPl and BRCA2 and have an effect on BRCA2 transcriptional activation.
The following describes two hybrid assay techniques for yeast . When performing a two hybrid assay to look for
substances which interfere with the interaction between two
polypeptides or peptides it may be preferred to use mammalian cells instead of yeast cells. The same principles apply and similar methods are well known to the person skilled in the
art .
Two Hybrid Screens for Interacting Molecules The following method is used to isolate molecules which ^interact with a BRCA2 polypeptide or peptide in accordance with the present invention.
The yeast two hybrid system is based on a protein
interaction assay in yeast (Fields and Song. 1989. Nature
340, 245-246). The following protocol contains several
modifications of the original Fields strategy and facilitates
large scale library screens. It has been designed and
optimised and was first used by Ann Vojtek to isolate c-raf
and a-raf clones (Vojtek et al . 1993. Cell 74, 205-214).- The
method described below is essentially identical to the one
outlined in Vojtek et al . It uses the same set of vectors (the different bait constructs are described below) and yeast strains. The Two Hybrid System is based on an in vivo yeast
protein interaction assay.
In general yeast are transformed with a reporter gene
construction which expresses a selective marker protein (e.g. encoding β-galactosidase) . The promoter of that gene has
been designed such that it contains binding site for the LexA
DNA-binding protein. Gene expression from that plasmid is
usually very low. Two more expression vectors are transformed into the yeast containing the selectable marker expression plasmid. The first of those two vectors is based on pBTM116. It contains the coding sequence for the full
JLength LexA gene linked to a multiple cloning site. This multiple cloning site is used to clone a gene of interest,
i.e. encoding a BRCA2 polypeptide or peptide in accordance with the present invention, in frame on to the LexA coding
region. The second yeast expression vector contains the
activation domain of the herpes simplex transactivator VP16
fused to random sequences of a cDNA library or a library of
sequences encoding peptides with diverse e.g. random
sequences (depending on whether the aim is to obtain a
naturally occurring ligand for the BRCA2 polypeptide or
peptide or to screen for peptides which interact) . Those two plasmids facilitate expression from the reporter construct
containing the selectable marker only when the LexA fusion construct (bait) interacts with a polypeptide or peptide
sequence derived from the cDNA or peptide library.
A modification of this when looking for peptides which
interfere with interaction between a BRCA2 polypeptide or
peptide and a ligand or other binding molecule, e.g. for the TAD or for IRl and/or IR2 , employs the BRCA2 polypeptide or
peptide as a fusion with the LexA DNA binding domain, and the binding molecule as a fusion with VP60, and involves a third
expression cassette, which may be on a separate expression vector, from which a peptide or a library of peptides of diverse and/or random sequence may be expressed. A reduction in reporter gene expression (e.g. in the case of β-
galactosidase a weakening of the blue colour) results from
the presence of a peptide which disrupts the interaction
between the BRCA1 polypeptide or peptide and the binding molecule, which interaction is required for transcriptional
activation of the β-galactosidase gene.
As noted, instead of using LexA and VP60, other similar
combinations of proteins which together form a functional
transcriptional activator may be used, such as the GAL4 DNA
binding domain and the GAL4 transcriptional activation
domain .
Two Hybrid Plasmids
Amino Acids 1-197 of a human BRCA2 clone are amplified
by PCR and cloned as in frame with the LexA gene in pBTM116,
which contains the TRP1 gene which allows selection of
transformed yeast on tryptophan negative plates.
The pVP16 library vector carries the LEU2 gene which allows selection on Leucine negative plates. A human cDNA library cloned next to the activation domain of VP16 is
generated by random primed synthesis of poly A+ RNA. The vast majority of inserts have a length of 400 - 600
nucleotides .
Two reporter constructs are in use and both are provided
