WO2001000677A1 - A method of modulating integrin mediated cellular activity and agents useful for same - Google Patents

A method of modulating integrin mediated cellular activity and agents useful for same Download PDF

Info

Publication number
WO2001000677A1
WO2001000677A1 PCT/AU2000/000729 AU0000729W WO0100677A1 WO 2001000677 A1 WO2001000677 A1 WO 2001000677A1 AU 0000729 W AU0000729 W AU 0000729W WO 0100677 A1 WO0100677 A1 WO 0100677A1
Authority
WO
WIPO (PCT)
Prior art keywords
integrin
map kinase
binding
polypeptide
agent
Prior art date
Application number
PCT/AU2000/000729
Other languages
French (fr)
Other versions
WO2001000677A9 (en
Inventor
Michael Valentine Agrez
Nuzhat Ahmed
Original Assignee
The University Of Newcastle Research Associates 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
Priority claimed from AUPQ1248A external-priority patent/AUPQ124899A0/en
Priority claimed from AUPQ8003A external-priority patent/AUPQ800300A0/en
Application filed by The University Of Newcastle Research Associates Limited filed Critical The University Of Newcastle Research Associates Limited
Priority to AU55106/00A priority Critical patent/AU781540B2/en
Priority to US10/019,816 priority patent/US8119594B1/en
Priority to EP00940024A priority patent/EP1196449B1/en
Priority to DE60037970T priority patent/DE60037970T2/en
Publication of WO2001000677A1 publication Critical patent/WO2001000677A1/en
Publication of WO2001000677A9 publication Critical patent/WO2001000677A9/en
Priority to AU2005204275A priority patent/AU2005204275B2/en
Priority to US13/349,129 priority patent/US8618062B2/en
Priority to US13/832,796 priority patent/US20140187612A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates to a method of modulating cell activity mediated by a
  • a cell such as a neoplastic cell.
  • a cell such as a neoplastic cell.
  • the present invention also provides agents
  • Colorectal cancer is the commonest internal malignancy affecting men and women
  • Colorectal cancer is a malignant tumour that starts in the bowel wall and is
  • Familial Adenomatous Polyposis Familial Adenomatous Polyposis (FAP) and
  • FAP and HNPCC contribute to
  • Adenomas are classified by histological architecture as tubular,
  • Villous change is associated with a higher malignant potential
  • colorectal cancer include diets low in fibre and vegetables and high in
  • tumour suppressor genes such as the Adenomatous
  • Polyposis Coli (APC) gene or Mutated in Colon Cancer (MCC) gene can proliferate and
  • tumour suppressor genes such as p53 and the Deleted in Colon Cancer gene (DCC) may
  • neoplasia development of neoplasia is believed to result, therefore, from a series of inherited and
  • tumour cell extravasation and proliferation at metastatic sites Detachment of cells from
  • tumour cells into blood and lymphatic vessels. It is the invasive and
  • tumourigenesis can be viewed as a tissue
  • Cell adhesion receptors on the surface of colon cancer cells are involved in complex cell signalling which may regulate cell proliferation, migration, invasion and
  • tumour cells cell and cell-matrix binding, the latter involving attachment of tumour cells to
  • extracellular scaffolding molecules such as collagen, fibronectin and laminin.
  • Integrins are involved in several fundamental fundamentals
  • transmembrane glycoproteins consisting of an alpha ( ⁇ ) and beta ( ⁇ ) chain in close
  • the integrin family comprises 17
  • the ⁇ l chain combines with any one of nine ⁇ chain members
  • ⁇ 2 ⁇ l binds collagen and laminin
  • ⁇ 3 ⁇ l binds
  • the abundant and promiscuous ⁇ v chain combines with any one of five ⁇ chains, and a distinguishing feature of ⁇ v integrins is that they all recognise and bind
  • This head containing the
  • the ⁇ -subunits have a molecular mass ranging
  • the ⁇ subunit cytoplasmic domain is required for linking integrins to the
  • FAK focal adhesion kinase
  • tumours including the regulation of proliferation and apoptosis.
  • the integrin ⁇ 6 subunit was first identified in cultured epithelial cells as part of the
  • the ⁇ 6 subunit is composed of 788 amino acids
  • subunit also contains 9 potential glycosylation sites on the extracellular domain
  • the cytoplasmic domain differs from other ⁇ subunits in that it
  • the integrin ⁇ v ⁇ 6 has previously been shown to enhance growth of colon cancer
  • integrin of particular interest in colon cancer is that it is not expressed in normal cells but
  • tumourigenesis is highly expressed during tumourigenesis (Breuss et al, 1995; Agrez et al, 1996).
  • tumour cells likely to reflect the ability of tumour cells to digest their surrounding matrix scaffold
  • matrix-degrading enzymes such as matrix metalloproteinases
  • MMPs The mechanisms whereby human colon cancer cells escape the constraints on
  • Integrins can signal through the cell membrane in either direction.
  • extracellular binding activity of integrins can be regulated from the cell interior as, for
  • ECM extracellular matrix
  • the first is 'direct
  • adhesion to ECM proteins can activate cytoplasmic tyrosine kinases (e.g. focal
  • APK activated protein kinase
  • PjP 2 5-biphosphate
  • MAP kinases behave as a convergence point for diverse receptor-initiated
  • the core unit of MAP kinase pathways is a
  • MEKs MAP kinase kinases
  • MAP kinase family are the extracellular signal-regulated kinases (ERKs) (Boulton et al,
  • serine/threonine kinases phosphorylate and modulate the function of many proteins with
  • upstream regulators such as the
  • MAP kinases can be activated through non-receptor tyrosine kinases such as focal
  • FAK adhesion kinase
  • cytoplasmic tyrosine kinase ⁇ 60 c-srk
  • the FAK pathway is activated by most integrins. In addition to activating
  • activate ERK may be especially important when the concentration of growth factors
  • PLC protein kinase C
  • PKC has been shown to regulate integrin-induced activation of the MAP kinase pathway
  • proteins such as ⁇ -actinin and talin and ⁇ l and ⁇ 3 integrin tails (Horwitz et al, 1986;
  • ILK integrin-linked kinase
  • integrin-binding molecules include the serine/threonine integrin-linked kinase, ILK,
  • ILK has
  • extracellular Ig region of the IAP molecule mediates association with ⁇ v ⁇ 3 and is
  • Integrins have also been shown to be physically linked with matrix-degrading
  • integrin ⁇ v ⁇ 6 has been shown to bind
  • PDGF platelet-derived growth factor
  • PDGF ⁇ -receptor is tyrosine phosphorylated upon growth factor stimulation.
  • this subset can associate with the ⁇ v ⁇ 3 integrin, and PDGF activity is
  • the present invention relates to modulation of integrin expression
  • MAP mitogen activated protein
  • the agent will be capable of binding with a binding site on the MAP
  • the agent may be capable of binding to the binding domain on the integrin for the MAP
  • the agent may be provided either isolated or for instance, coupled to another
  • binding domain on an integrin for the MAP kinase or a homolog, analog, variant or
  • the polypeptide will comprise the binding domain of the integrin or
  • polypeptide with the MAP kinase polypeptide with the MAP kinase.
  • polypeptide will comprise amino acid sequence
  • RSKAKWQTGTNPLYR more preferably RSKAKNPLYR, or one or both amino acid
  • polypeptide will be a fragment of
  • the integrin subunit is other than a ⁇ 6(770t) or ⁇ 6(777t) deletion mutant.
  • the polypeptide or fragment will have a length of about 150 amino acids
  • the length will be between about 5 to about 30
  • the fragment may comprise an amino acid sequence incorporating extracellular
  • the fragment will be a
  • polypeptide or fragment will be coupled to a carrier polypeptide for
  • the carrier polypeptide will be any polypeptide that will facilitate entry of the fusion protein into a cell.
  • the carrier polypeptide will be any polypeptide that will be derived from the same or different amino acids.
  • nucleic acid sequence encoding a fusion protein of the invention.
  • antisense nucleic acid sequences complimentary to the
  • oligonucleotides include oligonucleotides. Sense oligonucleotides coding for the binding domain of an
  • antisense oligonucleotides find particular application as primers or probes.
  • the vector will be an
  • expression vector and the nucleic acid sequence will be capable of being transcribed.
  • the host cell will be selected from the group consisting of a mammalian
  • the host cell will first construct an epithelial cell, a neoplastic cell, and a cancer cell.
  • the host cell will be any cell that will be used to generate a cancer cell.
  • the antibody may be a polyclonal or monoclonal antibody.
  • the antibody may be a polyclonal or monoclonal antibody.
  • antibody is a monoclonal antibody.
  • binding site binds with a binding domain of an integrin for the MAP kinase, comprising:
  • the molecule may be the integrin, or a fusion protein, a polypeptide, or a fragment
  • MAP kinase is capable of binding, or for instance an
  • the MAP kinase may be any suitable MAP kinase.
  • the MAP kinase may be any suitable MAP kinase.
  • integrin for the MAP kinase comprising: (a) testing a number of agents for ability to bind to either the MAP kinase or the integrin;
  • MAP kinase for the MAP kinase, comprising:
  • the or any of the selected said agents is capable of inhibiting the binding of the MAP
  • an agent is capable of inhibiting binding of a MAP kinase to a binding domain of an
  • integrin for the MAP kinase comprising:
  • integrin and thereby inhibit binding of the MAP kinase to the integrin, may comprise
  • kinase Rather than using the integrin, a polypeptide or fragment of the invention or
  • the integrin may be used.
  • the testing or assaying may comprise exposing
  • the or any of the selected said agents is capable of binding to the binding domain on the
  • a MAP kinase comprising:
  • polypeptide or fragment of the invention consisting of the binding
  • polypeptide or fragment will consist of the amino acid sequence
  • An agent of the invention will usually be provided in the form of a pharmaceutical
  • composition comprising an agent of the invention capable of inhibiting
  • the agent may or not be proteinaceous in nature.
  • the agent will be any suitable pharmaceutically acceptable carrier or diluent.
  • the agent may or not be proteinaceous in nature.
  • the agent will be any suitable pharmaceutically acceptable carrier or diluent.
  • the agent will be any suitable pharmaceutically acceptable carrier or diluent.
  • the agent may or not be proteinaceous in nature.
  • the agent will be any suitable pharmaceutically acceptable carrier or diluent.
  • the agent will be coupled to
  • a carrier molecule for facilitating entry of the agent into a cell.
  • nucleic acid sequence of the invention comprising a nucleic acid sequence of the invention and a pharmaceutically acceptable
  • nucleic acid sequence is incorporated into a vector as
  • nucleic acid sequence may be joined to a carrier
  • modulating activity of a cell comprising:
  • the integrin subunit for being expressed by the cell, wherein the integrin subunit has a mutagenised binding domain for a MAP kinase or in which the binding domain is deleted, or a
  • modulating activity of a cell comprising:
  • the polypeptide is capable of inhibiting binding of a MAP
  • the polypeptide will be capable of binding with the binding site on the
  • kinase is able to bind to be down-regulated.
  • down-regulation of the expression of the integrin is achieved using an
  • antisense nucleic acid sequence that inhibits expression of the gene encoding the
  • the antisense nucleic acid sequence may be administered to the cell or
  • the cell will be transformed with a vector
  • the antisense nucleic acid sequence will specifically hybridise with the
  • activity of a cell comprising contacting the cell with an effective amount of an agent for
  • activity of a cell comprising: contacting the cell with an effective amount of an agent capable of binding to
  • MAP kinase to thereby inhibit binding of the MAP kinase to a binding domain on the
  • the agent will be capable of binding to the binding site on the MAP
  • cell activity is desirable, comprising:
  • binding domain for a MAP kinase or in which the binding domain is
  • cell activity comprising: administering to the mammal an effective amount of a nucleic acid sequence encoding a polypeptide for being expressed, wherein the polypeptide is capable of
  • nucleic acid of the invention in another aspect there is provided use of a nucleic acid of the invention in the
  • the cellular activity desired to be down-regulated will typically but not
  • the cell may be any cell type in which functional activity of a MAP kinase arising
  • the cell will be an epithelial cell
  • a neoplastic cell By modulating the activity a neoplastic cell,
  • the MAP kinase will be selected from the group consisting of an
  • extracellular signal-regulated kinase ERK
  • JNK MAP kinase JNK MAP kinase
  • MAP kinase is ERK2 or JNK-1. Most preferably, the MAP kinase is ERK2.
  • the mammal may be any mammal treatable with a method of the invention.
  • the mammal may be a member of the bovine, porcine, ovine or equine families,
  • a laboratory test animal such as a mouse, rabbit, guinea pig, a cat or dog, a primate or a
  • the mammal will be a human being.
  • an integrin subunit or an integrin family member incorporating at least one such
  • the integrin is or incorporates an integrin subunit selected from the group consisting of ⁇ 3, ⁇ 5 and ⁇ 6.
  • the integrin is or incorporates an integrin subunit selected from the group consisting of ⁇ 3, ⁇ 5 and ⁇ 6.
  • integrin comprises ⁇ 6.
  • binding domain is meant the minimum length of contiguous amino acid
  • binding as distinct from any amino acids that do not directly participate in the binding
  • sequence similarity The homology between amino acid sequences can be determined by
  • sequences are considered homologous at a position if the
  • amino acid sequence homology of at least about 30% more preferably at least about 50%
  • Homology with the binding domain of an integrin may be greater than the overall amino
  • analog is to be taken to mean a molecule that has one or more aspects
  • analog may have substantial overall structural similarity with the molecule or only structural similarity with one or more regions or domains thereof responsible for the
  • an analog may not be proteinaceous at all.
  • An analog may for
  • variable is meant an isoform of an integrin subunit, an integrin
  • a variant may be prepared by introducing nucleotide
  • substitution of an amino acid may involve a conservative or non-conservative
  • Preferred variants include ones having amino acid sequences in which one or more
  • amino acids have been substituted with alanine or other neutrally charged amino acid
  • a variant may also be any variant of residue(s), or to which one or more such residues have been added.
  • a variant may also be any variant of residue(s), or to which one or more such residues have been added.
  • a variant may also be any variant of residue(s), or to which one or more such residues have been added.
  • derivative is meant a molecule that is derived or obtained from
  • a derivative may for instance arise as a result of the cleavage
  • a derivative may also result from post-translational or
  • reaction(s) resulting in structural modification(s) such as the alkylation or acetylation of
  • polypeptide is used interchangeably herein with “peptide” and
  • Neoplastic cell is to be taken to mean a cell exhibiting abnormal growth
  • Figure 1 Surface biotinylation and immunoprecipitation of integrin subunits ⁇ 5
  • mRNA Ethidium-stained agarose gels with amplification products following RT-PCR
  • PCR product obtained from RT-PCR reactions were loaded on each lane and the ⁇ 6 (141 basepairs) and GAPDH (216 basepairs) bands are indicated (WT, wild-type; S, sense ⁇ 6;
  • Figure 3 Non-transfected HT29 cells (wild) and cells transfected with vector alone
  • Figure 4 WiDr cells transfected with vector alone (mock) or antisense ⁇ 6 and
  • Figure 8 Graph showing average tumour size at weekly intervals for each
  • Figure 10 ⁇ 6-associated ERK identified by immunoprecipitafions of integrin
  • ERK mAb (SC-1647) against total ERK. Purified non-phosphorylated ERK2 is shown in
  • Figure 12 ⁇ 6-bound ERK shown for the high and low SW480 ⁇ 6-expressing
  • FIG. 14 Non-transformed (HaCaT) and Ras-transformed (HaRas) human
  • keratinocytes ⁇ 6 immunoprecipitation and ERK western blots probed with monoclonal
  • SW480 ⁇ 6 and SW480 mock transfectants full grey and hatched bars, respectively.
  • MAP kinase activity is shown as the mean of three independent experiments.
  • Figure 16 Shows the amino acid sequence of the cytoplasmic domain of the ⁇ 6
  • Figure 17 Graph showing binding of non-phosphorylated ERK2 (GST.ERK2) to
  • Figure 18 Graph showing binding of ERK2 (GST.ERK2) to the ⁇ 6 cytoplasmic
  • Figure 19 Graph showing binding of ERK2 (GST.ERK2) to the ⁇ 6 cytoplasmic
  • Figure 20 Graph showing binding of ERK2 (GST.ERK2) to a 15 mer fragment of
  • Figure 21 Shows regions of the cytoplasmic domain of the ⁇ 6 subunit
  • Figure 22 Graph showing assay results for ERK2 (GST.ERK) binding to
  • Figure 23 Graph showing binding of ERK2 (thrombin cleaved) to synthesised
  • Figure 24 Graph showing binding of JNK-1 to the ⁇ 6 cytoplasmic domain.
  • Figure 25 location of ⁇ 6 ⁇ 746-764, ⁇ 6(770t) and ⁇ 6(777t) deletions in the
  • FIG. 26 SW480 cells transfected with wild-type full length coding sequence for
  • FIGS. 27(A) and 27(B) (A) Western blotting: equal protein loads of cell lyates
  • mAb R6G9 immunoprecipitates from equal protein loads of cell lysates (A) probed with anti-ERK mAb (ElO).
  • Figure 28 Proliferation of HT29 colon cancer cells cultured for 48 hours and
  • Figure 34 Graph showing binding of ERK2 to RSKAKWQTGTNPLYR peptide
  • ⁇ 6 expression is restricted to epithelia and is up-regulated in parallel with morphogenetic
  • tumourigenesis a malignant neoplasm originating from tumourigenesis.
  • epithelial repair a malignant neoplasm originating from tumourigenesis.
  • ⁇ 6 is expressed by specific types of epithelial cells
  • ⁇ 6 is almost invariably neo-expressed in squamous cell carcinomas derived
  • ⁇ 6 is also up-regulated in migrating keratinocytes at the wound edge
  • TGF- ⁇ l has been shown to induce the de novo expression of ⁇ v ⁇ 6 at
  • ⁇ v ⁇ 6 expression is also upregulated in type II alveolar epithelial cells during lung
  • TGF- ⁇ l has been shown to dramatically increase expression of
  • EGF growth factor
  • tumour cell islands have been observed at the invading edges of tumour cell islands (Agrez et al,
  • degrading enzymes as has been previously suggested for invasive melanoma cells.
  • the ⁇ subunit is widely observed in cancers of various origins (Breuss et al,
  • ⁇ 6 is detected in at least 50% of bowel cancer tumours.
  • ⁇ 6 expression is similarly maximal at the advancing edges of
  • tumour cell islands (Agrez et al, 1996).
  • cells may be used in the prophylaxis or treatment of cancer of the lip, tongue, salivary
  • hypopharynx and other oral cavities oesophagus, stomach, small intestine, duodenum,
  • a method of the invention may find application in
  • breast cancer susceptibility gene BRCA1 or BRCA2 associated with breast cancer the breast cancer susceptibility gene BRCA1 or BRCA2 associated with breast cancer
  • the epithelial restricted integrin subunit ⁇ is shown herein to interact with at least
  • tumour cells be significantly upregulated on tumour cells compared to normal cells offers the
  • tumour cell specificity substantially without impairment of normal cellular
  • the integrin molecule or by inhibiting the signalling function of the integrin.
  • Gene therapy is one strategy for treating cancers of different types.
  • recombinant adenoviruses has for instance been utilised for restoring expression of
  • polypeptide encoded by a wild-type p53 tumour suppressor gene (Bookstein et al, 1996)
  • tumoural injection of recombinant virus (Spitz et al, 1996). Rather than replacing a defective gene with one encoding a polypeptide the
  • the binding domain has been wholly or partially deleted, to thereby achieve down
  • the defective integrin subunit and be expressed on the cell membrane.