by the yeast strain L40. The first construct has a
selectable marker, the LYS2 gene, which allows growth on
Lysine negative plates. It contains the coding region for
the histidine gene under the control of a promoter containing
four binding sites for the LexA operator. The second
reporter gene has a URA3 gene as selectable marker which
allows growth on uracil negative plates. It contains the
coding region for the lacZ gene controlled by a promoter
containing eight binding sites for the lexA protein.
Yeast Transformation Small Scale Transformation
10 ml of YPAD are inoculated with a colony of L40 and
incubated overnight at 30°C. Thereafter, the culture is
diluted to an OD600 of around 0.4 in 50 ml YPAD and grown for
an additional 2-4 hours. Cells are then pelleted at 2500 rpm
at room temperature and re-suspended in 40 ml TE . Yeast are then repelleted at 2500 rpm and resuspended in 2 ml of 100 mM
LiAc in 0.5 x TE. This yeast suspension is incubated at room temperature for 10 minutes. 1 μg of plasmid DNA together with 100 μg of sonicated sheared salmon sperm DNA is mixed
with 100 μl of the yeast suspension. After a further addition of a buffer of 700 μl containing 100 mM (LiAc) , 40%
PEG-3350 in 1 x TE, the solution is mixed well and incubated
at 30°C for 30 minutes. To stop the transformation process
88 μl DMSO is added and the mixture heat- shocked at 40°C for
7 minutes. Cells are pelletted in a microfuge for 10 seconds and re-suspended in 1 ml TE . Cells are then re-washed in 1
ml TE and re-suspended in 50-100 μl TE and plated on
selective plates. Plates are incubated at 30°C and colonies
picked after 2-3 days.
Small scale transformation may be used to test the
induction of the beta-galactosidase activity by the
LexA/BRCA2 fragment fusion plasmid. The beta-galactosidase
filter assay is described below.
Larσe Scale Library Transformation
The LexA/BRCA2 fragment fusion plasmid is introduced
into L40 by selecting for growth on tryptophan minus plates
after a small scale transformation. The resulting strain is
used to grow a 2 ml overnight culture in yeast selective medium minus tryptophan and minus uracil. Thereafter, the culture is diluted with 100 ml of the same medium. The next day the mid log phase culture is used to inoculate 1 litre of YPAD medium (pre-warmed to 30°C) . The optical density at 600
nm should be about 0.3. This culture is grown at 30°C for 3
hours. During this time the cells should roughly double in
number. Yeast are pelletted at 2500 rpm for 5 minutes at
room temperature and re-suspended in 500 ml of TE . After a
re-spin the cells are taken up in 10 ml of 100 mM Li Ac in
0.5 x TE. To this a mixture of 0.5 ml of 10 mg/ml denatured
salmon sperm DNA and 200 μg of library plasmid is added. The
suspension is mixed well. After this 70 ml of a solution
containing 100 mM LiAc, 40% PEG-3350 in 1 x TBE is added and
mixed well. This mixture is incubated for 30 minutes at
30°C. The transformation mixture is then transferred to a
sterile 2 litre beaker and 8.8 ml of DMSO was added. After mixing the suspension is heat shocked at 42°C in a water bath
for 6 minutes. Thereafter, the suspension is diluted with
200 ml of YPA and rapidly cooled to room temperature in a
water bath. Cells are then pelletted at 2500 rpm for 5 minutes at room temperature, washed with 250 ml YPA medium
and re-suspended in 1 litre of pre-warmed YPAD medium. At this stage incubation at 30°C is allowed for 1 hour with
gentle shaking. The culture is then pelletted at 2500 rpm for 5 minutes at room temperature and re-suspended in 500 ml of selective medium omitting uracil, tryptophan leucine (-
UTL) . After a further respin cells are resuspended in 1
litre of pre-warmed -UTL medium with shaking at 30°C for
about 4 hours. Thereafter, cells are pelletted at 2500 rpm for 5 minutes at room temperature and washed twice with
selective medium omitting tryptophan, histidine, uracil and
leucine (-THULL). The final pellet is resuspended in 10 ml
of -THULL medium and plated in aliquots of 100 μl on plates
made from -THULL media. If the bait alone can activate the
β-galactosidase gene, this is suppressed by the inclusion of
3 amino 124-triazole at an appropriate concentration (e.g.
50mM) . After 2-3 days colonies are picked to a grid. A
nitrocellulose filter lift is used in a beta-galactosidase