  • integrin subunit will be expressed at a higher level than the corresponding wild-type
  • such therapy may involve the introduction and expression of a nucleic acid
  • Another option is to introduce a nucleic acid sequence encoding a polypeptide
  • a gene or nucleic acid sequence encoding an integrin subunit may also be
  • amino acid sequence of a region of the integrin subunit distant from the binding domain or the amino acid sequence of either one or both regions flanking the
  • binding domain is altered to achieve a change in the three dimensional conformation of
  • the gene or nucleic acid sequence may be altered to achieve the desired outcome
  • MAP kinase to the integrin is inhibited. Inhibition in this context may be partial or total
  • the gene or nucleic acid sequence can be introduced into a cell in an appropriate
  • nucleic acid sequences e.g. oligonucleotides
  • nucleic acid construct incorporating all or
  • a method of down regulating an activity of a cell may comprise
  • first nucleic acid molecule that is capable of interacting with a target nucleic acid sequence, or which first nucleic acid molecule is capable of being
  • nucleic acid molecule capable of interacting with the target sequence
  • nucleic acid molecule which directly or indirectly facilitates reduction, inhibition or
  • target sequence such as mRNA encoding the integrin subunit or
  • molecule may also be a ribozyme capable for instance, specifically binding to that region
  • nucleic acid encoding the binding domain of an integrin subunit and cleaving the
  • vitronectin-binding integrin ⁇ v ⁇ 5 promotes adenovirus internalisation
  • a secondary benefit of inhibiting ⁇ expression during the course of therapy may be the concomitant rise of ⁇ 5 in cells already transduced with antisense ⁇ nucleic
  • an antisense nucleic acid sequence will hybridise with all or part of that
  • nucleic sequence may for instance be capable of hybridising to exon and/or intron
  • an antisense nucleic acid sequence will be any nucleic acid sequence.
  • an antisense nucleic acid sequence will be any nucleic acid sequence.
  • antisense nucleic sequence will have a complementarity of about 70% or greater, more
  • Preferred antisense sequences are oligonucleotides wherein the complementary sense
  • nucleotides encode for about 50 amino acids or less, preferably about 35 or 30 amino
  • Antisense nucleic acid sequences may be generated in vivo by transcription of a
  • Antisense sequences will desirably be designed to be resistant to endogenous exonucleases and/or endonucleases to provide in vivo stability in target
  • nucleotides and substituted sugar moieties linkage to liphophilic moieties and the such
  • Gene transfer methods known in the art include viral and non-viral
  • adenovirus packaged for delivery to target cells
  • adenovirus (Berkner, 1992; Gorziglia and
  • HSV Herpes Simplex Virus
  • EBV Margolskee, 1992; Johnson et al
  • poliovirus particularly preferred virus are replication deficient recombinant adenovirus
  • Engineered virus may be administered locally or systemically to
  • transgenic animals for studying in vivo the effect of for instance, modifying the binding
  • a transgenic animal is one with cells
  • nucleic acid may be introduced indirectly by viral transfer as indicated
  • nucleic acid sequence of interest may be introduced into an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in an embryonal stem (ES) cell in
  • a transgenic animal homozygous for a transferred gene for instance may be
  • a transgenic animal may for instance be a mouse, rat,
  • the transgenic animal will be a mouse.
  • MAP kinase like ERK2 or JNK-1 with a integrin include antagonists and inhibitors
  • Antagonists and inhibitors include those agents that act by binding the
  • a binding domain may be
  • binding domain for the MAP kinase may then be determined utilising progressively
  • test peptides are readily
  • MAP kinase is achieved with the use of further synthesised test peptides in which one or
  • substitution mutagenesis will involve substitution of selected amino acids
  • deletion is meant deletion of one or more of the amino acids between the N-
  • nucleic acid sequence encoding such peptide fragments as well as the amino acid
  • binding domain typically involve determining the physical properties of the binding domain such as size
  • At least the region of the integrin containing the binding domain is
  • the modelling will take into account the interaction of the binding domain with the MAP
  • the design of a mimetic of the binding domain will usually involve selecting or
  • polypeptide or the like may be incorporated into an amino acid sequence at
  • ⁇ -pleated sheet or helical structure such as an ⁇ -helix
  • a polypeptide or other agent may also be cyclised to provide enhanced rigidity and
  • Cyclisation may also be achieved by the formation of a peptide bond between the N-
  • Variation of cycle size for optimisation of binding affinity may be achieved by
  • peptide library technology provides an
  • kinase as the case may be or prior to the addition of the integrin or the MAP kinase, and
  • alternate screening method may for instance involve selecting a test agent capable of
  • test agent comparing that activity with cellular activity in the absence of the test
  • Cellular activity may be assessed by cell growth as indicated by [ H]-thymidine uptake or other measurement of cellular activity. As will be understood, a difference in
  • integrin for the MAP kinase or a homolog, analog, variant or derivative of such a
  • binding domain of the integrin or core amino acid sequence of the binding domain that
  • Polypeptides including fusion proteins and fragments of an integrin subunit

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Compounds Of Unknown Constitution (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

There is disclosed agents capable of inhibiting the binding of a MAP kinase to a binding domain of an integrin for the MAP kinase, and methods of modulating the activity of a cell utilising the agents. The methods are particularly suitable for inhibiting the growth of cancer cells.

Description

A METHOD OF MODULATING INTEGRIN MEDIATED CELLULAR
ACTIVITY AND AGENTS USEFUL FOR SAME
Field Of The Invention
The present invention relates to a method of modulating cell activity mediated by a
cell adhesion molecule and more particularly, finds application in down regulation of the
growth and/or the proliferation of a cell such as a neoplastic cell. In particular, the
invention finds use inter alia in the prophylaxis or treatment of conditions such as
cancers requiring modulation of cell activity. The present invention also provides agents
for use in the above.
Background Of The Invention
Colorectal cancer is the commonest internal malignancy affecting men and women
in Australia. About 4% of individuals develop this disease during the course of their
lifetime and it was responsible for 14% of cancer deaths in that country in 1990. In 1995
there were 10,615 cases of colorectal cancer and 4508 deaths in Australia.
Worldwide, an estimated 875,000 cases of colorectal cancer occurred in 1996,
accounting for 8.5% of all new cases of cancer. Incidence rates vary approximately 20-
fold around the world, with the highest rates seen in the developed world and lowest in
India. Australian incidence rates are towards the higher end of the scale internationally
alongside those for North America and New Zealand. Five-year survival following
diagnosis of colon cancer is around 55% in the developed world and has altered little
during the past few decades despite advances in chemo-, immuno- and radiotherapy.
Colorectal cancer is a malignant tumour that starts in the bowel wall and is
confined locally for a relatively long period before spreading through the bowel wall and metastasising to lymph nodes and other parts of the body. Survival rates are
significantly improved where the disease is detected and treated early.
The aetiology of colorectal cancer is complex and appears to involve interactions
between inherited susceptibility and environmental factors. Recognition of the genetic
component of colorectal cancer is growing. Mutations are present as inherited germline
defects or arise in somatic cells secondary to environmental insult. There are two main
inherited predisposition syndromes: Familial Adenomatous Polyposis (FAP) and
Hereditary Non-Polyposis Colorectal Cancer (FINPCC); the remaining cases are
attributed to so-called sporadic colorectal cancer. FAP and HNPCC contribute to
approximately 1% and 4%, respectively, of all colorectal cancers and a strong family
history of bowel cancer in first-degree relatives is obtained in another 10 - 15% of
patients.
However, in the vast majority of patients the aetiology of large bowel cancer
remains unknown. Most colon cancer arises within pre-existing benign precursor lesions
or adenomas. Adenomas are classified by histological architecture as tubular,
tubulovillous or villous. Villous change is associated with a higher malignant potential,
as are large and high-grade epithelial dysplasia. Environmental risk factors for
development of colorectal cancer include diets low in fibre and vegetables and high in
fat, red meat and alcohol and cigarette smoking which may induce mutations in somatic
cells.
Studies have shown that persistent genetic instability and accumulation of
mutations in several genes that are mainly concerned with cell growth or DNA repair,
may be critical for the development of all colorectal cancers. For example, a normal mucosal cell with inactivation of tumour suppressor genes such as the Adenomatous
Polyposis Coli (APC) gene or Mutated in Colon Cancer (MCC) gene can proliferate and
become a small adenomatous polyp. Mutations in oncogenes such as k ras and in
tumour suppressor genes such as p53 and the Deleted in Colon Cancer gene (DCC) may
then occur and lead to the transformation of the polyp into a large adenoma, from which
a carcinoma can eventually arise. The uncontrolled cell growth that leads to the
development of neoplasia is believed to result, therefore, from a series of inherited and
acquired accumulated genetic changes. This multistep process confers on cells the
capacity to survive and proliferate in a manner freed from the constraints imposed on
normal cell growth.
Spread of cancer cells involves tumour cell migration through the extracellular
matrix scaffold, invasion of basement membranes, arrest of circulating tumour cells, and
tumour cell extravasation and proliferation at metastatic sites. Detachment of cells from
the primary tumour mass and modification of the peri-cellular environment aid
penetration of tumour cells into blood and lymphatic vessels. It is the invasive and
metastatic potential of tumour cells that ultimately dictates the fate of most patients
suffering from malignant diseases. Hence, tumourigenesis can be viewed as a tissue
remodelling process that reflects the ability of cancer cells to proliferate and digest
surrounding matrix barriers. These events are thought to be regulated, at least in part, by
cell adhesion molecules and matrix-degrading enzymes.
Cell adhesion receptors on the surface of colon cancer cells are involved in complex cell signalling which may regulate cell proliferation, migration, invasion and
metastasis and several families of adhesion molecules have now been identified including integrins, cadherins, the immunoglobulin superfamily, hyaluronate receptors,
and mucins (Agrez, 1996.) In general, these cell surface molecules mediate both cell-
cell and cell-matrix binding, the latter involving attachment of tumour cells to
extracellular scaffolding molecules such as collagen, fibronectin and laminin. It is now
clear that multiple and varied cell adhesion receptors exist on colon cancer cells at any
one time and an understanding of the role of individual receptors in promoting growth
and spread of colon cancer is only just beginning to be elucidated.
Of all the families of cell adhesion molecules, the best-characterised at the present
time is the family known as integrins. Integrins are involved in several fundamental
processes including leucocyte recruitment, immune activation, thrombosis, wound
healing, embryogenesis, virus internalisation and tumourigenesis. Integrins are
transmembrane glycoproteins consisting of an alpha (α) and beta (β) chain in close
association that provide a structural and functional bridge between extracellular matrix
molecules and cytoskeletal components with the cell. The integrin family comprises 17
different α and 8 β subunits and the αβ combinations are subsumed under 3 subfamilies.
Excluding the leucocyte integrin subfamily that is designated by the β2
nomenclature, the remaining integrins are arranged into two major subgroups, designated
βl and αv based on sharing common chains.
In the βl subfamily, the βl chain combines with any one of nine α chain members
(αl-9), and the α chain which associates with βl determines the matrix-binding
specificity of that receptor. For example, α2βl binds collagen and laminin, α3βl binds
collagen, laminin and fibronectin, and α5βl binds fibronectin. In the αv subfamily of
receptors, the abundant and promiscuous αv chain combines with any one of five β chains, and a distinguishing feature of αv integrins is that they all recognise and bind
with high affinity to arginine-glycine-aspartate (RGD) sequences present in the matrix molecules to which they adhere (Hynes, 1992).
The current picture of integrins is that the N-terminal domains of α and β subunits
combine to form a ligand -binding head on each integrin. This head, containing the
cation binding domains, is connected by two stalks representing both subunits, to the
membrane-spanning segments and thus to the two cytoplasmic domains. The β subunits
all show considerable similarity at the amino acid level (Loftus et al, 1994). All have a
molecular mass between 90 and 110 kDa, with the exception of β4 which is larger at 210
kDa.. Similarly, they all contain 56 conserved cysteine residues, except for β4 which
has 48. These cysteines are arranged in four repeating patterns which are thought to be
linked internally by disulphide bonds. The α-subunits have a molecular mass ranging
from 150 - 200 kDa. They exhibit a lower degree of similarity than the β chains,
although all contain seven repeating amino acid sequences interspaced with non-
repeating domains.
The β subunit cytoplasmic domain is required for linking integrins to the
cytoskeleton (Hynes, 1992). In many cases, this linkage is reflected in localisation to
focal contacts, which is believed to lead to the assembly of signalling complexes that
include α-actinin, talin, and focal adhesion kinase (FAK) (Otey et al, 1990; Guan and
Shalloway, 1992; Kornberg et al, 1992). At least three different regions that are
required for focal contact localisation of βl integrins have been delineated (Reszka et al,
1992). These regions contain conserved sequences that are also found in the cytoplasmic
domains of the β2, β3, β5, β6 and β7 integrin subunits. The functional differences between these cytoplasmic domains with regard to their signalling capacity have not yet
been established.
Ligation of integrins by their extracellular matrix protein ligands induces a cascade
of intracellular signals that include tyrosine phosphorylation of focal adhesion kinase,
increases in intracellular Ca levels, inositol lipid synthesis, synthesis of cyclins and
expression of immediate early genes. In contrast, prevention of integrin-ligand
interactions suppresses cellular growth or induces apoptotic cell death (Meredith et al,
1993; Montgomery et al, 1994; Brooks et al, 1994; Varner et al, 1995; Boudreau et al,
1995). Thus, integrins play roles in a number of cellular processes that impact on the
development of tumours, including the regulation of proliferation and apoptosis.
The integrin β6 subunit was first identified in cultured epithelial cells as part of the
αvβ6 heterodimer, and the αvβ6 complex was shown to bind fibronectin in an arginine-
glycine-aspartate (RGD)-dependent manner in human pancreatic carcinoma cells
(Sheppard et al, 1990; Busk et al, 1992). The β6 subunit is composed of 788 amino acids
and shares 34 - 51% sequence homology with other β integrin subunits βl - β5. The β6
subunit also contains 9 potential glycosylation sites on the extracellular domain
(Sheppard et al, 1990). The cytoplasmic domain differs from other β subunits in that it
is composed of a 41 amino acid region that is highly conserved among integrin β
subunits, and a unique 11 amino acid carboxy-terminal extension. The 11 amino acid
extension has been shown not to be necessary for localisation of β6 to focal contacts; in
fact, its removal appears to increase receptor localisation. However, removal of any of
the three conserved regions previously identified as important for the localisation of βl integrins to focal contacts (Reszka et al, 1992) has been shown to eliminate recruitment
of β6 to focal contacts (Cone et al, 1994).
The integrin αvβ6 has previously been shown to enhance growth of colon cancer
cells in vitro and in vivo (Agrez et al, 1994). What makes this epithelial-restricted
integrin of particular interest in colon cancer is that it is not expressed in normal cells but
is highly expressed during tumourigenesis (Breuss et al, 1995; Agrez et al, 1996).
Invasion of the extracellular matrix and metastatic spread of colon cancer is also
likely to reflect the ability of tumour cells to digest their surrounding matrix scaffold
through secretion of matrix-degrading enzymes such as matrix metalloproteinases
(MMPs). The mechanisms whereby human colon cancer cells escape the constraints on
growth imposed on normal cells by cell crowding and dense pericellular matrices is
unclear. However, even colon cancer cells are subject to relative growth inhibition in
vitro in a dense extracellular matrix environment (Agrez, 1989).
Integrins can signal through the cell membrane in either direction. The
extracellular binding activity of integrins can be regulated from the cell interior as, for
example, by phosphorylation of integrin cytolasmic domains (inside-out signalling),
while the binding of the extracellular matrix (ECM) elicits signals that are transmitted
into the cell (outside-in signalling) (Giancotti and Ruoslahti, 1999). Outside-in
signalling can be roughly divided into two descriptive categories. The first is 'direct
signalling' in which ligation and clustering of integrins is the only extracellular stimulus.
Thus, adhesion to ECM proteins can activate cytoplasmic tyrosine kinases (e.g. focal
adhesion kinase FAK) and serine/threonine kinases (such as those in the mitogen-
activated protein kinase (MAPK) cascade) and stimulate lipid metabolism (eg phosphatidylinositol-4, 5-biphosphate (PjP2) synthesis. The second category of integrin
signalling is 'collaborative signalling', in which integrin-mediated cell adhesion
modulates signalling events initiated through other types of receptors, particularly
receptor tyrosine kinases that are activated by polypeptide growth factors (Howe et al,
1998). In all cases, however, integrin-mediated adhesion seems to be required for
efficient transduction of signals into the cytosol or nucleus.
MAP kinases behave as a convergence point for diverse receptor-initiated
signalling events at the plasma membrane. The core unit of MAP kinase pathways is a
three-member protein kinase cascade in which MAP kinases are phosphorylated by MAP
kinase kinases (MEKs) which are in turn phosphorylated by MAP kinase kinase kinases
(e.g. Raf-1) (Garrington and Johnson, 1999). Amongst the 12 member proteins of the
MAP kinase family are the extracellular signal-regulated kinases (ERKs) (Boulton et al,
1991) activated by phosphorylation of tyrosine and threonine residues (Payne et al,
1991) which is the type of activation common to all known MAP kinase isoforms. ERK
1/2 (44kD and 42kD MAPks, respectively) share 90% amino acid identity and are
ubiquitous components of signal transduction pathways (Boulton et al, 1991). These
serine/threonine kinases phosphorylate and modulate the function of many proteins with
regulatory functions including other protein kinases (such as p90rs ) cytoskeletal proteins
(such as microtubule-associated phospholipase A2 ), upstream regulators (such as the
epidermal growth factor receptor and Ras exchange factor) and transcription factors
(such as C-Myc and Elk-1).
MAP kinases can be activated through non-receptor tyrosine kinases such as focal
adhesion kinase (FAK), cytoplasmic tyrosine kinase (ρρ60 c-srk) (Schlaepher and Hunter, 1998), and growth factors acting through membrane-associated receptor tyrosine
kinases. The FAK pathway is activated by most integrins. In addition to activating
FAK, some βl and αv integrins also activate the tyrosine kinase Fyn and through it, the
adaptor protein She (Wang et al, 1996). It is likely that both FAK and She contribute to
the activation of Ras and thence to the downstream kinase cascade of Raf-1 , MEK, and
MAP kinases (Schlaepfer et al, 1994; 1997). It is now generally accepted that the
activation of ERK in response to integrin ligation requires Ras signalling (Wary et al,
1996; Schlaepfer and Hunter, 1997). The laminin receptor α6β4, the laminin/collagen
receptor αlβl, the fibronectin receptor α5βl and the RGD binding receptor αvβ3 are
linked to the Ras-Raf-MEK-ERK signalling pathway and control of immediate early
gene expression (Wary et al, 1996; Maniero et al, 1995; 1997). The ability of integrins to
activate ERK may be especially important when the concentration of growth factors
available to the cell is limited. In this setting, proliferation is likely to require co-
stimulation of ERK through integrins and growth factor receptors (Giancotti and
Ruoslahti, 1999). Moreover, activation of ERK in response to integrin ligation may play
a role in regulating cell migration (Klemke et al, 1997) possibly by initiating matrix-
degrading enzyme secretion.
While there is a good deal of evidence in support of a key role for FAK and the
phosphotyrosine-domain-containing adaptor protein She (Howe et al, 1998; Giancotti &
Ruoslahti, 1999) in the Ras-Raf MEK-MAP kinase activation pathway there are also
data implicating alternate pathways independent of MEKs. For example, MEK-
independent regulation of MAP kinase activation in NIH3T3 fibroblasts has been shown
to be mediated by phosphatidylinositol-3 -kinases and the conventional protein kinase C (PKC) isoforms and is thought to be due to inactivation of a MAP kinase inhibitor
(Grammer and Blenis, 1997).
Although the mechanism by which PKC regulates integrin function is not known,
PKC has been shown to regulate integrin-induced activation of the MAP kinase pathway
upstream of She. For example, PKC inhibition has been shown to inhibit ERK2
activation by fibronectin receptors without any effect on integrin-induced FAK or
paxillin tyrosine phosphorylation (Miranti et al, 1999). Hence, MAP kinase activation is
more complicated than a simple linear pathway, and the mechanistic basis for the
commonly observed integrin-mediated activation of MAP kinases remains controversial.