filter assay for analysis of lacZ induction.
Beta-galactosidase Filter Assay
Filters are removed from the plates and immersed for 3-5 seconds in liquid nitrogen. Filters are then placed, colony
side up, at room temperature until thawed. The beta- galactosidase assay is performed in the lid of a petri dish. 3 ml of Z-buffer (60 mM Na2HP04, 40 mM NaH2P04, 10 mM KC1, 1
mM MgS04, pH7.0) containing 30 ml of 50 mg/ml X-gal . Circularised Whatman filters (#1) are placed into the buffer, followed by the nitrocellulose filters, colonies facing up.
The lid is then covered with the bottom of the petri dish and
placed at 30°C. Interactions are detectable by the
appearance of a blue colour after 20 to 40 minutes.
Recovering of Plasmids from Yeast and Shuttling into E. coli
Viable cells are recovered from colonies and grown in a
50 ml overnight culture with the appropriate selection. The
next morning cells are pelletted at 2500 rpm for 5 minutes at
room temperature. Pellets are resuspended in 0.3 ml of lysis
buffer (2.5 M LiCl, 50 mM Tris-Cl (pH 8.0), 4% Triton X-100,
62.5 mM EDTA) . At this stage solution is transferred to 1.5
ml tubes and 150 ml of glass beads (0.45 - 0.50 mm) together with 0.3 ml phenol/chloroform are added. After vigorous shaking for 1 minute samples are centrifuged for 1 minute and
the aqueous phase transferred to a new tube. DNA is
precipitated twice with ethanol and resuspended in 25 ml TE followed by electroporation of DNA into E.coli.
Verification of Interacting Partners
Recovered library plasmids from positive yeast colonies
are retransformed into the L40 strain containing the LexA/BRCA2 fragment bait vector using the small scale transformation procedure. Using the beta-galactosidase filter assay positive colonies are identified with the
LexA/BRCA2 fragment bait. No induction of beta-galactosidase
activity is detected in colonies transformed with a LexA
instead of a LexA/BRCA 2 fragment clone. The underlying
protein interactions of the positive colonies are significant and consequently further analysed.
Identification of a Full Length cDNA of A BRCA2 Binding
Molecule
In order to isolate a full length cDNA clone of a
molecule which binds BRCA2 TAD, IRl and/or IR2 , a plasmid
cDNA library is plated on ampicillin resistant plates.
Nitrocellulose filter lifts of those plates are hybridised to
DNA sequences obtained through the yeast two hybrid screen. Washes are done at high stringency and positive signals are identified. Isolated DNA is sequenced and if necessary partial clones are ligated to provide a full length coding
sequence. A full length sequence may be verified as such by
primer walking sequencing in both directions.
Identification of a kinase able to bind the activation domain of BRCA2 Using residues 18-141 of BRCA2 as an affinity column a
kinase has been purified from HeLa nuclear extracts which phosphorylates within amino acids 60-105 of BRCA2 ,
specifically the region of BRCA2 which is homologous to the
JNK binding site in c-jun. This was demonstrated by deletion of residues 80-107 in BRCA2 , which abolished binding of the
kinase. This mutation also reduced the activation capacity
of BRCA2. The region of BRCA2 residues 60-105 is
phosphorylated in vivo .
The kinase was found not to be stimulated by uv and is
therefore not JNK. This was confirmed by experiments in
which recombinant JNK was found not to bind the region of
BRCA2.
REFERENCES 1. Wooster, R. et al . (1994). Science, 265, 2088-2090.
2. Derijard, B. et al . (1994). Cell, 76, 1025-1037.
3. Friend, S. and the Breast Cancer Information Core Steering Committee (1995) . Breast cancer information on
the web. Nature Genetics 11: 238-239.
4. Brown, H.J. et al . (1995). EMBO J. , 14, 124-131.
5. Dennig, J. et al . (1996). EMBO J., 15, 5659-5667.
6. Dubendorff, J.W. , et al . (1992). Genes & Development, 6, 2524-2535. 7. Thakur, S. et al . (1997). Mol & Cell. Biol . 17, 444-
452.
8. Chapman, M.S. and Verma, I.M. (1996). Nature, 382, 678-
679.
9. Varo, A. et al . PNAS, 93, 13595-13599.
10. Nordling, M. , et al . , Cancer Research, 58, 1372-1375,
April 1, 1998
TABLE 1 shows the results of a β-galactosidase liquid assay
using various fragments of BRCA2 fused to the DNA binding
domain of lexA.
TABLE 1
bait activity TUl
18-105 112±2
919-1171 (BRC1) 0
1500-1589 (BRC4) 0
2034-2223 (BRC8) 0
3200-3326 0