Various intracellular proteins may be linked directly or spatially to integrin
cytoplasmic domains. Direct interactions have been identified between cytoskeletal
proteins such as α-actinin and talin and βl and β3 integrin tails (Horwitz et al, 1986;
Otey et al, 1990; Knezevic et al, 1996; Pfaff et al, 1998). A direct association between
FAK and the βl integrin tail has been suggested based on in vitro βl peptide studies, but
this remains to be confirmed (Schaller et al, 1995). More recently, the cytoplasmic
domain of the α4 subunit has been found to be physically associated with the signalling
adaptor protein paxillin in Jurkat T cells, and this binding event regulates the kinetics of
FAK tyrosine phosphorylation (Liu et al, 1999).
Direct integrin links with the intracellular calcium-binding protein, calreticulin,
and integrin-linked kinase (ILK) (Hannigan et al, 1996) have been shown to regulate
"inside-out" integrin signalling. For example, calreticulin has been shown to bind to α
chain cytoplasmic domains (Rojiani et al, 1991) and modify α2βl integrin activation by
phorbol esters and anti-integrin antibodies (Coppolino et al, 1995). Newly identified integrin-binding molecules include the serine/threonine integrin-linked kinase, ILK,
which can associate with the β 1 , β2 and β3 subunits. When over-expressed, ILK has
been shown to reduce anchorage-independent growth and tumourigenicity in nude mice
(Hannigan et al, 1996). Co-immunoprecipitation strategies have also demonstrated
interactions between integrins and the integral plasma membrane protein IAP, and
members of the four transmembrane domain protein family (tetraspans). The
extracellular Ig region of the IAP molecule mediates association with αvβ3 and is
required for cell binding to vitronectin-coated particles (Lindberg et al, 1996). An
emerging model for tetraspans is that they recruit signalling enzymes such as
phosphatidylinositol-4-kinase and PKC into complexes with integrins (Hemler, 1998).
Integrins have also been shown to be physically linked with matrix-degrading
enzymes and growth factors. For example, the integrin αvβ6 has been shown to bind and
activate latent TGFβl in keratinocytes (Munger et al, 1999) which is thought to be
important in modulating the inflammatory process following epithelial injury. In
melanoma cells, αvβ3 binds activated gelatinase A (Brooks et al, 1996), and both insulin
and platelet-derived growth factor (PDGF) co-immunoprecipitate with this integrin in
NIH3T3 mouse fibroblasts (Schneller et al, 1997). Synergism between integrin-
mediated signalling processes and growth factor responses is now well-recognised and
Schneller et al (1997) showed that a small subset of each of the insulin receptor and
PDGF β-receptor is tyrosine phosphorylated upon growth factor stimulation.
Interestingly, this subset can associate with the αvβ3 integrin, and PDGF activity is
enhanced in association with increased MAP kinase activity in cells plated on the αvβ3
ligand, vitronectin. Summary Of The Invention
Broadly stated, the present invention relates to modulation of integrin expression
in neoplastic cells to inhibit the growth of the cells, and the surprising finding that
members of the mitogen activated protein (MAP) kinase family associate with the
cytoplasmic domain of an integrin molecule. It is believed that no member of the MAP
kinase family has previously been found to directly associate with any integrin or for that
matter, with any transmembrane protein. The identification of this functional
relationship permits the rational design of agents for therapeutically or prophylactically
modulating cellular activity mediated by the MAP kinase and integrin interaction.
In an aspect of the present invention there is provided an agent capable of
inhibiting binding of a MAP kinase with an integrin.
Typically, the agent will be capable of binding with a binding site on the MAP
kinase that binds to a binding domain on the integrin for the MAP kinase. Alternatively,
the agent may be capable of binding to the binding domain on the integrin for the MAP
kinase or other site on the integrin such that inhibition of binding of the MAP kinase to
the integrin is thereby caused.
The agent may be provided either isolated or for instance, coupled to another
molecule for facilitating transport of the agent into a cell.
In another aspect of the present invention there is provided an isolated polypeptide
capable of binding with a binding site on a MAP kinase which binding site binds with a
binding domain on an integrin for the MAP kinase, or a homolog, analog, variant or
derivative of the polypeptide, with the proviso that the polypeptide is other than a full
length integrin subunit or a β6(770t) or β6(777t) deletion mutant. Preferably, the polypeptide will comprise the binding domain of the integrin or
sufficient core amino acid sequence of the binding domain to enable binding of the
polypeptide with the MAP kinase.
Preferably, the polypeptide will comprise amino acid sequence
RSKAKWQTGTNPLYR, more preferably RSKAKNPLYR, or one or both amino acid
sequences RSKAK and NPLYR. Most preferably, the polypeptide will be a fragment of
an integrin subunit
Accordingly, in a further aspect of the present invention there is provided a
fragment of an integrin subunit wherein the fragment is capable of binding with a MAP
kinase, or a homolog, analog, variant or derivative of the fragment, with the proviso that
the integrin subunit is other than a β6(770t) or β6(777t) deletion mutant.
Preferably, the polypeptide or fragment will have a length of about 150 amino
acids or less, more preferably about 100 or 50 amino acids or less and more usually
about 40 amino acids or less. Typically, the length will be between about 5 to about 30
amino acids.
The fragment may comprise an amino acid sequence incorporating extracellular
and cytoplasmic regions of the integrin subunit. Preferably, the fragment will be a
fragment of the cytoplasmic domain of the integrin subunit.
In another aspect of the present invention there is provided an integrin subunit with
a mutagenised binding domain for a MAP kinase or in which the binding domain is
deleted, wherein capability of the integrin subunit to bind with the MAP kinase is thereby
reduced, or a homolog, analog, variant or derivative of the integrin subunit, with the
proviso that the integrin subunit is other than a β6 Δ746-764 deletion mutant. In still another aspect of the present invention there is provided a fusion protein
incorporating a polypeptide of the invention or a homolog, analog or variant of the
polypeptide.
In a further aspect of the present invention there is provided a fusion protein
incorporating a fragment of the invention or a homolog, analog or variant of the
fragment.
Typically, the polypeptide or fragment will be coupled to a carrier polypeptide for
facilitating entry of the fusion protein into a cell. Preferably, the carrier polypeptide will
be penetatin.
In another aspect of the present invention there is provided an isolated nucleic acid
sequence encoding a polypeptide of the invention or a homolog, analog, or variant of the
polypeptide.
In yet another aspect of the invention there is provided an isolated nucleic acid
sequence encoding a fragment of the invention or a homolog, analog, or variant of the
fragment.
In another aspect of the invention there is provided a nucleic acid sequence
encoding an integrin subunit with a mutagenised binding domain for a MAP kinase or in
which the binding domain is deleted, wherein capability of the integrin subunit to bind
with the MAP kinase is thereby reduced, or a homolog, analog, or variant of the integrin
subunit, with the proviso that the integrin subunit is other than a β6 Δ746-764 deletion
mutant.
In a still further aspect of the present invention there is provided an isolated
nucleic acid sequence encoding a fusion protein of the invention. There are also provided antisense nucleic acid sequences complimentary to the
sense nucleic sequences of the invention. Such antisense sequences find application in
antisense therapy of cells in which down regulation of cellular activity is desired, and
include oligonucleotides. Sense oligonucleotides coding for the binding domain of an
integrin subunit or that of a homolog, analog or variant thereof, and complimentary
antisense oligonucleotides find particular application as primers or probes. A nucleic
acid primer or probe of the invention may be labelled with a suitable reporter molecule
for enabling detection of hybridisation of the primer or probe to a target nucleic acid
sequence.
In yet another aspect of the present invention there is provided a vector
incorporating a nucleic acid sequence of the invention. Typically, the vector will be an
expression vector and the nucleic acid sequence will be capable of being transcribed.
In a further aspect of the present invention there is provided a host cell transformed
with a vector of the invention.
Preferably, the host cell will be selected from the group consisting of a mammalian
cell, an epithelial cell, a neoplastic cell, and a cancer cell. Preferably, the host cell will
be a mammalian cell and most preferably, a colon cancer cell
In a further aspect of the present invention there is provided a transgenic animal
with cells containing heterologous nucleic acid of the invention.
In yet another aspect of the present invention there is provided an antibody
generated with the use of a polypeptide or fragment of the invention.
In a still further aspect of the invention there is provided an antibody capable of
binding to a binding domain on an integrin for a MAP kinase. The antibody may be a polyclonal or monoclonal antibody. Preferably, the
antibody is a monoclonal antibody.
In still another aspect of the present invention there is provided a method of
isolating a MAP kinase from a sample utilising a molecule immobilised on a solid
support and which is capable of binding to a binding site on the MAP kinase which
binding site binds with a binding domain of an integrin for the MAP kinase, comprising:
(a) contacting the molecule immobilised on the solid support with the sample
under conditions suitable for binding of the MAP kinase to the molecule;
(b) eluting the MAP kinase from the solid support;
(c) collecting the eluted MAP kinase
The molecule may be the integrin, or a fusion protein, a polypeptide, or a fragment
of the invention to which the MAP kinase is capable of binding, or for instance an
analog, homolog, variant or derivative of the polypeptide or fragment.
In yet another aspect of the present invention there is provided a MAP kinase
isolated by a method of the invention.
Rather than isolating the MAP kinase from a sample, the MAP kinase may be
immobilised on a solid support and used to isolate the molecule from a sample, and all
such methods are also expressly encompassed as is the molecule when isolated in this
way.
In another aspect of the present invention there is provided a method of screening
for an agent capable of inhibiting binding of a MAP kinase to a binding domain of an
integrin for the MAP kinase, comprising: (a) testing a number of agents for ability to bind to either the MAP kinase or the integrin; and
(b) determining if any said agent is capable of inhibiting binding of the MAP
kinase to the binding domain of the integrin on the basis of the testing.
In another aspect of the invention there is provided a method of screening for an
agent capable of inhibiting binding of a MAP kinase to a binding domain on an integrin
for the MAP kinase, comprising:
(a) testing a number of agents for ability to bind to either the MAP kinase or the
integrin;
(b) selecting an agent or agents identified as being able to bind to the MAP
kinase or the integrin on the basis of the testing; and
(c) utilising the selected said agent or agents in an assay for indicating whether
the or any of the selected said agents is capable of inhibiting the binding of the MAP
kinase to the binding domain of the integrin.
In another aspect of the invention there is provided a method of evaluating whether
an agent is capable of inhibiting binding of a MAP kinase to a binding domain of an
integrin for the MAP kinase, comprising:
(a) selecting the agent;
(b) utilising the agent in an assay for indicating whether the agent is capable of
inhibiting the binding of the MAP kinase to the binding domain of the integrin; and
(c) determining if the agent is capable of inhibiting the binding of the MAP
kinase to the binding domain of the integrin on the basis of the assay. Testing or assaying of an agent for ability to bind to the MAP kinase or the
integrin and thereby inhibit binding of the MAP kinase to the integrin, may comprise
exposing the integrin to the agent(s) to enable binding of the agent(s) to the integrin to
occur either in the presence of the MAP kinase or prior to the addition of the MAP
kinase. Rather than using the integrin, a polypeptide or fragment of the invention or
other molecule capable of binding with the binding site on the MAP kinase that binds to
the integrin may be used. Alternatively, the testing or assaying may comprise exposing
the MAP kinase to the agent(s) to enable binding of the agent(s) to the MAP kinase to
occur either in the presence of the integrin or other molecule capable of binding with the
binding site on the MAP kinase, or prior to the addition of the integrin or molecule.
In still another aspect of the present invention there is provided a method of
screening for an agent capable of binding to a binding domain of an integrin for a MAP
kinase, comprising:
(a) testing a number of agents for ability to bind to the binding domain of the
integrin for the MAP kinase; and
(b) determining if any said agent is capable of binding to the binding domain of
the integrin on the basis of the testing.
In yet another aspect of the invention there is provided a method of screening for
an agent capable of binding to a binding domain of an integrin for a MAP kinase,
comprising:
(a) testing a number of agents for ability to bind to the integrin;
(b) selecting an agent or agents identified as being able to bind to the integrin on
the basis of the testing; and (c) utilising the selected said agent or agents in an assay for indicating whether
the or any of the selected said agents is capable of binding to the binding domain on the
integrin for the MAP kinase.
In another aspect of the present invention there is provided a a method of
evaluating whether an agent is capable of binding to a binding domain of an integrin for
a MAP kinase, comprising:
(a) testing the agent for ability to bind to the binding domain of the integrin for the
MAP kinase; and
(b) determining if the agent is capable of binding to the binding domain on the
basis of the testing.
Preferably, a polypeptide or fragment of the invention consisting of the binding
domain of the integrin or core amino acid sequence of the binding domain or a homolog,
analog or variant of the polypeptide or fragment is used in the testing or assaying for
whether an agent is capable of binding to the binding domain of the integrin. Most
preferably, the polypeptide or fragment will consist of the amino acid sequence
RSKAKWQTGTNPLYR or RSKAKNPLYR.
An agent of the invention will usually be provided in the form of a pharmaceutical
composition. Accordingly, in another aspect of the present invention there is provided a
pharmaceutical composition comprising an agent of the invention capable of inhibiting
binding of a MAP kinase to a binding domain on an integrin for the MAP kinase, and a
pharmaceutically acceptable carrier or diluent. The agent may or not be proteinaceous in nature. Preferably, the agent will
comprise a fusion protein, or polypeptide. Most preferably, the agent will be coupled to
a carrier molecule for facilitating entry of the agent into a cell.
In a further aspect of the invention there is provided a pharmaceutical composition
comprising a nucleic acid sequence of the invention and a pharmaceutically acceptable
carrier or diluent. Preferably, the nucleic acid sequence is incorporated into a vector as
described herein. Alternatively, the nucleic acid sequence may be joined to a carrier
molecule for facilitating entry of the nucleic acid sequence into a target cell.
In a further aspect of the present invention there is provided a method of
modulating activity of a cell, comprising:
transfecting the cell with a nucleic acid sequence encoding an integrin
subunit for being expressed by the cell, wherein the integrin subunit has a mutagenised binding domain for a MAP kinase or in which the binding domain is deleted, or a
homolog, analog or variant of the integrin subunit.
In yet another aspect of the present invention there is provided a method of
modulating activity of a cell, comprising:
transfecting the cell with a nucleic acid encoding a polypeptide for being
expressed by the cell wherein the polypeptide is capable of inhibiting binding of a MAP
kinase with a binding domain on an integrin for the MAP kinase, or a homolog, analog
or variant of the polypeptide.
Preferably, the polypeptide will be capable of binding with the binding site on the
MAP kinase which binding site binds to the binding domain on the integrin. In still another aspect of the invention there is provided a method of modulating
activity of a cell, comprising causing the expression of an integrin to which a MAP
kinase is able to bind to be down-regulated.
Preferably, down-regulation of the expression of the integrin is achieved using an
antisense nucleic acid sequence that inhibits expression of the gene encoding the
integrin. The antisense nucleic acid sequence may be administered to the cell or
generated in vivo within the cell. Preferably, the cell will be transformed with a vector
of the invention for generation of the antisense nucleic acid sequence in vivo.
Preferably, the antisense nucleic acid sequence will specifically hybridise with the
sense nucleic acid sequence coding for the binding domain of the integrin for the MAP
kinase and/or intron sequence between such coding sequence.
In a still further aspect of the invention there is provided a method of modulating
activity of a cell, comprising contacting the cell with an effective amount of an agent for
down regulating functional activity of an integrin expressed by the cell and thereby the
activity of the cell.
In another aspect of the present invention there is provided a method of modulating
activity of a cell, comprising:
contacting the cell with an effective amount of an agent capable of inhibiting
binding of a MAP kinase to a binding domain on an integrin expressed by the cell.
In a further aspect of the invention there is provided a method of modulating
activity of a cell, comprising: contacting the cell with an effective amount of an agent capable of binding to
a MAP kinase to thereby inhibit binding of the MAP kinase to a binding domain on the
integrin for the MAP kinase.
Preferably, the agent will be capable of binding to the binding site on the MAP
kinase which binding site binds to the binding domain on the integrin for the MAP
kinase.
In a further aspect of the present invention there is provided a method of
prophylaxis or treatment of a disease or condition in a mammal wherein modulation of
cell activity is desirable, comprising:
administering to the mammal an effective amount of a nucleic acid sequence
encoding an integrin subunit for being expressed, wherein the integrin subunit has a
mutagenised binding domain for a MAP kinase or in which the binding domain is
deleted, or a homolog, analog or variant of the integrin subunit.
In another aspect of the invention there is provided a method of prophylaxis or
treatment of a disease or condition in a mammal wherein modulation of cell activity is
desirable, comprising:
administering to the mammal an effective amount of a nucleic acid of the invention
capable of causing the expression of an integrin to which a MAP kinase is able to bind to
be down regulated.
In still another aspect of the present invention there is provided a method of
prophylaxis or treatment of a disease or condition in a mammal wherein modulation of
cell activity is desirable, comprising: administering to the mammal an effective amount of a nucleic acid sequence encoding a polypeptide for being expressed, wherein the polypeptide is capable of
inhibiting binding of a MAP kinase with a binding domain of an integrin for the MAP
kinase to thereby cause down-regulation of MAP kinase integrin binding, or a homolog,
analog or variant of the polypeptide.
In another aspect of there is provided a method of treatment or prophylaxis of a
disease or condition in a mammal, wherein said condition is responsive to an agent of the
invention capable of inhibiting binding of a MAP kinase to binding domain on an
integrin for the MAP kinase and the method comprises administering an effective
amount of the agent to the mammal.
In another aspect there is provided use of a nucleic acid of the invention in the
manufacture of a medicament for administration to a mammal in the prophylaxis or
treatment of a disease or condition in which down regulation of cellular activity is
desirable.
In a still further aspect of the invention there is provided the use of an agent
capable of inhibiting binding of a MAP kinase to a binding domain of an integrin in the
manufacture of a medicament for administration to a mammal for the prophylaxis or
treatment of a disease or condition where down regulation of cellular activity is
desirable.
The cellular activity desired to be down-regulated will typically but not
exclusively, be cell growth. Indeed, any activity influenced by signalling mediated by
MAP kinase activation is expressly included within the scope of the invention. The cell may be any cell type in which functional activity of a MAP kinase arising
from binding with an integrin may occur. Preferably, the cell will be an epithelial cell
and most preferably, a neoplastic cell. By modulating the activity a neoplastic cell,
methods of the invention find particular application in the prophylaxis or treatment of
cancer. In particular, methods of the invention find particular application in the
treatment of colon cancer.
Usually, the MAP kinase will be selected from the group consisting of an
extracellular signal-regulated kinase (ERK) and a JNK MAP kinase. Preferably, the
MAP kinase is ERK2 or JNK-1. Most preferably, the MAP kinase is ERK2.
The mammal may be any mammal treatable with a method of the invention. For
instance, the mammal may be a member of the bovine, porcine, ovine or equine families,
a laboratory test animal such as a mouse, rabbit, guinea pig, a cat or dog, a primate or a
human being. Preferably, the mammal will be a human being.
The term "modulating" is to be taken as reference to down-regulating the activity
of the cell or the functional activity of the integrin. Reference to "down-regulating"
should be understood to include preventing, reducing or otherwise inhibiting one or
more aspects of the activity of the cell or the functional activity of the integrin molecule
or the MAP kinase
In the broadest sense, the term "integrin" unless otherwise specified, is to be taken
to encompass an integrin family member or a homolog, derivative, variant or analog of
an integrin subunit, or an integrin family member incorporating at least one such
homolog, derivative, variant or analog of an integrin subunit. Usually, the integrin will
be a member of the αv subfamily. Preferably, the integrin is or incorporates an integrin subunit selected from the group consisting of β3, β5 and β6. Most preferably, the
integrin comprises β6.
By "binding domain" is meant the minimum length of contiguous amino acid
sequence required for binding by the MAP kinase. By "core amino acid sequence" is
meant regions or amino acids of the binding domain that directly participate in the
binding as distinct from any amino acids that do not directly participate in the binding
interaction with the MAP kinase. Typically, the core amino acid sequence of the binding
domain will comprise regions of the binding domain linked together by a number of
intervening amino acids which do not directly participate in the binding.
The term "homolog" is to be taken to mean a molecule that has amino acid
sequence similarity. The homology between amino acid sequences can be determined by
comparing amino acids at each position in the sequences when optimally aligned for the
purpose of comparison. The sequences are considered homologous at a position if the
amino acids at that position are the same. Typically, a homolog will have an overall
amino acid sequence homology of at least about 30% more preferably at least about 50%
or 70% and most preferably, greater than about 80%, 90% or 98% sequence homology.
Homology with the binding domain of an integrin may be greater than the overall amino
acid sequence homology of the homolog, and will usually be greater than about 60% or
80%, and more usually greater than about 90%, 95% or 98%.
The term "analog" is to be taken to mean a molecule that has one or more aspects
of biological function characteristic of the molecule on which at least part of the analog
is based or which was otherwise utilised in the design or preparation of the analog. An
analog may have substantial overall structural similarity with the molecule or only structural similarity with one or more regions or domains thereof responsible for the
desired characteristic biological function. By "structural" similarity is meant similarity
in shape, conformation and/or other structural features responsible for the provision of
the biological function or which otherwise have involvement in the provision of the
biological function. Alternatively, it will be understood that with knowledge of the
region(s) or domain(s) of a molecule that provide(s) the characteristic biological
function, analogs may be designed that while differing in structure nevertheless possess
such biological function. Indeed, it is not necessary that an analog have amino acid
sequence homology, and an analog may not be proteinaceous at all. An analog may for
instance be a mimetic of a molecule.
By the term "variant" is meant an isoform of an integrin subunit, an integrin
subunit encoded by an allelic variant of a gene for an integrin subunit, a naturally
occurring mutant form of a gene for an integrin subunit, or an integrin subunit or
polypeptide having an amino acid sequence that differs in one or more amino acids but
which retains one or more aspects of desired characteristic biological function. This may
be achieved by the addition of one or more amino acids to an amino acid sequence,
deletion of one or more amino acids from an amino acid sequence and/or the substitution
of one or more amino acids with another amino acid or amino acids. Inversion of amino
acids and any other mutational change that results in alteration of an amino acid
sequence are also encompassed. A variant may be prepared by introducing nucleotide
changes in a nucleic acid sequence that encodes for an integrin subunit or amino acid
sequence such that the desired amino acid changes are achieved upon expression of the
mutagenised nucleic acid sequence, or for instance by synthesising an integrin subunit or amino acid sequence incorporating the desired amino acid changes, both of which
possibilities are well within the capability of the skilled addressee.
Substitution of an amino acid may involve a conservative or non-conservative
amino acid substitution. By conservative amino acid substitution is meant replacing an
amino acid residue with another amino acid having similar stereochemical properties
(eg. structure, charge, acidity or basicity characteristics) and which does not substantially
effect conformation or the desired aspect or aspects of characteristic biological function.
Preferred variants include ones having amino acid sequences in which one or more
amino acids have been substituted with alanine or other neutrally charged amino acid
residue(s), or to which one or more such residues have been added. A variant may also
incorporate an amino acid or amino acids that are not encoded by the genetic code.
By the term "derivative" is meant a molecule that is derived or obtained from
another molecule and which retains one or more aspects of characteristic biological
function of that molecule. A derivative may for instance arise as a result of the cleavage
of the parent molecule, cyclisation and/or coupling with one or more additional moieties
that improve solubility, lipophilic characteristics to enhance uptake by cells, stability or
biological half-life, decreased cellular toxicity, or for instance to act as a label for
subsequent detection or the like. A derivative may also result from post-translational or
post-synthesis modification such as the attachment of carbohydrate moieties or chemical
reaction(s) resulting in structural modification(s) such as the alkylation or acetylation of
amino acid residues or other changes involving the formation of chemical bonds. The term "polypeptide" is used interchangeably herein with "peptide" and
encompasses amino acid sequences incorporating only a few amino acid residues or
many amino acid residues coupled by peptide bonds.
The term "neoplastic cell" is to be taken to mean a cell exhibiting abnormal growth
and may or may not be a malignant cell. "Growth" is to be taken in its broadest sense
and includes proliferation of the cell. In this regard, an example of abnormal cell growth
is the uncontrolled proliferation of a cell.
Unless the context clearly requires otherwise, throughout the description and the
claims, the words 'comprise', 'comprising', and the like are to be construed in an
inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense
of "including, but not limited to".
The features and advantages of the present invention will become further apparent
from the following detailed description of preferred embodiments and the accompanying
drawings.
Brief Description Of The Accompanying Drawings
Figure 1 : Surface biotinylation and immunoprecipitation of integrin subunits β5
and β6 in HT29 colon cancer cells stably transfected with either vector alone (mock
transfectants) or antisense β6 construct;
Figure 2: Amplification of β6 and glyceraldehyde dehydrogenase (GAPDH)
mRNA: Ethidium-stained agarose gels with amplification products following RT-PCR
from total RNA extracted from transfected HT29 and WiDr cell lines. Equal amounts of
PCR product obtained from RT-PCR reactions were loaded on each lane and the β6 (141 basepairs) and GAPDH (216 basepairs) bands are indicated (WT, wild-type; S, sense β6;
A/S, antisense β6; mock, vector alone);
Figure 3: Non-transfected HT29 cells (wild) and cells transfected with vector alone
(mock), sense β6 and antisense β6 analysed by FACScan for expression of the β6
subunit. White and black histograms represent cells stained in the absence and presence,
respectively, of mAb E7P6 (anti-β6);
Figure 4: WiDr cells transfected with vector alone (mock) or antisense β6 and
analysed by FACScan for expression of the β6 subunit. White and black histograms
represent cells stained in the absence and presence, respectively, of mAb E7P6 (anti-
β6).;
Figure 5 : Tumor cell proliferation in vitro assessed by ( H)-thymidine uptake for
WiDr wild-type cells and transfectants (mock and antisense β6) cultured on plastic for
the times indicated.
Figure 6: Tumor cell proliferation in vitro assessed by ( H)-thymidine uptake for
HT29 wild-type cells and transfectants (mock, sense β6 and antisense β6) cultured on
plastic for the times indicated.
Figure 7: Tumour growth after 6 weeks following subcutaneous inoculation of 10
viable cells of HT29 mock (vector alone) and antisense β6 transfectants;
Figure 8: Graph showing average tumour size at weekly intervals for each
subgroup of 10 animals following subcutaneous inoculation of clone 1 from WiDr mock
(vector alone) and antisense β6 transfectants; Figure 9: Graph showing average tumour size at weekly intervals for each
subgroup of 10 animals following subcutaneous inoculation of HT29 mock (vector
alone) and antisense β6 transfectants;
Figure 10: β6-associated ERK identified by immunoprecipitafions of integrin
subunits from SW480 β6 transfectants. (Top) Cell lysates were immunoprecipitated
with mAbs QE2E5 (anti-βl), L230 (anti-αv), P1F6 (anti-β5), R6G9 (anti-β6), or
matched isotype control Abs (IgGl and IgG2A), and (bottom) blotted with anti-ERK 1/2
mAb, SC-1647, which recognises total ERK (phosphorylated and non-phosphorylated).
Purified, non-phosphorylated ERK2 is shown in the left hand lane;
Figures 11(A) and 1 1(B): (A) Western blotting: equal protein loads of cell lysates
from one representative clone each from WiDr mock and antisense β6 blotted with anti-
ERK mAb (SC-1647) against total ERK. Purified non-phosphorylated ERK2 is shown in
the left hand lane. (B) β6 immunoprecipitates (mAb R6G9) from equal protein loads of
the cell lysates in (A) probed with anti-ERK mAb (ElO). Purified phosphorylated ERK2
is shown in the left hand lane;
Figure 12: β6-bound ERK shown for the high and low SW480 β6-expressing
clones by probing β6 immunoprecipitates with anti-ERK mAb (ElO) against
phosphorylated forms of ERK1/2. Purified, phosphorylated ERK2 is shown in the left
hand lane;
Figures 13(A) and 13(B): (A) Surface biotinylation of WiDr wild-type cells and β6
immunodepletion of the cell lysates by three successive rounds of β6 and β5
immunoprecipitations using mAb R6G9 and P1F6, respectively or control mAb
(IgG2A). The β6 and partner αv bands are arrowed. (B) β6-immunodepleted WiDr cell lysates after 3 successive rounds of β6-immunoprecipitations probed with anti-ERK
mAb SC-1647 recognising both phosphorylated and non-phosphorylated forms of
ERK 1/2 and compared with non-β6 immunodepleted lysates and control lysates sequentially immunoprecipitated 3 times with either isotype matched control mAb
(IgG2 A) or mAb P 1 F6 (anti-β5);
Figure 14: Non-transformed (HaCaT) and Ras-transformed (HaRas) human
keratinocytes: β6 immunoprecipitation and ERK western blots probed with monoclonal
antibody ElO (against Phosphorylated ERK 1/2) and monoclonal antibody SC1647
(against total ERK 1/2), respectively.
Figure 15(A) and 15(B): (A) Depletion of β6 with sequential rounds of ERK
immunodepletion of cell lysates using anti-ERK mAb (SC-1647). (B) MAP kinase
activity in cell lysates before and after 5 rounds of sequential β6 immunodepletion from
SW480 β6 and SW480 mock transfectants (full grey and hatched bars, respectively).
MAP kinase activity is shown as the mean of three independent experiments. The
reduction in MAP kinase activity following β6 immunodepletion was highly signigicant
(P<0.005, students T test).
Figure 16: Shows the amino acid sequence of the cytoplasmic domain of the β6
subunit as well as the amino acid sequences for the βl to β3 subunits, respectively;
Figure 17: Graph showing binding of non-phosphorylated ERK2 (GST.ERK2) to
overlapping fragments corresponding to different regions of the cytoplasmic domain of
the β6 subunit; Figure 18: Graph showing binding of ERK2 (GST.ERK2) to the β6 cytoplasmic
domain and the peptide fragments indicated in Fig. 16 over a range of concentrations of
ERK2;
Figure 19: Graph showing binding of ERK2 (GST.ERK2) to the β6 cytoplasmic
domain and fragments thereof;
Figure 20: Graph showing binding of ERK2 (GST.ERK2) to a 15 mer fragment of
the β6 cytoplasmic domain and which has the amino acid sequence
RSKAKWQTGTNPLYR; and
Figure 21 : Shows regions of the cytoplasmic domain of the β6 subunit
corresponding to synthesised fragments thereof evaluated for capacity to be bound by
ERK2.
Figure 22: Graph showing assay results for ERK2 (GST.ERK) binding to
synthesised 10 mer fragments identified in fig. 21.
Figure 23: Graph showing binding of ERK2 (thrombin cleaved) to synthesised
peptide having the amino acid sequence RSKAKNPLYR compared to the 15 mer
RSKAKWQTGTNPLYR fragment of the cytoplasmic domain of the β6 subunit.
Figure 24: Graph showing binding of JNK-1 to the β6 cytoplasmic domain.
Figure 25: location of β6 Δ746-764, β6(770t) and β6(777t) deletions in the
cytoplasmic domain of the β6 subunit.
Figure 26: SW480 cells transfected with wild-type full length coding sequence for
β6 or β6 Δ746-764 deletion mutant analysed by FACScan for expression of wild-type or
mutant β6. White and black histograms represent cells stained in the absence and
presence of the integrin subunit, respectively. Figures 27(A) and 27(B): (A) Western blotting: equal protein loads of cell lyates
from SW480 cells expressing wild-type β6 or β6 Δ746-764 deletion mutant. (B) β6
immunoprecipitates (mAb R6G9) from equal protein loads of cell lysates (A) probed with anti-ERK mAb (ElO).
Figure 28: Proliferation of HT29 colon cancer cells cultured for 48 hours and
treated with penetratin, the fragment of β6 cytoplasmic domain having amino acid
sequence RSKAKWQTGTNPLYR alone or the fragment coupled to penetratin for the
final 24 hours of the incubation period.
Figure 29: Proliferation of SW480 cells expressing wild-type β6 cultured on
plastic for 48 hours and treated with penetratin, the RSKAKWQTGTNPLYR peptide
alone or the peptide coupled to penetratin for the final 24 hours of the incubation period.
Figures 30 (A) to 29 (C): (A) SW480 cells cultured with control additive for 24
hours; (B) SW480 cells cultured with penetratin for 24 hours; (C) SW480 cells cultured
with RSKAKWQTGTNPLYR bound to penetratin for 24 hours.
Figures 31 (A) and 31 (B): (A) S W480 mock (-β6) and S W480 β6 transfectants
(+β6) cultured in presence of the RSKAKWQTGTNPLYR peptide coupled to
penetratin. (B) Photomicrographs of cells shown in (A) cultured in the presence/absence
of the peptide penetratin complex.
Figures 32(A) to 32(C): (A) SW480 cells cultured in a 3-dimensional collagen type
I matrix photographed in the gel at the end of 10 days. (B) The SW480 cells following
dissolution of the collagen with collagenase. (C) Graph showing numbers of colonies of
the SW480 cells having a diameter exceeding 200μ. Figures 33(A) and 33(B): Graphs showing inhibition of proliferation of SW480
cells expressing full length wild-type β6 in the presence of RSKAKWQTGTNPLYR
peptide bound to penetratin.
Figure 34: Graph showing binding of ERK2 to RSKAKWQTGTNPLYR peptide
and peptides corresponding to regions of the cytoplasmic domain of β3 and β5.
Detailed Description Of Preferred Embodiments Of The Present Invention
The distribution of β6 integrin subunit within various tissues has been assessed by
both in situ hybridisation and immunostaining and reported in the art. For instance, β6
mRNA in adult primate tissues was detected only in epithelial cells and at very low or
undetectable levels in most normal tissues (Breuss et al, 1993). High-level expression of
β6 has been observed in secretory endometrial glands while low-level expression was
detected in the ductal epithelia of salivary gland, mammary gland and epididymis, in gall
and urinary bladder, and in the digestive tract. Immunostaining data has also shown that
β6 expression is restricted to epithelia and is up-regulated in parallel with morphogenetic
events, tumourigenesis and epithelial repair (Breuss et al, 1993; 1995). During
development of the kidney, lung and skin, β6 is expressed by specific types of epithelial
cells, whereas it is mostly undetectable in normal adult kidney, lung and skin. In
contrast, high level expression of β6 has been observed in several types of carcinoma.
For example, β6 is almost invariably neo-expressed in squamous cell carcinomas derived
from the oral mucosa, and often focally localised at the infiltrating edges of tumour cell
islands (Breuss et al, 1995; Thomas et al, 1997). Moreover, expression of the β6 subunit
has been observed in renal cell carcinoma and testicular tumour cell lines (Takiuchi et al, 1994) and 50% of lung cancers have been shown to express this subunit (Smythe et al,
1995).
Expression of β6 is also up-regulated in migrating keratinocytes at the wound edge
during experimental epidermal wound healing. αvβ6 is not expressed in normal
epithelium (Jones et al, 1997). However, following experimental wounding, αv appears
to switch its heterodimeric association from β5 to β6 subunit during re-epithelialisation.
At day 3 after wounding, β6 is absent but then appears around the perimeter of the basal
cells of the migrating epidermis (Clark et al, 1996). By day 14 after wounding, when re-
epithelialisation is complete, all suprabasalar cells overlying the wound express β6 but
not β5. In human mucosal wounds, maximal expression of β6 has been observed
relatively late when epithelial sheets are fused and granulation tissue is present
(Haapasalmi et al, 1996). Furthermore, those investigators observed maximal expression
of tenascin with αvβ6 expression. Interestingly, freshly isolated keratinocytes have not
been found to express β6 but begin to express this after subculturing (Haapasalmi et al,
1996). Moreover, TGF-βl has been shown to induce the de novo expression of αvβ6 at
the cell surface on keratinocytes (Zambruno et al, 1995). This is particularly relevant in
view of the recent observation that αvβ6 binds and activates latent TGF-βl which may
be a means of locally regulating TGF-βl function in vivo during tissue response to injury
(Munger et al, 1999).
αvβ6 expression is also upregulated in type II alveolar epithelial cells during lung
injury caused by injection of live bacteria and αvβ6 mRNA is induced within 5 hours of
acute injury (Breuss et al, 1995). Interestingly, αvβό has been shown to be expressed on
proximal airway epithelial cells in 50% of smokers undergoing lung resection (Weinacker et al, 1995). Just as in human keratinocytes, in primary cultures of human
airway epithelial cells, TGF-βl has been shown to dramatically increase expression of
αvβό without affecting surface expression of any other integrin. Moreover, epidermal
growth factor (EGF) also induces αvβό in these cells, and the effect of both growth
factors on β6 expression has been shown to be additive (Wang et al, 1996). αvβό
expression has also been observed in adult lungs and kidneys at focal sites of sub-clinical
inflammation as well as in a variety of clinical specimens from patients with chronic or
acute inflammation of the lungs or kidneys (Breuss et al, 1995). Taken together these
data indicate that αvβό affects cell spreading, migration and growth during re-
organisation of epithelia in development, tissue repair and neoplasia.
Recent studies have shown that αvβό is a major fibronectin-binding receptor in
colorectal cancer (Agrez et al, 1996). Moreover, normal colonic epithelium from cancer
patients does not express αvβό in immunostaining studies, and as with squamous cell
carcinomas from the oral mucosa (Thomas et al, 1997) maximal β6 expression in colon
cancer has been observed at the invading edges of tumour cell islands (Agrez et al,
1996). Importantly, heterologous expression of the βό subunit in colon cancer cells
lacking constitutive expression of this receptor has been shown to stimulate tumour cell
proliferation in vitro and tumour growth in athymic immune-deficient mice, and this
growth-promoting effect is mediated through the 1 1 amino acid C-terminal cytoplasmic
extension unique to the β6 subunit (Agrez et al, 1994). These findings suggest that one
mechanism of the enhanced growth effect involves induced secretion of matrix-
degrading enzymes as has been previously suggested for invasive melanoma cells.
Support for this arises from findings that the proliferative capacity of colon cancer cells within 3-dimensional collagen matrices is inversely related to the density of the
extracellular matrix (Agrez, 1989), and the observation that induced expression of αVβό
in colon cancer cells markedly enhances gelatinase B secretion (Agrez et al, 1999).
The βό subunit is widely observed in cancers of various origins (Breuss et al,
1995). As described above, β6 is detected in at least 50% of bowel cancer tumours.
Others have reported its presence in oropharyngeal cancers where it is also present and
strongly expressed in the invading margins of the cancer cell islands as is commonly
found in bowel cancer. In the oropharyngeal mucosa, no β6 is observed in the normal
lining cells of the mouth but in both primary and metastatic tumours from the
oropharyngeal mucosa, strong β6 expression is seen which does not correlate with
degree of differentiation and in particular, is restricted to the basal layer of epithelial
cells. In colon cancer, β6 expression is similarly maximal at the advancing edges of
tumour cell islands (Agrez et al, 1996).
Hence, modulation of MAP kinase interaction with the β6 subunit in epithelial
cells may be used in the prophylaxis or treatment of cancer of the lip, tongue, salivary
glands, gums, floor and other areas of the mouth, oropharynx, nasopharynx,
hypopharynx and other oral cavities, oesophagus, stomach, small intestine, duodenum,
colon, rectum, gallbladder, pancreas, larynx, trachea, bronchus, lung, female and male
breast, uterus, cervix, ovary, vagina, vulva, prostate, testes, penis, bladder, kidney,
thyroid and skin.