Claims

1. A fragment of BRCA2 which is able to act as
transcriptional activator when operably linked to a heterologous DNA binding domain.
2. A fragment according to claim 1 which is of less than
about 200 amino acids.
3. A fragment according to claim 1 or claim 2 which includes amino acids 23-105 of the human BRCA2 polypeptide
the sequence of which is shown in Figure 2.
4. A fragment according to claim 3 which includes amino acids 18-105 of the human BRCA2 polypeptide the sequence of
which is shown in Figure 2.
5. A fragment according to claim 1 or claim 2 which includes amino acids 18 to 60 of the human BRCA2 polypeptide the sequence of which is shown in Figure 2.
6. A fragment according to claim 1 or claim 2 which includes amino acids 60 to 105 of the human BRCA2 polypeptide the sequence of which is shown in Figure 2.
7. A fragment according to any of claims 1 to 6 including
amino acids 1-17 and/or amino acids 106-125 of the human BRCA2 polypeptide the sequence of which is shown in Figure 2.
5 8. A mutant, variant or derivative of a BRCA2 fragment
according to any of claims 1 to 7 , which mutant, variant or
derivative is able to act as transcriptional activator when
operably linked to a heterologous DNA binding domain and has
at least 80% sequence similarity with said fragment.
10
9. A fragment according to any of claims 1 to 7 or a
mutant, variant or derivative thereof according to claim 8 fused to a sequence of amino acids heterologous to BRCA2.
15 10. A substance including a fragment according to any of claims 1 to 7 or a mutant, variant or derivative thereof according to claim 8 operably linked to a heterologous DNA
binding domain.
20 11. An isolated nucleic acid molecule encoding a fragment
according to any of claims 1 to 7 or a mutant, variant or derivative thereof according to claim 8.
12. Nucleic acid according to claim 11 wherein said
fragment, mutant, variant or derivative is fused to a
sequence of amino acids heterologous to BRCA2.
13. Nucleic acid according to claim 11 or claim 12 operably linked to regulatory sequences for expression of the encoded
product .
14. A host cell transformed with nucleic acid according to
claim 13.
15. A method for production of a fragment according to any
of claims 1 to 7 or a mutant, variant or derivative thereof according to claim 8, the method including causing expression
from nucleic acid according to claim 13.
16. A method according to claim 15 including culturing a
host cell transformed with said nucleic acid under conditions
for expression of the encoded product.
17. A method according to claim 15 or claim 16 wherein said
fragment, mutant, variant or derivative is isolated and/or
purified.
18. A method according to claim 17 including formulating said fragment, mutant, variant or derivative into a
composition including at least one additional component.
19. A method of producing a transcription factor, the method
including operably linking a fragment according to any of claims 1 to 7 or a mutant, variant or derivative according to
claim 8 to a DNA binding domain to form a transcription
factor.
20. A method of activating transcription from a promoter
including a motif for a DNA binding domain, the method
including bringing into contact the promoter and a substance
according to claim 10 wherein said heterologous DNA binding
domain is able to bind said motif, under conditions wherein the DNA binding domain binds said motif and transcription from the promoter is activated.
21. An assay method which includes: (a) bringing into contact a substance including a
fragment according to any of claims 1 to 6 or a mutant, variant or derivative thereof according to claim 8 and a test compound; and (b) determining interaction between said substance and
said test compound.
22. An assay method which includes:
(a) bringing into contact a substance including a fragment according to any of claims 1 to 5 or a mutant,