In terms of prophylactic use, a method of the invention may find application in
protecting against ultraviolet-induced skin cancer, lung cancer in smokers, cancer of the
gut where polyps are present as in polyposis coli or other inheritable disease where a pre- disposition to the development of polyps exists, breast cancer in high risk patients with a
familial history of breast cancer or otherwise identified as carrying known mutations of
the breast cancer susceptibility gene BRCA1 or BRCA2 associated with breast cancer,
transitional cell cancers arising from bladder papillomas, and cancer of the cervix in
individuals deemed to be at high risk.
The epithelial restricted integrin subunit βό is shown herein to interact with at least
MAP kinases ERK2 and JNK-1, and its down-regulation in the present study
dramatically suppresses growth of colon cancer. Hence, therapeutic strategies to inhibit
growth and proliferation of colon cancer and growth of other malignant cancer cells
growth by switching off either the permissive βό integrin and/or inhibiting the β6 MAP
kinase interaction are of particular interest. In particular, the fact that βό expression may
be significantly upregulated on tumour cells compared to normal cells offers the
potential for tumour cell specificity substantially without impairment of normal cellular
function. Down-regulation of the functional activity of an integrin can be achieved in a
number of ways and in particular, by preventing or down-regulating the expression of
the integrin molecule or by inhibiting the signalling function of the integrin.
Gene therapy is one strategy for treating cancers of different types. The use of
recombinant adenoviruses has for instance been utilised for restoring expression of
polypeptide encoded by a wild-type p53 tumour suppressor gene (Bookstein et al, 1996),
and adenoviral vectors for expression of wild-type p53 have been shown to suppress
growth of human colon cancers by as much as 60% in animal models following intra-
tumoural injection of recombinant virus (Spitz et al, 1996). Rather than replacing a defective gene with one encoding a polypeptide the
expression of which restores normal function of the cell as in the above examples, the
possibility exists to achieve down regulation of cancer cells such as colon cells by
introducing a gene that encodes an integrin subunit in which the binding domain for
interacting with a MAP kinase has been rendered defective by mutagenesis, or in which
the binding domain has been wholly or partially deleted, to thereby achieve down
regulation through the inhibition of the MAP kinase integrin interaction. The defective
integrin subunit will nevertheless usually be able to associate with its normal partner
integrin subunit and be expressed on the cell membrane. Preferably, the defective
integrin subunit will be expressed at a higher level than the corresponding wild-type
integrin subunit such that down regulation is achieved by a dominant negative effect.
Alternatively, such therapy may involve the introduction and expression of a nucleic
acid sequence that encodes a fragment or truncated form of an integrin subunit that
excludes the binding domain for the MAP kinase or in which the binding domain has
been partially or wholly deleted or otherwise mutagenised so as to be defective.
Another option is to introduce a nucleic acid sequence encoding a polypeptide
capable of binding to the binding domain on the integrin for the MAP kinase or to the
binding site on the MAP kinase upon being expressed within the cell to thereby inhibit
binding of the MAP kinase to the integrin and thereby achieve down regulation of
cellular activity.
A gene or nucleic acid sequence encoding an integrin subunit may also be
modified such that although the encoded binding domain for the MAP kinase remains
unaltered, the amino acid sequence of a region of the integrin subunit distant from the binding domain, or the amino acid sequence of either one or both regions flanking the
binding domain, is altered to achieve a change in the three dimensional conformation of
the integrin subunit such that the binding of the MAP kinase with the binding domain is
inhibited.
The gene or nucleic acid sequence may be altered to achieve the desired outcome
by the deletion, insertion or substitution of one or more nucleotides such that the
corresponding amino acid sequence is modified to the extent that the binding of the
MAP kinase to the integrin is inhibited. Inhibition in this context may be partial or total
inhibition.
The gene or nucleic acid sequence can be introduced into a cell in an appropriate
expression vector for expression of the gene or nucleic acid sequence
extrachromosomally or more preferably, for facilitating integration of the gene or nucleic
acid sequence into genomic DNA by heterologous or homologous recombination events.
In another strategy, down-regulation of the expression of an integrin subunit such
as βό and hence an integrin heterodimer such as αvβό, is achieved using antisense
nucleic acid sequences (e.g. oligonucleotides) for inhibiting expression of the subunit.
Typically, this will involve expression of a nucleic acid construct incorporating all or
part of a coding region of the gene for the integrin subunit inserted in the reverse
orientation resulting in the synthesis of antisense RNA which inhibits translation of
mRNA encoding the integrin subunit by hybridisation of the antisense RNA to the
mRNA.
More broadly, a method of down regulating an activity of a cell may comprise
contacting the cell with a first nucleic acid molecule that is capable of interacting with a target nucleic acid sequence, or which first nucleic acid molecule is capable of being
transcribed to a nucleic acid molecule capable of interacting with the target sequence
whereby the interaction of the first nucleic acid molecule with the target sequence
inhibits expression of the integrin subunit.
Reference to the first nucleic acid molecule is to be understood as a reference to
any nucleic acid molecule which directly or indirectly facilitates reduction, inhibition or
other form of down regulation of the expression of the integrin molecule. Nucleic acid
molecules which fall within the scope of this definition include antisense sequences
administered to a cell and antisense sequences generated in situ which have sufficient
complementarity with target sequence such as mRNA encoding the integrin subunit or
for instance, a transcription regulatory sequence controlling transcription of the gene
encoding the integrin subunit, to thereby be capable of hybridising with the target
sequence and inhibit the expression of the integrin subunit. The first nucleic acid
molecule may also be a ribozyme capable for instance, specifically binding to that region
of nucleic acid encoding the binding domain of an integrin subunit and cleaving the
nucleic acid.
On a priori grounds, targeting the expression of the βό subunit in malignant cells
such as in colon cancer by means of adenoviral-mediated antisense therapy is preferred
because down-regulating β6 by means of a non-adenoviral approach may increase cell
surface expression of the β5 subunit. The relevance to such therapy is that the
vitronectin-binding integrin αvβ5 promotes adenovirus internalisation (Thomas et al,
1993). Given that abundant αvβ5 is always present on the surface of colon cancer cells
for example, a secondary benefit of inhibiting βό expression during the course of therapy may be the concomitant rise of β5 in cells already transduced with antisense βό nucleic
acid. This is likely to facilitate further virus uptake into such cells since the amount of
integrin αvβ5 present has been shown to be closely related to levels of gene expression
following adenovirus-mediated gene transfer (Takayama et al, 1998).
Typically, an antisense nucleic acid sequence will hybridise with all or part of that
region of the target sense nucleic acid encoding the binding domain. An antisense
nucleic sequence may for instance be capable of hybridising to exon and/or intron
sequences of pre-mRNA. Preferably, an antisense nucleic acid sequence will be
designed for specifically hybridising to mature mRNA in which intron sequences have
been spliced out by normal cellular processing events.
It is not necessary that an antisense nucleic acid sequence have total
complementarity with its target sequence only that substantial complementarity exists
for specificity and to allow hybridisation under cellular conditions. Preferably, an
antisense nucleic sequence will have a complementarity of about 70% or greater, more
preferably about 80% or greater and most preferably, about 90% or 95% or greater.
Preferred antisense sequences are oligonucleotides wherein the complementary sense
nucleotides encode for about 50 amino acids or less, preferably about 35 or 30 amino
acids or less, more preferably less than about 25 or 20 amino acids, most preferably
about 15 amino acids or less and usually between about 5 to about 15 amino acids.
Antisense nucleic acid sequences may be generated in vivo by transcription of a
suitable expression vector within a cell transformed with the vector, or ex vivo and then
be introduced into a target cell to effect down regulation of the MAP kinase integrin
interaction. Antisense sequences will desirably be designed to be resistant to endogenous exonucleases and/or endonucleases to provide in vivo stability in target
cells. Modification to the phosphate backbone, sugar moieties or nucleic acid bases may
also be made to enhance uptake by cells or for instance solubility, and all such
modifications are expressly encompassed. Such modifications include modification of
the phosphodiester linkages between sugar moieties, the utilisation of synthetic
nucleotides and substituted sugar moieties, linkage to liphophilic moieties and the such
like as described in US Patent No. 5,877,309. Methods for the construction of
oligonucleotides for use in antisense therapy have previously been described (see Van
der Krol et al, 1998 Biotechniques 6:958-976; and Stein et al, 1998 Cancer Res 48:2659-
2668; Bachman et al, 1998, J. Mol. Med. 76:126-132).
Any means able to achieve the introduction of a gene or a nucleic acid into a target
cell may be used. Gene transfer methods known in the art include viral and non-viral
transfer methods. Suitable virus into which appropriate viral expression vectors may be
packaged for delivery to target cells include adenovirus (Berkner, 1992; Gorziglia and
Kapikian, 1992); vaccinia virus (Moss, 1992); retroviruses of avian (Petropoulos et al,
1992); murine (Miller, 1992) and human origin (Shimada et al, 1991); herpes viruses
including Herpes Simplex Virus (HSV) and EBV (Margolskee, 1992; Johnson et al,
1992; Fink et al, 1992; Breakfield and Geller, 1997; Freese et al, 1990); papovaviruses
such as SV40 (Madzak et al, 1992), adeno-associated virus (Muzyczka, 1992); BCG and
poliovirus. Particularly preferred virus are replication deficient recombinant adenovirus
(eg. He et al, 1998), Engineered virus may be administered locally or systemically to
achieve delivery of the gene or nucleic acid sequence of interest into a target cell. Gene transfer methods as described above may be used in the provision of
transgenic animals for studying in vivo the effect of for instance, modifying the binding
site of an integrin for a MAP kinase interaction. A transgenic animal is one with cells
that contain heterologous nucleic acid as a result of the deliberate introduction of the
nucleic acid. The nucleic acid may be introduced indirectly by viral transfer as indicated
above or directly by micro injection into a pronucleus of a fertilised egg prior to transfer
of the egg to a surrogate mother for development to term. Alternatively, a gene or
nucleic acid sequence of interest may be introduced into an embryonal stem (ES) cell in
culture and the transformed cell injected into a recipient blastocyst which is then
transferred to a surrogate mother for development to term. Techniques for generation of
transgenic animals are for instance described in US Patent No. 4,873,191; Van der
Putten, 1985; Thompson et al, 1989 and Lo, 1983.
A transgenic animal homozygous for a transferred gene for instance may be
obtained by the mating of animals heterozygous for the gene as will be appreciated.
Both animal cells expressing a heterologous gene or nucleic acid sequence and
transgenic animals in which a particular gene has been mutagenised by homologous
recombination may be provided. A transgenic animal may for instance be a mouse, rat,
hamster or pig. Typically, the transgenic animal will be a mouse.
Agents for modulating the functional activity of a cell arising from the interaction
of a MAP kinase like ERK2 or JNK-1 with a integrin include antagonists and inhibitors
capable of associating with the integrin to thereby inhibit the MAP kinase and integrin
interaction. Antagonists and inhibitors include those agents that act by binding the
integrin adjacent to the binding domain and stearically hindering the interaction of the MAP kinase with the integrin, as well as allostearic inhibitors that distort the binding
domain upon associating with the integrin.
The binding domain of an integrin may be identified and characterised using
protocols and techniques described herein. Specifically, a binding domain may be
localised by assessing the capacity of respective overlapping peptide fragments
corresponding to different regions of the cytoplasmic domain of an integrin subunit to
associate with a MAP kinase. The specific amino acid sequence which constitute the
binding domain for the MAP kinase may then be determined utilising progressively
smaller peptide fragments of the region of the cytoplasmic domain of the integrin
subunit observed to interact with the MAP kinase. In particular, test peptides are readily
synthesised to a desired length involving deletion of an amino acid or amino acids from
either or both ends of the amino acid sequence corresponding to that region each time,
and tested for their ability to associate with the MAP kinase. This process is repeated
until the minimum length peptide capable of associating with the MAP kinase
substantially without compromising the optimum observed level of association is
identified. The specific amino acids that play an active role in the interaction with the
MAP kinase is achieved with the use of further synthesised test peptides in which one or
more amino acids of the sequence are deleted or substituted with a different amino acid
or amino acids to determine the effect on the ability of the peptide of associate with the
MAP kinase. Typically, substitution mutagenesis will involve substitution of selected
ones of the amino acid sequence with alanine or other relatively neutrally charged amino
acid. By deletion is meant deletion of one or more of the amino acids between the N-
terminal and C-terminal amino acid residues of the identified amino acid sequence. Nucleotide and amino acid sequence data for the β6 integrin subunit for instance is
found in Sheppard et al, 1990. The amino acid sequence for βό is also set out in SEQ ID
NO: 1. Reference to such published data allows the ready design of peptide fragments of
an integrin subunit cytopasmic domain for use in the identification and localisation of
the binding domain for a MAP kinase, and the indentification of the corresponding
nucleic acid sequence encoding such peptide fragments as well as the amino acid
sequence of the binding domain.
Localisation and characterisation of the binding domain for a MAP kinase enables
the design of agents for use in down regulating the functional activity of the integrin and
more particularly, the binding interaction of the MAP kinase with the integrin. This will
typically involve determining the physical properties of the binding domain such as size
and charge distribution, and the tertiary structure of the binding domain.
Specifically, at least the region of the integrin containing the binding domain is
modelled taking into account the stereochemistry and physical properties of the binding
domain such as size and charge distribution as well as its tliree dimensional structure as
determined using x-ray chrstallography, nuclear magnetic resonance and/or
commercially available computer modelling software. In a variation of this approach,
the modelling will take into account the interaction of the binding domain with the MAP
kinase such that any change in conformation arising from the interaction may be taken in
to account in the design of an agent. Modelling flanking regions adjacent the binding
domain also allows .the design of agents for associating with such flanking regions but
which are nevertheless capable of inhibiting the binding domain MAP kinase interaction either by stearic hinderence or by distorting the conformation of the binding domain (eg.
allostearic inhibitors).
The design of a mimetic of the binding domain will usually involve selecting or
deriving a template molecule onto which chemical groups are grafted to provide required
physical and chemical characteristics. The selection of template molecule and chemical
groups is based on ease of synthesis, likely pharmacological acceptability, risk of or
potential for degradation in vivo, stability and maintenance of biological activity upon
administration. Pharmacological acceptability and the like are also taken into
consideration in the design of other agent types.
In order to constrain a polypeptide or other agent in a three dimensional
conformation required for binding, it may be synthesised with side chain structures or
otherwise be incorporated into a molecule with a known stable structure in vivo. In
particular, a polypeptide or the like may be incorporated into an amino acid sequence at
least part of which folds into a β-pleated sheet or helical structure such as an α-helix (eg.
see Dedhar et al., 1997).
A polypeptide or other agent may also be cyclised to provide enhanced rigidity and
thereby stability in vivo. Various methods for cyclising peptides, fusion proteins or the
like are known (eg. Schiller et al., 1985). For example, a synthetic peptide incorporating
two cysteine residues distanced from each other along the peptide may be cyclised by the
oxidation of the thiol groups of the residues to form a disulfide bridge between them.
Cyclisation may also be achieved by the formation of a peptide bond between the N-
terminal and C-terminal amino acids of a synthetic peptide or for instance through the
formation of a bond between the positively charged amino group on the side chain of a lysine residue and the negatively charged carboxyl group on the side chain of a
glutamine acid residue. As will be understood, the position of the various amino acid
residues between which such bonds are formed will determine the size of the cycle.
Variation of cycle size for optimisation of binding affinity may be achieved by
synthesising peptides in which the position of amino acids for achieving cyclisation has
been altered. The formation of direct chemical bonds between amino acids or the use of
any suitable linker to achieve cyclisation is also well within the scope of the skilled
addressee.
Further strategies for identifying possible agents include large scale screening
techniques as are known in the art. For example, peptide library technology provides an
efficient way of testing a vast number of potential agents. Such libraries and their use
are well known. Prospective agents identified may be then further evaluated in suitable
activity, competitive and other immunoassays. A method of screening for an agent or
evaluating whether an agent is capable of binding to a MAP kinase or an integrin and
thereby inhibiting binding of the MAP kinase to the binding domain of the integrin may
for instance involve utilising the agent in an assay whereby the agent has the opportunity
of binding to the MAP kinase or the integrin in the presence of the integrin or the MAP
kinase as the case may be or prior to the addition of the integrin or the MAP kinase, and
determining whether inhibition of binding of the MAP kinase to the integrin results. An
alternate screening method may for instance involve selecting a test agent capable of
binding with the integrin or MAP kinase, measuring cellular activity in the presence of
the test agent, and comparing that activity with cellular activity in the absence of the test
agent. Cellular activity may be assessed by cell growth as indicated by [ H]-thymidine uptake or other measurement of cellular activity. As will be understood, a difference in
observed functional activity in the presence of the test agent is indicative of the
modulating effect provided by the test agent.
It will also be understood that the integrin in the context of such assays may be an
integrin subunit or polypeptide or fragment incorporating the binding domain of the
integrin for the MAP kinase, or a homolog, analog, variant or derivative of such a
molecule to which the MAP kinase is capable of binding. In addition, determination of
whether an agent is capable of binding to the binding domain of an integrin may be
readily achieved by using a polypeptide or fragment as described herein consisting of the
binding domain of the integrin or core amino acid sequence of the binding domain that
directly participates in the binding interaction with the MAP kinase or analogs or the like
of such molecules.
It is not necessary that an agent be proteinaceous in character and indeed, mimetics
may be prepared which may not be a polypeptide at all but which nevertheless possess
the capability of binding with the integrin.
Polypeptides including fusion proteins and fragments of an integrin subunit
comprising the binding domain for a MAP kinase or incorporating sufficient core amino
acid sequence of the binding domain for binding by the MAP kinase are encompassed by
the present invention. Typically, a polypeptide of the invention will have a length of
about 150 amino acids or less, more preferably about 100 or 50 amino acids or less and
generally, less than about 40 amino acids. Preferably, the length will be from between
about 5 to about 30 amino acids, and more preferably from between about 5 amino acids
and about 25 amino acids. Preferably, a polypeptide will comprise or incorporate the amino acid sequence RSKAKWQTGTNPLYR, more usually the amino acid sequence
RSKAKNPLYR, or one or both of sequences RSKAK and NPLYR.
Polypeptides and fusion proteins or the like may be synthesised or produced using
conventional recombinant techniques. Nucleic acid encoding a fusion protein may for
instance be provided via the joining together of separate DNA fragments encoding
peptides or polypeptides having desired three dimensional conformations and/or other
characteristics by employing blunt-ended termini and oligonucleotide linkers, digestion
to provide staggered termini as appropriate, and ligation of cohesive ends prior to
insertion of the resultant chimeric sequence into a suitable expression vector.
Alternatively, PCR amplification of DNA fragments can be utilised employing primers
which give rise to amplicons with complementary termini which can be subsequently
ligated together (eg. see Current Protocols in Molecular Biology. John Wiley & Sons,
1992).
Nucleic acid sequences encoding for the polypeptides, mutagenised integrin
subunits and the like as described herein are also encompassed by the present invention
as are the respective complementary antisense nucleic acid sequences.
Sense oligonucleotides encoding for the binding site of an integrin subunit or a
partial amino acid sequence of a binding domain, and the complementary antisense
oligonucleotides are particularly suitable for use as primers in polymerase chain reaction
(PCR) amplification methods or as probes for detection of the presence the respective
target nucleic acid sequences with which they hybridise such as in Southern blotting
protocols, or in affinity chromatography purification of the target nucleic acid sequence.
32
Probes may be labelled with for instance commonly used isotopes such as P , fluorophores, chemiluminescent agents and enzymes (see eg. Essential Molecular
Biology. A Practical Approach Vol. II, Oxford University Press, 1993; Current
Protocols in Molecular Biology, Ausubel FM., John Wiley & Sons Inc., 1998). The
choice of a label will vary depending on the degree of sensitivity required, ease of
conjugation with the probe, safety and other factors.