variant or derivative thereof according to claim 8 , or a
fragment of BRCA2 including amino acids 18-46 as shown in
Figure 2, a substance including a fragment of BBPl of which
the amino acid sequence is shown in Figure 3 or Figure 4, or
a mutant, variant or derivative thereof which is able to bind
BRCA2; and a test compound, under conditions in which in the
absence of the test compound being an inhibitor, the two said
substances interact ;
(b) determining interaction between said substance.
23. An assay method which includes:
(a) bringing into contact a substance according to
claim 10 including a DNA binding domain able to bind a motif within a promoter, the method including bringing into contact
a substance according to claim 10 and a putative inhibitor compound under conditions where the substance, in the absence of inhibitor, is capable of binding the nucleotide sequence within the promoter to activate transcription;
(b) providing a nucleic acid molecule which includes a
promoter which includes the motif to which said DNA binding
domain is capable of binding to activate transcription of a
sequence operably linked to the promoter; and
(c) measuring the degree of modulation or alteration of
transcriptional activation caused by said inhibitor compound.
24. An assay method which includes:
(a) bringing into contact a substance including a
fragment according to any of claims 1 to 6 or a mutant,
variant or derivative thereof according to claim 8 and a test
compound in the presence of a kinase under conditions in
which the kinase normally phosphorylates said fragment,
mutant, variant or derivative; and
(b) determining phosphorylation of said fragment,
mutant, variant or derivative.
25. An assay method which includes:
(a) bringing into contact a substance including amino
acids 1-17 and/or 106-125, and a putative binding molecule or
other test compound; and
(b) determining interaction or binding between the substance and the test compound.
26. An assay method according to any of claims 21 to 25
wherein the test compound is a peptide fragment of BRCA2 or BBPl or a mimetic thereof.
27. A method wherein a test compound identified as testing
positive in a method according to any of claims 21 to 26 is
formulated into a composition including at least one
additional component.
28. Use of a fragment according to any of claims 1 to 7 or a
mutant, variant or derivative thereof according to claim 8 in an assay to identify a compound which is able to modulate transcriptional activation by BRCA2.
29. A peptide which is a fragment of a fragment according to
any of claims 1 to 7 or a mutant, variant or derivative thereof according to claim 8 or a fragment of BBPl the amino acid sequence of which is shown in Figure 3 or Figure 4, or a
non-peptidyl mimetic of said peptide, which peptide or mimetic thereof is able to modulate transcriptional activation by BRCA2.
30. A peptide according to claim 29 including residues 18-46
of the amino acid sequence of human BRCA2 polypeptide which
is shown in Figure 2.
31. A peptide which includes amino acids 1-17 of the human
BRCA2 polypeptide the sequence of which is shown in Figure 2.
32. A peptide which includes amino acids 106-125 of the
human BRCA2 polypeptide the sequence of which is shown in
Figure 2.
PCT/GB1998/001181 1997-04-23 1998-04-23 Brca2 transcriptional activator domain and uses thereof WO1998048013A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA002288335A CA2288335A1 (en) 1997-04-23 1998-04-23 Brca2 transcriptional activator domain and uses thereof
EP98917445A EP0977847A1 (en) 1997-04-23 1998-04-23 Brca2 transcriptional activator domain and uses thereof
AU70674/98A AU7067498A (en) 1997-04-23 1998-04-23 Brca2 transcriptional activator domain and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9708221.8 1997-04-23
GBGB9708221.8A GB9708221D0 (en) 1997-04-23 1997-04-23 Substances and uses thereof