Oligonucleotides for use as probes or primers will usually have a length of less
than about 60 nucleotides, usually less than about 50 or 40 nucleotides preferably,
between about 14 and about 30 nucleotides, and more preferably, between about 14 and
about 25 nucleotides. While it is desirable that a primer or probe has 100%
complementarity with its target sequence, oligonucleotides may be designed with less
complementarity but which nevertheless hybridise with the target sequence. Typically, a
primer or probe will have a complementarity of about 70% or greater, more preferably
about 80% or greater and most preferably about 90% or 95%, or greater. A probe will
generally be designed for being capable of hybridising with its target nucleic acid
sequence under moderate or high stringency wash conditions. Moderate stringency wash
conditions are for example those that employ 0.2 x SSC (0.015M NaCl/0.0015M sodium
citrate) /0.1 % SDS (sodium dodecylsulfate) wash buffer at 42°C. High stringency wash
conditions employ for instance, 0.1 x SSC wash buffer at 68°C. Generally, the content
of purine relative to the content of pyrimidine nucleotides in the region of target nucleic
acid of interest will be taken into account in the design of such primers and probes as
will be their length in accordance with well accepted principles known in the art.
In addition, the present invention provides vectors incorporating nucleic acid
sequences of the invention. The term "vector" is to be taken to mean a nucleic acid molecule capable of facilitating the transport of a nucleic acid sequence inserted therein
into a cell and includes expression vectors and cloning vectors.
Suitable expression vectors include plasmids and cosmids capable of expression of
a DNA (eg. genomic DNA or cDNA) insert. An expression vector will typically include
transcriptional regulatory control sequences to which the inserted nucleic acid sequence
is operably linked. By "operably linked" is meant the nucleic acid insert is linked to the
transcriptional regulatory control sequences for permitting transcription of the inserted
sequence without a shift in the reading frame of the insert. Such transcriptional
regulatory control sequences include promotors for facilitating binding of RNA
polymerase to initiate transcription, expression control elements for enabling binding of
ribosomes to transcribed mRNA, and enhancers for modulating promotor activity. A
promotor may be a tissue specific promotor which facilitates transcription of the nucleic
acid insert only in specific cell lineages and not in other cell types or only to a relatively
low level in such other cell types. The design of an expression vector will depend on the
host cell to be transfected, the mode of transfection and the desired level of transcription
of the nucleic acid insert.
Numerous expression vectors suitable for transfection of prokaryotic (eg. bacterial)
or eukaryotic (eg yeast, insect or mammalian cells) are known in the art. Expression
vectors suitable for transfection of eukaryotic cells include pSV2neo, pEF.PGK.puro,
pTk2, pRc/CNV, pcDNAI/neo, non-replicating adenoviral shuttle vectors incorporating
the polyadenylation site and elongation factor 1-α promotor and pAdEasy based
expression vectors most preferably incorporating a cytomegalovirus (CMV) promotor
(eg. see He et al, 1998). For expression in insect cells, baculovirus expression vectors may be utilised examples of which include pVL based vectors such as pVL1392, and
pVL941, and pAcUW based vectors such as pAcUWl. Viral expression vectors are
particularly preferred.
Typical cloning vectors incorporate an origin of replication (ori) for permitting
efficient replication of the vector, a reporter or marker gene for enabling selection of host
cells transformed with the vector, and restriction enzyme cleavage sites for facilitating
the insertion and subsequent excision of the nucleic acid sequence of interest.
Preferably, the cloning vector has a polylinker sequence incorporating an array of
restriction sites. The marker gene may be drug-resistance gene (eg. Ampr for ampicillin
resistance), a gene encoding an enzyme such as chloramphenicol acetyltransferase
(CAT), β-lactamase, adenosine deaminase (ADA), aminoglycoside phosphotransferase
(APH), dihydrofolate reductase (DHFR), hygromycin-B-phosphotransferase (HPH),
thymidine kinase (TK), or for instance β-galactosidase encoded by the E. coli lacZ gene
(LacZ'). Yeast reporter genes include imidazole glycerolphosphate dehydratase (HIS3),
N-(5'-phosphoribosyl)-anthranilate isomerase (TRP1) and β-isopropylmalate
dehydrogenase (LEU2). As will be appreciated, expression vectors of the invention may
also incorporate such marker genes.
Cloning vectors include cloning vectors for mammalian, yeast and insect cells.
Particular vectors that may find application include pBR322 based vectors and pUC
vectors such as pUC 118 and pUC 119. Suitable expression and cloning vectors are for
instance described in Molecular Cloning. A Laboratory Manual., Sambrook et al., 2nd
Ed. Cold Spring Harbour Laboratory., 1989. Host cells suitable for being transformed by vectors of the invention include
bacteria such as E. coli, Bacillus such as B. subtilis, Streptomyces and Pseudomonas
bacterial strains, yeast such as Sacchromyces and Pichia, insect cells, avian cells and
mammalian cells such as Chinese Hamster Ovary cells (CHO), COS, HeLa, HaRas,
WI38, SW480, and NIH3T3 cells. Host cells may be cultured in a suitable culture
medium and under conditions for facilitating expression of nucleic acid sequences of the
invention or replication of cloning vectors, prior to purification from the host cells,
and/or supernatants as the case may be using standard purification techniques.
Rather than utilising viral mediated transfection of cells, nucleic acid sequences
and other molecules of the invention may also be delivered to a cell in vitro or in vivo by
liposome mediated transfection. The liposomes may carry targeting molecules for
maximising delivery of the agent or agents contained therein to specific cell types of
interest. Such targeting molecules may be for instance antibodies, ligands or cell surface
receptors for facilitating fusion of liposomes to the specific cells of interest. Agents may
also be intracellularly delivered in vitro using conventional cold or heat shock techniques
or for instance, calcium phosphate coprecipitation or electroporation protocols. Yet
another strategy is to design the agent to have the inherent ability to pass across the lipid
bilayer of a cell.
A particularly preferred way of achieving intracellular delivery of an agent is to
use "carrier peptides" which have the ability to deliver macro-molecules across cell
membranes in an energy-independent manner (Prociantz, 1996). Indeed, such peptides
provide the possibility of both testing potential agents in cell culture without drastically
altering cell membrane integrity and of delivering agents in vivo. Carrier peptides that are known in the art include penetratins and variants thereof (Derossi et al, 1994, 1996),
human immunodeficiency virus Tat derived peptide (Prociantz, 1996), and transportan
derived peptide (Pooga et al. 1998). Indeed, carrier peptides have been successfully used
to facilitate internalisation of mimetics of Src homology 2 binding sites, and peptides
which inhibit protein kinase C mediated axon development and CD44 (hyaluronate
receptor) dependent migration (Theodore et al, 1995; Williams et al, 1997; Peck Isacke,
1998; Derossi et al, 1998).
Specific targetting to β6-expressing cancer cells may also be achieved by coupling
humanised anti-β6 antibody to carrier molecules such as penetratin coupled to an agent
capable of inhibiting binding of a MAP kinase with an integrin expressed by the cell or
down regulation of the expression of the integrin. Coupling may for instance be by a
peptide bond or disulfide bridge. Given that β6 expression enhances effective
proteolysis at the cell surface by matrix metalloproteinase-9 (Agrez et al, 1999), such
targetting approaches may include engineering an MMP-9 cleavage site between the
antibody and the carrier peptide penetratin to facilitate internalisation of the pentratin-
agent complex. Another approach may employ coupling the penetratin-agent complex to
β6 integrin receptor-targetted peptides, targetted for binding to the extracellular βό
domain by virtue of their DLXXL sequence. For example, a ligand recognition motif for
αVβό integrin, RTDLDSLRTYTL (Kraft et al, 1999) may be used in conjunction with
or without an engineered MMP-9 cleavage site to release the penetratin-agent complex at
the cell surface. Further protocol for targetting nucleic acids to cells by targetting
integrins is described in Bachmann et al, 1998. The toxicity profile of an agent of the invention may be tested on normal and
malignant cells by evaluation of cell morphology, trypan-blue exclusion, assessment of
apoptosis and cell proliferation studies (eg cell counts, H-thymidine uptake and MTT
assay).
Agents of the invention may be co-administered with one or more other
compounds or drugs. For example, an agent or agents may be administered in
combination with antisense therapy or chemotherapeutic drugs. Alternatively, an agent
may be administered in conjunction with antisense therapy and/or chemotherapeutic
drugs. By "co-administered" is meant simultaneous administration in the same
formulation or in two different formulations by the same or different routes, or
sequential administration by the same or different routes. By "sequential" administration
is meant a time difference of between the administration of the agents, drugs and other
therapies which can be administered in any order. The time difference may range from
very short times up to hours or for instance days or weeks.
The agent or agents will typically be formulated into pharmaceutical composition
incorporating pharmaceutically acceptable carriers, diluents and/or excipients for
administration to the intended subject.
Pharmaceutical forms include sterile aqueous solutions suitable for injection,
(where the agent or agents is water soluble) and sterile powders for the extemporaneous
preparation of sterile injectable solutions. Such injectable compositions will be fluid to
the extent that the syringability exists and typically, will be stable to allow for storage
after manufacture. The carrier may be a solvent or dispersion medium containing one or
more of ethanol, polyol (eg glycerol, propylene glycol, liquid polyethylene glycol and the like), vegetable oils, and suitable mixtures thereof. Fluidity may be maintained by
the use of a coating such as lecithin, by the maintenance of the required particle size in
the case of a dispersion and by the use of surfactants.
Sterile injectable solutions will typically be prepared by incorporating the active
agents in the desired amount in the appropriate solvent with various other components
enumerated above, prior to sterilising the solution by filtration. Generally, dispersions
will be prepared by incorporating the sterile active agents into a sterile vehicle which
contains the dispersion medium and other components. In the case of sterile powders for
the preparation of sterile injectable solutions, preferred methods of preparation are
vacuum drying and freeze-drying techniques which yield a powder of the active agent
plus any additional desired ingredient from previously sterile filtered solutions thereof.
For oral administration, the active agents may be formulated into any orally
acceptable carrier deemed suitable. In particular, the active ingredient may be
formulated with an inert diluent, an assimilable edible carrier or it may be enclosed in a
hard or soft shell gelatin capsule. Alternatively, it may be incorporated directly into
food. Moreover, an active agent may be incorporated with excipients and used in the
form of ingestable tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups,
and the like.
Such compositions will generally contain at least about 1% by weight of the active
agent or agents. The percentage may of course be varied and may conveniently be
between about 5 to about 80% w/w of the composition or preparation. As will be
appreciated, the amount of active agent or agents in such compositions will be such that
a suitable effective dosage will be delivered to the subject taking into account the proposed mode of administration. Preferred oral compositions according to the
invention will contain between about 0.1 μg and 2000mg of each active agent,
respectively.
Active agents may also be formulated into topically acceptable carriers
conventionally used for forming creams, lotions, ointments and the like for internal or
external application.
Typically, a composition of the invention will incorporate one or more
preservatives such as parabens, chlorobutanol, phenol, sorbic acid, and thimersal. In
many cases, a composition may furthermore include isotonic agents such as sugars or
sodium chloride. In addition, prolonged absoφtion of the composition may be brought
about by the use in the compositions of agents for delaying absoφtion such as
aluminium monosterate and gelatin.
Tablets, troches, pills, capsules and the like may also contain one or more of the
following: a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients
such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch,
alginic acid and the like; a lubricant such as magnesium sterate; a sweetening agent such
as sucrose, lactose or saccharin or a flavouring agent such as peppermint, oil of
wintergreen, orange or cherry flavouring. When the dosage unit form is a capsule, it
may contain in addition to one or more of the above ingredients a liquid carrier. Various
other ingredients may be present as coatings or to otherwise modify the physical form of
the dosage unit. For instance, tablets, pills or capsules may be coated with shellac,
sugars or both. In addition, an active agent may be incoφorated into any suitable
sustained-release preparation or formulation. Pharmaceutically acceptable carriers, diluents and/or excipients include any
suitable conventionally known solvents, dispersion media and isotonic preparations or
solutions. Use of such ingredients and media for pharmaceutically active substances is
well known. Except insofar as any conventional media or agent is incompatible with the
active agent, use thereof in therapeutic and prophylactic compositions is contemplated.
Supplementary active ingredients can also be incoφorated into the compositions if
desired.
It is particularly preferred to formulate parenteral compositions in dosage unit form
for ease of administration and uniformity of dosage. Dosage unit form as used herein is
to be taken to mean physically discrete units suited as unitary dosages for the subject to
be treated, each unit containing a predetermined quantity of active agent calculated to
produce the desired therapeutic or prophylactic effect in association with the relevant
carrier, diluent and/or excipient.
A unit dosage formed will generally contain each active agent in amounts ranging
from about 0.5 μg to about 2000mg/ml of carrier respectively.
A pharmaceutical composition may also comprise vectors capable of transfecting
target cells where the vector carries a nucleic acid molecule for modulating functional
activity or expression of an integrin or MAP kinase. The vector may for instance, be
packaged into a suitable virus for delivery of the vector into target cells as described
above.
The dosage of an active agent will depend on a number of factors including
whether the agent is to be administered for prophylactic or therapeutic use, the condition
for which the agent is intended to be administered, the severity of the condition, the age of the subject, and related factors including weight and general health of the subject as
may be determined by the physician or attendant in accordance with accepted principles.
Indeed, a low dosage may initially be given which is subsequently increased at each
administration following evaluation of the subjects response. Similarly, frequency of
administration may be determined in the same way that is, by continuously monitoring
the subjects response between each dosage and if necessary, increasing the frequency of
administration or alternatively, reducing the frequency of administration.
The route of administration of a pharmaceutical composition will again depend on
the nature of the condition for which the composition is to be administered. Suitable
routes of administration include but are not limited to respiritoraly, intratracheally,
nasopharyngeally, intraveneously, intraperitonealy, subcutaneously, intracranialy,
intradermially, intramuscularly, intraoccularly, intrathecally, intranasally, by infusion,
orally, rectally, via IV group patch, topically and by implant. With respect to
intravenous routes, particularly suitable routes are via injection into blood vessels which
supply a tumour or particular organs to be treated. Agents may also be delivered into
cavities such for example the pleural or peritoneal cavity, or be injected directly into
tumour tissue.
The production of antibodies and monoclonal antibodies is well established in the
art (eg. see Antibodies, A Laboratory Manual. Harlow & Lane Eds. Cold Spring Harbour
Press, 1988). For polyclonal antibodies, a mammal such as a sheep or rat for instance is
immunised with a polypeptide of the invention and antisera subsequently isolated prior
to purification of the antibodies therefrom by standard affinity chromatography
techniques such as Sepharose-Protein A chromatography. Desirably, the mammal is periodically challenged with the relevant antigen to establish and/or maintain high
antibody titre. To produce monoclonal antibodies, B lymphocytes can be isolated from
the immunised mammal and fused with immortalising cells (eg. myeloma cells) by
standard somatic cell fusion techniques (eg. utilising polyethylene glycol) to produce
hybridoma cells (Kohler and Milstein, 1975; see also Handbook of Experimental
Immunology, Weir et al Eds. Blackwell Scientific Publications. 4th Ed. 1986). The
resulting hybridoma cells may then be screened for production of antibodies specific for
the peptide by an enzyme linked immunosorbant assay (ELISA) or other immunoassay.
Conventionally used methods for preparing monoclonal antibodies include those
involving the use of Epstein-Barr virus (Cole et al. Monoclonal Antibodies and Cancer
Therapy, Allen R. Liss Inc. pp. 77-96, 1985). The term "antibody" or "antibodies" as
used herein is to be taken to include within its scope entire intact antibodies as well as
binding fragments thereof such as Fab and (Fab')2 fragments which may be obtained by
papain or pepsin proteolytic cleavage, respectively.
An antibody of the invention may be labelled for enabling detection of antibody
binding in immunoassays including competitive inhibition assays. A "label" may be any
molecule which by its nature is capable of providing or causing the production of an
analytically identifiable signal which allows the detection of an antibody and antigen
complex. Such detection may be qualitative or quantitative. An antibody can for
instance be labelled with radioisotopes including 32 P, 125 I or 131 I, an enzyme, a
fluorescent label, chemiluminescent molecule or for instance an affinity label such as
biotin, avidin, streptavidin and the like. An enzyme can be conjugated with an antibody by means of coupling agents such
as gluteraldehyde, carbodiimides, or for instance periodate although a wide variety of
conjugation techniques exist. Commonly used enzymes include horseradish peroxidase,
glucose oxidase, β-galactosidase and alkaline phosphatase amongst others. Detection
utilising enzymes is achieved with use of a suitable substrate for the selected enzyme.
The substrate is generally chosen for the production upon hydrolysis of a detectable
colour change. However, fluorogenic substrates may also be used which yield a
fluorescence product rather than a chromogen. Suitable fluorescent labels are those
capable of being conjugated to an antibody substantially without altering the binding
capacity of the antibody and include fluorescein, phycoerythrin (PE) and rhodamine
which emit light at a characteristic wavelength in the colour range following illumination
with light at a different wavelength. Methods for labelling of antibodies can be found in
Current Protocols in Molecular Biology. Ausubel FM., John Wiley & Sons Inc.
Immunoassays in which antibodies of the invention may be utilised include
radioimmunoassays (RIA) and ELISA (eg., see Handbook of Experimental Immunology,
Weir et al., Vol. 1-4, Blackwell Scientific Publications 4th Edition, 1986). Such assays
include those in which a target antigen is detected by direct binding with a labelled
antibody, and those in which the target antigen is bound by a first antibody, typically
immobilised on a solid substrate (eg., a microtitre tissue culture plate formed from a
suitable plastics material such as polystyrene or the like) and a labelled second antibody
specific for the first antibody used to form an antigen-first antibody-second antibody
complex that is detected by a signal emitted by the label. Sandwich techniques in which
the antigen is immobilised by an antibody for presentation to a labelled second antibody specific for the antigen are also well known. An antibody can be bound to a solid
substrate covalently utilising commonly used amide or ester linkers, or by adsoφtion.
Optimal concentrations of antibodies, temperatures, incubation times and other assay
conditions can be determined by the skilled addressee with reference to conventional
assay methodology and the application of routine experimentation.
Antibodies and other molecules of the invention including polypeptides and
oligonucleotides as described herein when bound to a solid support can be used in
affinity chromatography for the purification of a binding partner for which they are
specific. In particular, a polypeptide either alone or as a fusion protein comprising the
binding domain for a MAP kinase for instance can be utilised in the purification,
isolation or concentration of the MAP kinase. It may also be used for assaying levels of
MMP kinase in cell extracts. Similarly, an antibody that specifically binds to the
binding domain of an integrin has use in the purification of the integrin or fragments
thereof incoφorating the binding domain from a relatively crude preparation or mixture.
Suitable solid supports include agarose, sepharose and other commercially available
supports such as beads formed from latex, polystyrene, polypropylene, dextran, glass or
synthetic resins, typically packed in an affinity column through which the relevant
sample containing the binding partner is passed at a pH and conditions (eg., low salt
concentration) under which the binding partner becomes bound by the antibody,
polypeptide or other such molecule. The column is then washed utilising a suitable
buffer whereby the binding partner is retained bound on the column, prior to being
eluted therefrom utilising a suitable elution buffer (eg., with a higher salt concentration
and at an altered pH, typically pH 2.5 or pH 11) that facilitates the release of the binding partner from the affinity column, and collected. Protocols for affinity chromatography
are described in Current Protocols in Molecular Biology-Ausubel FM., John Wiley &
Sons Inc. Buffers and conditions utilised for the purification, isolation or concentration
of a binding partner will vary depending on the affinity the antibody, polypeptide or the
like for the binding partner.
A kit for use in assays as described herein may include one or more of an antibody,
polypeptide, vector, nucleic acid or other molecule of the invention. The kit may also
comprise one or more other reagents such as washing solutions, dilution buffers and the
like together with instructions for use. The antibody or other molecule of the invention
may or may not be labelled, bound to a solid support or be coupled with another
molecule. Particularly preferred kits are those provided for use in affinity
chromotography or RIA, ELISA or other type of immunoassay.
The present invention will be described herein after with reference to a number of
examples.
Example 1
Anti-integrin antibodies and antibodies against the β 1 subunit have been shown to
inhibit proliferation of retinal pigment epithelial cells (Hergott et al, 1993). In
endothelial cells, inhibition of cell-matrix interactions by anti-integrin antibodies
specifically against α2βl converts the cells from a proliferative to a differentiated
phenotype (Gamble et al, 1993). In another study, synthetic peptides containing the
integrin recognition sequence arginine-glycine-aspartate (RGD) have been shown to inhibit tumour cell invasion in vitro and tumour metastases from melanoma in an animal
model (Humphries et ;al, 1986; Gehlsen et al, 1988).
In colon cancer, the contribution made by the α5βl receptor to regulation of
growth appears to be ligand (fibronectin)-dependent. For example, induced expression
of α5βl in human colon cancer cells constitutively lacking this integrin has been shown
to result in decreased tumour cell proliferation in vitro (Varner et al, 1995).
Interestingly, when the appropriate ligand was present, cell proliferation was restored,
indicating that the unoccupied receptor mediated a negative growth signal in these cells
(Varner et al, 1995). Moreover, induction of α5βl expression was associated with a
marked reduction of tumourigenicity in immune-deficient mice. Failure to ligate all of
the tumour cell α5βl molecules with sufficient murine fibronectin most likely accounts
for the in vivo tumour suppression in these studies (Varner et al, 1995). In the present
study the effect of down-regulating αvβό expression on colon cancer growth was
examined.
1.1 Methods
1.1.1 Generation of sense and antisense βό constructs in pEF.PGK.puro vector.
For βό antisense constructs, βό cDNA was excised from the vector pcDNAlneo βό
(Weinacker et al, 1994) using the restriction enzymes SnaBl and Xbal. This produced a
5' overhang (Xbal) and a 3' blunt end (SnaBl). The 5' overhang was blunted with
Klenow (Promega) prior to ligation. pEF.PGK.puro vector (a gift from D. Huang, the
Walter & Eliza Hall Institute, Melbourne Australia) was cut with EcoRV which
produced blunt ends. The pEF.puro vector was de-phosphorylated using calf intestinal
alkaline phosphatase (CIAP, Promega) and the βό cDNA ligated overnight into pEF.puro using T4 DNA ligase (Promega) at a ratio of vector: insert of 1 :5. After
ligation all the reaction mix was used for transformation into competent JM109 cells and
the cells plated onto LB plates containing ampicillin. After overnight incubation at
37°C, colonies were selected, the plasmid DNA extracted by microprepping, and the
DNA cut with BstEl 1 to confirm the antisense orientation of β6. Digestion with BstEl 1
produced the expected two fragments, one of 6.0 basepairs and the other 3.5 basepairs.
To provide β6 sense constructs, βό cDNA was also excised from the pcDNAlneo
b6 vector using the restriction enzymes Snabl and Xbal as described above. The
pEF.puro vector was then cut with Xbal and EcoRV and the insert was ligated in sense
direction into the pEF.PGK.puro vector without having to blunt the insert. The ligation
reaction was prepared at a ratio of vector: insert of 1 : 1. Digestion with BstEl 1 produced
two expected fragments, one of 7.5 basepairs and the other 2.0 basepairs.
1.1.2 Transfection of WiDr and HT29 colon cancer cells
The human colon cancer cell lines, WiDr and HT29 were obtained from the
American Type Culture Collection (ATCC), Rockville, Maryland, USA and maintained
as monolayers in standard medium comprising Dulbecco's Modified Eagle's medium
(DMEM; 4.5gm/litre of glucose) with 10% heat-inactivated foetal bovine serum (FBS)
supplemented with HEPES and penicillin/streptomycin. WiDr and HT29 cell lines
constitutively express αvβό. Stable transfectants of WiDr and HT29 cells expressing βό
in sense and antisense direction were generated using lipofectamine and puromycin as
the selection antibiotic. One stable antisense βό transfectant from HT29 cells and three
stable clones from WiDr cells were successfully established for use in all experiments.
Mock transfectants using vector alone were also generated as controls. Initial killing curves performed using a range of concentrations of puromycin established that WiDr
and HT29 cells could be stably transfected with puromycin concentrations of 1.Oμg and
2.5μg/ml, respectively.
1.1.3 Assessment of β6 expression in the transfected cell lines
βό expression was assessed by means of FACScan analyses of parent cell lines and
clones generated therefrom by means of limiting-dilution experiments. Stability of the
transfectants was confirmed regularly by repeated FACScan analyses, and surface
biotinylation and immunoprecipitation as described below.
1.1.4 FACScan analyses
Monolayer cultures of cell lines were harvested with trypsin/EDTA and then
blocked with goat serum at 4°C for 10 min. Cells were washed once with PBS,
incubated with primary antibody against integrin subunits for 20 min at 4°C and then
washed twice with PBS. Cells were then stained with secondary antibody conjugated
with phycoerythrin for 20 min at 4°C, washed twice with PBS and resuspended in 0.5ml
PBS prior to FACScan analysis (Becton Dickinson, Rutherford, New Jersey, USA).
1.1.5 Integrin immunoprecipitation
Cells were harvested with trypsin EDTA, the trypsin neutralised with standard
culture medium, and the cell pellets washed once with cold PBS. Cell pellets were then
exposed to biotin-CNHS-ester (Sigma) in biotinylation buffer (lOmM sodium borate,
150mM sodium chloride, pH 8.8) for 30 mins at 4°C with continuous slow mixing. Cell
pellets were then centrifuged at 4°C, washed twice with cold PBS and exposed to lysis
buffer (containing lOOmM Tris, 150mM NaCl, lmM CaCl2 , 1% Triton, 0.1% SDS and
0.1% NP-40 at pH 7.4 and containing lmM phenylmethylsulfonyl fluoride (PMSF)) for 30 min at 4°C. Lysates were then clarified by ultracentrifugation (10,000g) for 30 min
and the protein content measured using the BCA protein assay reagent kit. Lysates
containing equal protein amounts were pre-cleared with rabbit anti-mouse (RAM)
immunoglobulin coupled to Sepharose-4B beads for 12 hrs. Immunoprecipitations were
carried out indirectly using RAM-Sepharose 4B and analysed by 7.5% SDS-PAGE
under non-reducing conditions.
1.1.6 Reverse transcriptase-PCR (RT-PCR)
mRNA levels for βό expression were evaluated by reverse transcriptase-
polymerase chain reaction (RT-PCR). Total RNA was extracted from cell cultures using
the commercial TriPure isolation reagent based on the method of Chomozynski and
Sacchi (1987). 0.4-2μg of RNA was used to prepared cDNA by reverse transcription.
Briefly, a reaction mixture in a final volume of 40μl containing 8μl of 5 x RT reaction
buffer (250mM Tris, 15mM MgCl2 , 375mM KCL, pH 8.3), 8μl of 2.5mM of each
dNTP, 4μl of lOOmM DTT, 40U of an Rnase inhibitor, Rnasin (Promega, Madison,
Wisconsin, USA), 0.5μg of random hexamers (Promega) and 200U of Moloney murine
leukaemia virus (M-MLV) reverse transcriptase (Promega) were mixed and incubated at
38°C for a minimum of 90min. The reaction was stopped by heating at 95°C for 5 min
and cDNA stored at 4°C until PCR. 2-5μl of this cDNA was combined with 5μl of 10 x
PCR buffer (lOOmM Tris, 500mM KC1, 15mM MgCl2 , pH8.3) 8μl of 1.25mM dNTP
each and 1.25μl of 20μM of both forward and reverse primers. The forward primer
sequence was 5ΑGGATAGTTCTGTTTCCTGC3' and the reverse primer sequence
5ΑTCATAGGAATATTTGGAGG3'. The reaction was initiated by 2.5U of Taq
polymerase in a final volume of 50μl. After an initial 5 min incubation at 94°C, 30 cycles of amplification were performed under the following conditions: 94°C 1 min,
54°C 1 min and 72°C for 1 min. The reaction was stopped by incubating at 72°C for 10
min. To verify that equal amounts of RT product from cells were subjected to PCR
amplification, the same amounts of cDNA were amplified for the "house-keeping" gene
GAPDH using specific primers. The same reaction conditions were used except that the
annealing temperature was changed to 48°C and PCR amplification performed for 35
cycles.
1.2 Results
1.2.1 αvβό expression in HT29 and WiDr transfected cell lines
Transfection of the colon cancer cell lines HT29 and WiDr with the βό gene
construct in an antisense orientation resulted in a marked reduction of βό expression at
the transcript level and on the cell surface as shown in Figs 1 to 4. Transfection of cells
with β6 in the sense orientation did not enhance βό surface expression. However, a
consequence of down-regulation of the β6 subunit in antisense transfectants was a
marked increase in surface expression of the β5 subunit. The changes in surface
expression of β6 and β5 subunits noted on FACScan analyses of antisense β6
transfectants was confirmed by surface-labelling cells with biotin and
immunoprecipitating integrin subunits with either anti-βό mAb (R6G9) or anti-β5 mAb
(P1F6).
1.2.2 Effect of Suppression of αvβό expression on cell binding to fibronectin
The major substrate for αvβό is fibronectin and to investigate the effect that
reduction in βό surface expression in antisense βό transfectants might have on cell-
matrix adhesion, WiDr and HT29 antisense βό transfectants were seeded on fibronectin in adhesion assays. Since both cell lines can adhere to fibronectin through members of
the βl integrin subfamily (either α3βl or α5βl) αvβό-mediated adhesion to fibronectin
was assessed in the absence/presence of blocking anti-βl antibody.
The cell-adhesion assays were performed in wells of non-tissue culture-treated
polystyrene 96-well flat bottom microtitre plates (Nunc, Roskilde, Denmark). Culture
wells were coated with fibronectin, washed with phosphate-buffered saline (PBS) and
then blocked with 0.5% bovine serum albumin (Sigma) in PBS for lhr at 37°C.
Harvested cells were seeded at a density of 10 cells/well for HT29 and 1.5 x 105
cells/well for WiDr cells in 200μl of standard DMEM which lacked FBS but contained
0.5% bovine serum albumin. To block adhesion, cells were incubated with anti-βl
blocking antibodies for 15 min at 4°C before plating. The plates were centrifuged (top
side up) at 10 x g for 5 min, then incubated for 1 hr at 37°C in humidified 5% carbon
dioxide. Non-adherent cells were removed by centrifugation top side down at 48 x g for
5 min. The attached cells were fixed and stained with 0.5% crystal violet (in 20%
methanol and 1% formaldehyde) and the wells washed with phosphate-buffered saline.
The relative number of cells in each well was evaluated by measuring the absorbance at
595nm in a Microplate Reader (Bio-Rad).
With reduction in cell surface expression of βό, βl -independent adhesion of cells
to fibronectin was reduced compared with mock transfectants (containing vector alone)
which express surface βό at similar levels to wild-type cells. The further addition of
blocking anti-αv antibody completely prevented binding to fibronectin. 1.2.3 Effect of suppression of αvβό expression on tumour cell proliferation and tumour growth in vivo
To investigate the effect of diminished αvβό surface expression on cell
proliferation in vitro, WiDr and HT29 antisense β6 transfectants were seeded as
monolayers in 96-well microtitre culture plates (5,000 viable cells per well) in standard
culture medium containing the puromycin selection antibiotic. Cells were pulsed with
lμCi ( H)-thymidine (Amersham) per well for the last 24 hours of each experiment
before automated harvesting and measurement of radioactivity. WiDr wild-type, mock
and antisense β6 transfectants, and HT29 wild-type, mock, sense βό and antisense β6
transfectants were harvested second daily during a six-day culture period.
A marked increase in thymidine incoφoration was observed for WiDR and HT29
cells expressing normal levels of αvβό compared with antisense βό transfectants (see
Figs. 5 and 6).
1.2.4 Effect of Suppression of αvβό expression on Tumour Formation
The ability of HT29 antisense βό transfactants to form tumours in immune-
deficient mice was assessed.
BALB/C female athymic mice (8 weeks of age purchased from the Animal
Resource Centre, Perth, Western Australia) were maintained under pathogen-free
conditions and fed standard mouse chow and water ad lib. The mice were divided into
groups often each and all mice within each group inoculated with a single cell line.
Cells used were WiDr mock (transfected with vector alone) and antisense βό transfected
clones (clones 1 - 3) and HT29 mock, sense and antisense βό cell lines. Mice received
subcutaneous flank injections of 106 viable tumour cells suspended in 0.2ml of standard DMEM culture medium. Animal weights and tumour sizes (breadth and length as
measured with calipers) were recorded weekly. Six weeks following the last injection,
visible subcutaneous tumours were excised, weighed and fixed in 4% formalin. At the
time of euthanasia, all internal organs were routinely inspected for presence of
metastases.
Tumour growth after six weeks following inoculation of HT29 mock and antisense
β6 transfectants is shown in Fig. 7. Measurements of tumour growth for WiDr and
HT29 cells are shown in Figs 8 and 9, respectively. Similar tumour growth profiles for
mock and antisense βό WiDr clones 2 and 3 each inoculated into 10 mice are not shown.
Of a total of 40 mice injected with cells expressing antisense βό (HT29 - 10 mice and
WiDr cells lines, 3 clones - 30 mice) tumours completely disappeared in 93% of animals.
In the remaining 3 animals tumour sizes diminished to 1 mm in size during the six week
period compared with tumours at least 15mm in size from cells expressing normal
levels of β6. To confirm the presence of tumour xenografts histologically at one week
following subcutaneous inoculation of mock and antisense β6 transfectants, tumour
nodules were excised, fixed in formalin and stained with haematoxylin and eosin.
1.2.5 Effect of suppression of αvβό expression on gelatinase B secretion
Serum-free tumour-conditioned medium was collected from each of the 3 WiDr
mock and antisense βό clones and concentrated x 44 for measurement of gelatinase B
using the Biotrak MMP-9 activity assay system (Amersham Pharmacia Biotech, Uppsala
Sweden). Down-regulation of αvβό expression resulted in a marked reduction in
gelatinase B secretion. 1.3 Discussion of Results
Induced expression of βό in Chinese hamster ovary (CHO) cells has been shown to
result in decreased surface expression of the β5 integrin subunit which also partners αv
(Weinacker et al, 1994). The concept of integrin switching depends on the availability
of the promiscuous αv partner subunit. In the present study, the reverse was observed.
As a consequence of down-regulation of βό in colon cancer cells which constitutively
express αvβό, β5 surface expression increased, most likely secondary to increased
availability of the αv subunit partner.
Heterologous expression of αvβό in colon cancer cells has previously been
reported to enhance tumour growth in immune-deficient mice (Agrez et al, 1994).
Suppression of αvβό expression in the present study was shown to result in nearly
complete disappearance of tumours in 93% of animals following subcutaneous
inoculation of tumour cells. Moreover, in the remaining 7% of animals, a 95% reduction
in tumour size was observed over a six week period compared with large tumours seen in
all animals injected with cells in which αvβό expression had not been perturbed. Similar
findings have been described with loss of the classical vitronectin receptor αvβ3 in
melanoma. For example, in experimental animal models, the loss of αvβ3 expression in
melanoma cells has been shown to lead to reduced in vivo proliferation which is restored
upon re-expression of the receptor (Felding-Habermann et al, 1992).
Although the mechanisms involved in αvβό-mediated tumour growth remain to be
elucidated, the present in vitro data show that loss of βό expression is associated with
decreased proliferative capacity of the cells. Taken together with the marked reduction
in gelatinase B secretion seen for colon cancer cells transfected with antisense βό, the findings reported in the present study suggest that intracellular signalling pathways
activated via the integrin αvβό play a major role in promoting progression of this tumour
type.
Example 2
The association of αvβό expression and MAP kinase activity were evaluated using
WiDr, HT29 and SW480 cell lines.
2.1 Methods
2.1.1 S W480 colon cancer βό transfectants
Stable transfectants of S W480 colon cancer cells (ATCC) expressing gene
constructs of either wild-type or mutant forms of the βό integrin subunit or the
expression plasmid only (pcDNAlneo) have been previously described (Agrez et al,
1994). The transfected SW480 cell lines were maintained in standard medium
supplemented with the neomycin analogue G418. Stable transfectants of WiDr and
HT29 cells expressing wild-type β6 in antisense orientation were generated as described
in Example 1.1.2 and maintained in standard medium supplemented with puromycin.
2.1.1 MAP kinase assay.
Cultures of WiDr and HT29 mock and antisense β6 transfectants were established
by seeding 1 x 10 cells/5ml of culture medium in 25cm tissue culture flasks. Cells
were incubated at 37°C in humidified CO2 for 24 hours before serum starvation in
serum-free medium for the next 16 hours. Foetal calf serum was then added to a final
concentration of 10% for 30 mins before MAP kinase assays were performed. Before
each experiment the cells were washed twice with PBS, resuspended in extraction buffer
(lOmM Tris-HCl, 150mM NaCl, 2mM EDTA, 2mM DTT, lmM orthovanadate, lmM PMSF, 4μg/ml aprotonin, 2μg/ml leupeptin and lμg/ml pepstatin, pH 7.4) and sonicated
at a setting of 7, using a Soniprep 150 watt ultrasonic disintegrator for a total of 90
seconds in three 30 second pulses with an interval of 30 seconds between each pulse.
Cellular debris was removed by centrifugation at 900g for 10 min at 4°C. The assay was
performed on equal cell numbers using a MAP kinase assay system (Amersham
Pharmacia Biotech, Uppsala Sweden). The ability of cells to transfer phosphate from
32
[γ PJ-ATP to a synthetic peptide that contains specifically a p42/p44 MAP kinase
phosphorylation site was measured as described in the manufacturer's instructions.
[ P]-labelled peptides were spotted onto PEl -cellulose paper, unbound radioactivity was
washed with 75mM phosphoric acid and bound [ P]-labelled peptides were measured
by liquid scintillation counting. Protein estimation was performed on each cell lysate
used and enzyme activity calculated as described in the manufacturer's instructions.
Where MEK inhibitors, PD98059 and U0126 were used, cells were cultured as described
above and the inhibitors, at a final concentration of 40μM, were added one hour before
the addition of serum to the medium.
2.1.2 Western blotting
To detect the MAP kinases ERK 1/2, cells were lysed in lysis buffer containing
lOOmM Tris, 150mM NaCl, lmM CaC12, 1% Triton, 0.1% SDS and 0.5% NP-40 at
pH7.4, supplemented with enzyme inhibitors (lmM PMSF, lmM sodium orthovanadate,
1 μg/ml pepstatin A, 2.5 μg/ml aprotonin, lmM benzamidine, lμg/ml leupeptin). Lysates
were clarified by ultracentrifugation and equal protein loads electrophoresed in 8% or
10% SDS-PAGE under non-reducing conditions. Electrophoresed proteins were
transferred to nitrocellulose membranes (Biotrace NT, Gelman Sciences, Ann Arbor, MI) in transfer buffer (25mM Tris, 192mM glycine, 20% methanol, 0.1% SDS) for 2
hours at a constant voltage of 40 volts in a Transfer Blot Cell (Bio-Rad). Membranes
were blocked with 5% casein for 1 hr at room temperature and probed with monoclonal
anti-ERK antibodies (ElO which recognises only phosphorylated ERK1/2 ( New
England BioLabs) and SC-1647 which recognises total ERKs, non-phosphorylated and
phosphorylated (Santa Cruz Biotechnology). .In some experiments, membranes were
probed with polyclonal anti-ERK antibody (New England BioLabs) which also
recognises total ERKs. Membranes were then washed three times in Tris buffered
saline, containing 0.1% Tween and incubated with HRP-conjugated goat anti-mouse or
goat anti-rabbit antibody. Blots were visualised by the enhanced chemi-luminescence
detection system according to the manufacturer's instructions (Du Pont).