Publications (1)

Publication Number Publication Date
WO1998048013A1 true WO1998048013A1 (en) 1998-10-29

Family

ID=10811226

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1998/001181 WO1998048013A1 (en) 1997-04-23 1998-04-23 Brca2 transcriptional activator domain and uses thereof

Country Status (5)

Country Link
EP (1) EP0977847A1 (en)
AU (1) AU7067498A (en)
CA (1) CA2288335A1 (en)
GB (1) GB9708221D0 (en)
WO (1) WO1998048013A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022689A1 (en) * 1995-12-18 1997-06-26 Myriad Genetics, Inc. Chromosome 13-linked breast cancer susceptibility gene

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022689A1 (en) * 1995-12-18 1997-06-26 Myriad Genetics, Inc. Chromosome 13-linked breast cancer susceptibility gene

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK 26 May 1996 (1996-05-26), HILLIER L. ET AL.: "EST; H. sapiens clone 324322", XP002076944 *
MILNER J. ET AL.: "Transcriptional activation functoions in BRCA2.", NATURE, vol. 386, 24 April 1997 (1997-04-24), pages 772 - 773, XP002076943 *
SHARAN S. & BRADLEY A.: "Murine Brca2: sequence, map position and expression pattern", GENOMICS, vol. 40, no. 2, 1 March 1997 (1997-03-01), pages 234 - 241, XP002076941 *
TAVTIGIAN S.V. ET AL.: "The complete BRCA2 gene and mutations in chromosome 13q-linked kindreds", NATURE GENETICS, vol. 12, no. 3, March 1996 (1996-03-01), pages 333 - 337, XP002076942 *

Also Published As

Publication number Publication date
EP0977847A1 (en) 2000-02-09
GB9708221D0 (en) 1997-06-11
CA2288335A1 (en) 1998-10-29
AU7067498A (en) 1998-11-13

Similar Documents

Publication Publication Date Title
US5691147A (en) CDK4 binding assay
AU687764B2 (en) Cyclin complex rearrangement and uses related thereto
US5889169A (en) Cell cycle regulatory protein p16 gene
US6753158B1 (en) Assays, agents, therapy and diagnosis relating to modulation of cellular DNA repair activity
US20040072268A1 (en) Methods for regulating BRCA1-BRCA2-containing complex activity
WO1998048013A1 (en) Brca2 transcriptional activator domain and uses thereof
CA2262479A1 (en) Brca1 associated protein (bap-1) and uses therefor
US6890709B1 (en) Assays, methods and means for modulating e2f activity
WO1997041433A1 (en) METHOD AND MEANS FOR DISRUPTION OF p53 AND RB INTERACTION
US7348407B2 (en) Stress-responsive activator of p300 (strap) protein
EP1105738B1 (en) Assays for modulating nuclear localisation
US6331390B1 (en) Cell-cycle regulatory proteins, and uses related thereto
WO1997035975A1 (en) E2f-like transcription repressor and dna encoding it
JP2002508180A (en) Human RAD1 nucleic acids, polypeptides, assays, therapeutic methods and means
WO2001081587A1 (en) Acetyltransferase and uses thereof
EP1222264A1 (en) Human sit4 associated proteins like (sapl) proteins and encoding genes; uses thereof
JP2000512133A (en) Identification of altered genes in multiple myeloma

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2288335

Country of ref document: CA

Ref country code: CA

Ref document number: 2288335

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 70674/98

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 1998917445

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09402610

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1998917445

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 1998545300

Format of ref document f/p: F

WWW Wipo information: withdrawn in national office

Ref document number: 1998917445

Country of ref document: EP