2.1.3 Integrin β-sύbunit immunoprecipitations
Tumour cells were harvested and divided into two equal aliquots based on cell
counts. One aliquot was surface biotinylated and the cells lysed for integrin
immunoprecipitations as described in Example 1.1.5, with the exception that three
sequential rounds of immuno-precipitation were performed to deplete the lysates of
integrin subunits β5 and β6. Lysis buffer was the same as that used for Western blotting.
The other aliquot, intended for subsequent ERK2 immunoblotting to examine the effect
of integrin immunodepletion on ERKs was not biotinylated but lysed immediately and
divided into three samples each at a protein concentration of lmg/ml. Three sequential
rounds of immunoprecipitation were performed against anti-βό monoclonal antibody,
R6G9 (using isotype matched antibody, IgG2A, and anti-β5 monoclonal antibody, P1F6,
in control immunoprecipitations). The integrin-depleted lysates were electrophoresed in 8% or 10% SDS-PAGE
under non-reducing conditions and transferred to nitrocellulose membranes. Membranes
were blocked with casein as for Western blotting and probed with anti-ERK monoclonal
antibodies ElO and SC-1647. In parallel experiments, the sequentially
immunoprecipitated βό subunit bound to rabbit anti-mouse (RAM) coupled Sepharose
B4 beads was also electrophoresed in 10% SDS-PAGE under non-reducing conditions,
transferred to nitrocellulose membranes, and the membranes probed with anti-ERK
monoclonal antibody ElO which recognises only phosphorylated forms of ERK1/2.
2.3 Results
2.3.1 Effect of serum on MAP kinase activity
The effect of MEK (MAP kinase kinase) inhibitors UO126 and PD98059 on MAP
kinase activity in WiDr and HT29 wild-type cells was tested in the absence/presence of
serum. Adherent cell monolayers on plastic were grown for 24 hrs in standard culture
medium, then washed three times in PBS followed by 16 hrs in culture under serum-free
conditions. Serum was then added for 30 min and MAP kinase activity assessed before
and after addition of serum. The addition of serum markedly stimulated MAP kinase
activity for both cell lines and was inhibitable by both MEK inhibitors.
2.3.2 Effect of altered β6 expression on MAP kinase activity following serum
stimulation.
In these experiments, WiDr transfectants (3 mock and 3 antisense βό clones),
HT29 transfectants (mock and antisense β6) and SW480 β6 transfectants (mock and
sense βό) were serum-starved for 16 hrs followed by 30 mins exposure to serum.
Increased expression of αvβό was associated with a marked increase in MAP kinase activity upon serum stimulation compared with cells lacking αvβό altogether (SW480
mock). Cells in which βό expression had been down-regulated (antisense βό
transfectants) exhibited suppressed MAP kinase activity. Induced expression of β6 in
the non-β6-expressing colon cancer cell line SW480 resulted in a three fold increase in
serum-dependent MAP kinase activity.
2.3.3 αvβό binds an extracellular signal-related kinase (ERK).
A highly surprising finding arising from phage display screening which undeφins
the present work is that the integrin βό cytoplasmic domain binds a MAP kinase. Use of
phage display to screen a γgt cDNA colon cancer cell library with the βό cytoplasmic
domain as bait yielded a clone which coded for ERK2 at the 3' end with 100%
nucleotide sequence identity (across 393 bases) to the published sequence of ERK2
(Boulton et al, 1991). This putative association was investigated further.
Integrin immunoprecipitations were performed on equal protein loads of tumour
cell lysates and the transferred proteins blotted with antibodies recognising
phosphorylated and non-phosphorylated forms of ERK1/2. The specificity of this
interaction was examined in integrin immunoprecipitations against βl, β5, βό and αv
integrin subunits. As shown in Fig. 10, the anti- ERK mAb SC-1647 identified a β6-
specific band migrating at the position of purified ERK2 protein. The integrin-
associated ERK band was identified only in immunoprecipitations of the βό subunit and
its partner αv and not in immunoprecipitations of the βl/β5 subunits or in
immunoprecipitations using isotype-matched control antibodies. 2.3.4 Effect of altered βό expression on total cellular ERK and βό-bound ERK.
Equal protein loads from one representative clone each from WiDr mock and
antisense β6 transfectants were electrophoresed, transferred to nitrocellulose and blotted
with anti-ERK mAb (SC-1647) as shown in Fig.11(A). β6 immuno-precipitates from
equal protein loads of the WiDr mock and antisense βό clones were electrophoresed,
transferred and probed with anti-ERK mAb (ElO) as shown in Fig. 11(B). As indicated,
suppression of βό expression resulted in a reduction of both total cellular and
phosphorylated integrin-associated ERK compared with WiDr mock transfectants.
Similarly, βό immunoprecipitations from SW480 βό transfected clones expressing high
and low levels of βό (confirmed by FACScan and βό immunoprecipitations) showed
parallel changes in βό-bound phosphorylated ERK (Fig. 12).
2.3.5 Effect of βό immunodepletion on total cellular ERK and β6-bound ERK
WiDr wild-type cells were surface biotinylated and the cell lysates
immunodepleted of β6 in three rounds of sequential immunoprecipitation using anti-βό
mAb (R6G9) resulting in a marked loss of β6 from the lysates as shown in Fig. 13(A).
βό-immunodepleted lysates were then transferred and blotted with anti-ERKl/2 antibody
(SC-1647), which recognises both phosphorylated and non-phosphorylated forms of
ERK1/2. As shown in Fig. 13(B), following three rounds of βό immunodepletion, levels
of ERK1/2 in βό-depleted lysates compared with non-immunodepleted lysates, were
markedly reduced suggesting a significant contribution of β6-bound ERK to total
cellular ERK. In contrast, immunodepletion of β5 by three successive rounds of
immunoprecipitation with mAb P1F6 or isotype-matched control antibody IgG2A did not result in any reduction of cellular ERK levels compared with non-immunodepleted
cell lysates.
2.3.6 Effect of ERK immunodepletion on β6-bound ERK
Cell lysates from WiDr wild-type cells were immunodepleted of ERK1/2 by means
of sequential immunoprecipitations using the anti-ERK mAb, SC-1647. ERK-
immunodepleted cell lysates probed with either ElO or SC-1647 mAbs contained
markedly less ERKs. To examine the effect of ERK-immunodepletion on β6-bound
ERK, the ERK-immunodepleted cell lysates were immunoprecipitated with anti-βό mAb
(R6G9) and the βό-immunoprecipitates probed with anti-ERK mAb (ElO recognising
phosphorylated ERK1/2). ERK immunodepletion effectively reduced levels of βό-
bound ERK.
2.3.7 Effect of preventing cell attachment on αvβό-bound ERK
SW480 mock- and β6 expressing transfected cells were grown to 80-90%
confluency. Cells were washed twice with PBS and harvested after trypsinization. Cells
were divided into three equal portions (approximately 4 x 10 /batch). The first group
was plated on a 75 cm plastic flask in normal 10% serum containing medium (attached
cells) while the other two groups were plated on 0.3% agarose underlay in serum free
medium (non-attached cells). Cells were allowed to grow for 24 hours at 37°C, after
which in one group of 0.3% agarose underlay, 15 ml of serum-free medium was added
while in the other an equal volume of 10% serum-containing medium was added for 30
mins. Cells were collected washed with PBS and lysed in cell lysis buffer (100 mM
Tris-HCl pH-7.5, 150 mM NaCl, lmM Cacl2, 1% Triton, 0.1% SDS, 0.1% Np-40, 1
mM vanadate, 1 μg/ml pepstatin, 1 mM PMSF, 5 μg/ml aprotonin and lμg/ml of leupeptin). 10 μl (10 μg of protein) was used for the analyses of cell lysates after adding
equal volumes of non-reducing Laemmli buffer. For SW480-β6-transfected cells the
rest of the cell lysate was used for immunoprecipitation of αvβό integrin. Cell lysates
and βό immunoprecipitates were subjected to western blotting and probed with ElO
monoclonal antibody (recognising phosphorylated ERK 1/2).
Cell lysates from the non-attached cells were found to require serum factors to
maintain phosphorylation of total cellular ERK. In contrast, non-attached SW480 βό
colon cancer cells do not require serum factors to maintain the activation
(phosphorylation) state of βό-bound ERK.
2.3.8 Effect of PP2A phosphatase on αvβό-bound ERK
In experiments to examine the effect of protein phosphatase 2 A (PP2A) on βό-
bound ERK, SW480 β6-transfected cells were sonicated in buffer comprising 50mM
Tric-HCl (pH 8.0), lOmM MgCl2 and O.OlmM EGTA together with enzyme inhibitors,
and cell lysates from both serum-starved and serum-induced cells treated with 0.5 units
of PP2A (Promega) at 30°C for 10 minutes. The reaction mixture was stopped by
addition of equal volumes of non-reducing Laemmli sample buffer. In parallel, PP2A-
treated cell lysates were immunoprecipitated with mAb R6G9 (anti-β6) followed by
Western blot with anti-ERK mAb El O. MAP kinase activity assays were performed
32 according to the manufacturer's instructions (Amersham Pharmacia Biotec) using γ P-
ATP.
Exposure of cell lysates prepared from serum-supplemented and serum-starved
cells to the PP2A catalytic subunit resulted in dephosphorylation of total ERK. In
contrast, dephosphorylation of βό-bound ERK was not observed in βό immunoprecipitates prepared from serum-supplemented or serum-starved cells βό
Bound ERK may therefore serve to maintain adjacent growth factor receptors in an
activated state and thereby alter their sensitivity to exogenous co-factors.
2.3.9 Inhibition of MAP kinase activity inhibits secretion of gelatinase B.
SW480 βό transfectants were cultured under serum-free conditions for 48 hours in
the absence/presence of the MEK inhibitor PD98059 (40μM) or DMSO (vehicle
control). Tumour-conditioned medium was assayed for gelatinase B by analysis of equal
protein loads in a gelatin zymogram.
Inhibition of MAP kinase activity by the MEK inhibitor reduced gelatinase B
secretion compared with controls.
2.3.10 βό-ERK2 association in HaCaT and HaRas Cell Lines
β6 immunoprecipitates were prepared from human kerotinocyte cell lines (HaCaT
and HaRas were obtained from Prof. N. Fusenig, The German Cancer Research Institute,
Heidelberg, Germany) using mAb R6G9 (anti-βό) and the immunoprecipitates probed
with mab ElO (against phosphorylated ERK 1/2). Fig. 14 shows that ERK2 associates
with βό in both of HaCat and HaRas cells.
2.4 Discussion of Results
The MAP kinase pathway has been shown to be important in experimental tumour
metastases (Mansour et al, 1994) and recent data implicate MAP kinases in tumour
growth and invasiveness of colon cancer cells (Sebolt-Leopold et al, 1999). In the
present study, up-/down-regulation of βό expression in various colon cancer cell lines
was shown to enhance/suppress respectively, MAP kinase activity. The presence of
serum induced a three-fold increase in MAP kinase activity above that observed for serum-starved β6-expressing cells. In contrast, only a one-fold increase in serum-
dependent MAP kinase activity was observed for cells in which βό had been down-
regulated consequent upon transfection with antisense βό. Moreover, induced
expression of β6 in the non-β6-expressing colon cancer cell line SW480, was associated
with a three-fold increase in serum-dependent MAP kinase activity. Serum contains a
mixture of growth factors raising the possibility that one role for αvβό in colon cancer
cells is to lower the threshold for activation of MAP kinase signalling pathways at times
when the supply of serum-containing growth factors is limited.
The ERK band co-immunoprecipitated with βό migrated either at or 1 - 2 kD
higher than the mobility of purified phosphorylated ERK2 depending on the acrylamide
concentration used in SDS-PAGE showing that the kinase does indeed, associate with
the βό subunit. Slight differences in mobility of the βό-associated ERK band compared
with the pure ERK protein could arise if β6-bound ERK is hyper-phosphorylated and/or
exists in an altered conformation consequent upon its association with βό. ERK bound
to βό may also be an alternatively spliced variant of ERK2 causing it to migrate
differently. Finally^ the purified ERK2 protein is derived from mouse which differs
slightly from human ERK2 by being two amino acid residues shorter and also containing
a single amino acid substitution.
In the present study, increased/decreased β6 expression in colon cancer cell lines
was associated with increased/decreased β6-bound ERK, respectively. In addition,
immunodepletion experiments suggest that βό-bound ERK makes a substantial
contribution to total phosphorylated ERK within the cell and overall MAP kinase
activity (see Figs. 15(A) and 15(B)). The observation that β6-mediated colon cancer growth in vitro is inhibitable by a
matrix metalloproteinase inhibitor (Agrez et al, 1999) suggests that the increased
gelatinase B secretion by βό-expressing colon cancer cells contributes to tumour
progression. Taken together with the finding that inhibition of MAP kinase activity by
the MEK inhibitor PD98059 diminished gelatinase B secretion in βό-expressing cells, it
seems that activation of MAP kinase signalling plays a role, at least in part, in β6-
mediated induction of gelatinase B secretion.
Example 3
3.1 Identification of the binding domain on the βό subunit cytoplasmic tail domain
for ERK2
Peptide fragments corresponding to regions of the cytoplasmic tail domain of the
βό subunit were screened in an enzyme-linked immunosorbent assay (ELISA) for
binding with ERK2. The 52 amino acid long β6 cytoplasmic tail is shown in Fig. 16 as
are the amino acid sequences for the cytoplasmic domains of the βl to β3 subunits. In
particular, four synthetic peptides designated fragment 1 to fragment 4 were prepared
and biotinylated at the N-terminal end of each, respectively (Auspep Pty Ltd, Melbourne
Australia).
The region of the β6 tail to which each corresponds is indicated in Fig. 16 and set
out below.
Fragment 1 : HDRKEVAKFEAERSKAKWQTGT
Fragment 2: RSKAKWQTGTNPLYRGSTST
Fragment 3: NPLYRGSTSTFKNVTYKHRE Fragment 4: FKNVTYKHREKQKVDLSTDS
The fragments overlap by 10 amino acids and are each 20 amino acids long with
the exception of the fragment 1 with a length of 22 amino acids. Fragment 4 was
synthesised with a terminal serine rather than a cysteine as found in wild-type βό to
avoid formation of a disulfide bridge between peptides.
Overlapping biotinylated fragments 1 to 4 were coated onto streptavidin coated
polystyrene plates (Pierce, Rockford IL USA, Cat No. 15125) and the ELISA performed
substantially according to manufacturers instructions. Briefly, wells are washed with 3 x
200μl of wash buffer (TBS, 0.1% BSA, 0.05% Tween 20 or SuperBlock™ blocking
buffer in TBS, Pierce, Prod. No. 37535) prior to addition of biotinylated peptides
(lOOμl) and incubation for 1 hr at room temperature to allow for capture of the peptides
on the plate. Following another washing step, peptides were overlayed with GST-ERK,
ERK (or JNK-1) alone at a volume of lOOμl per well, and the plates incubated for a
further 1 hr at room temperature before removal of any unbound ERK by further
washing.
Binding of ERK to the peptides is detected using lOOμl anti-ERKl/2 mAb SCI 647
(Santa Cruz) as the primary antibody at a dilution of 1 :700 (isotype matched antibody
IgG2b is used as a control). This is followed by another washing step and addition of
lOOμl rabbit anti-mouse antibody (Biorad) conjugated to alkaline phosphatase at a
concentration of 1 : 1000 for 30 minutes again at room temperature. A lOOμl aliquot of
detection reagent (alkaline phosphatase detection kit-Biorad) is then introduced into each
well after a final washing step and allowed to react for 15-30 minutes at room
temperature in the dark before absorbance is measured at 405nm. All dilutions of peptides, MAP kinase and antibodies were performed using the wash buffer. GST-ERK
is a fusion protein consisting of ERK coupled to glutathione-S-transferase and purified
from host cells transfected with pGEX vector.
As shown in Fig. 17, significant binding of non-phosphorylated GST.ERK2
(0.25μg/100μl) to peptide fragment 2 was observed (lμg/lOOμl) while only negligible or
low level binding for the other fragments was found.
Significant binding of non-phosphorylated ERK2 to both fragment 2 and βό
cytoplasmic tail peptide compared to fragments 1, 3 and 4 over a range of concentrations
of ERK2 was also observed (see Fig.18). Similar results were observed using a range of
concentrations of the peptide fragments as shown in Fig. 19.
To further localise the binding domain on the cytoplasmic tail of the βό subunit,
progressively shorter peptides from the region of the βό cytoplasmic tail corresponding
to peptide fragment 2 were synthesised, biotinylated and the capacity to associate or
otherwise bind to ERK2 assessed as described above. The binding of GST.ERK2 to a 15
mer test peptide (seq. 4) having the amino acid sequence RSKAKWQTGTNPLYR and a
10 mer test peptide having the sequence RSKAKWQTGT is shown in Fig. 20 compared
to fragment 2 over a range of concentrations of the peptides. As can be seen, no
reduction in binding to the seq. 4 peptide compared to fragment 2 was found. Binding of
ERK2 to the seq. 3 peptide was substantially less than that observed for seq. 4.
A number of 10 mer biotinylated peptides corresponding to regions of fragment 2
or fragment 3 were then tested. The amino acid sequence for each peptide is as follows
and their location in the βό cytoplasmic domain is indicated in Fig. 21.
10(1): NPLYRGSTST 10(2): WQTGTNPLYR
10(3): KFEAERSKAK
The results are set out in Fig. 22 and show that GST. ERK binding to the 10 mer
peptides is substantially reduced compared to binding to the seq. 4 peptide suggesting
that opposite end regions of seq. 4 participate in the binding of ERK2.
Comparable binding of ERK2 to seq. 4 was found using a further 10 mer peptide
identified as 10(4) in which amino acid sequence WQTGT of seq. 4 is omitted indicating
that WQTGT is a linker sequence that does not participate directly in the binding of
ERK to seq. 4. Negligible binding of ERK2 to the 5 mer peptide RSKAK was observed
as shown in Fig. 23. ERK2 cleaved from GST-ERK2 by thrombin was used in this
assay. Results (not shown) indicate that greater than a 3 fold increase in assay sensitivity
can be achieved using thrombin cleaved ERK2 rather than GST-ERK2.
Example 4
4.1 MAP kinase JNK- 1 binds to the cytoplasmic tail domain of β6
In view of the observation that ERK2 associates with the cytoplasmic tail of the βό
subunit, the MAP kinase JNK-1 was tested to evaluate whether it also could associate
with the cytoplasmic tail of βό.
Briefly, 0.05-1.5μm/100μl of JNK-1 (Santa Cruz) was aliquoted into wells of a 96
well culture place containing increasing concentrations of the β6 cytoplasmic domain
tail peptide used in Example 3. For comparison purposes, non-phosphorylated GST-
ERK2 (0.05-1.5μm/100μl) was aliquoted into wells containing βό cytoplasmic tail
peptide or peptide fragments 1 or 2, respectively. Binding of JNK-1 was detected using mouse anti- JNK-1 mAb SC 474-G (Santa Cruz) and HRP-conjugated goat anti-mouse
antibody. Absorbance was read at 405nm and the results are shown in Fig. 24.
Significant binding of JNK-1 to the βό cytoplasmic tail peptide was found.
Example 5
5.1 Evaluation of ability of ERK2 to bind to βό Δ746-764 deletion mutant.
To examine the role of the amino acid sequence RSKAKWQTGTNPLYR in the
β6 cytoplasmic domain in situ, a β6 deletion construct lacking the coding sequence for
AERSKAKWOTGTNPLYRG was transfected into colon cancer cell line SW480 which
does not constitutively express the αVβό integrin using the calcium phosphate method
previously described for transfections into this cell line (Agrez et al, 1994). The location
of the βό Δ746-764 deletion is indicated in Fig. 25. Construction of the β6 Δ746-764
deletion mutant in the vector pcDNAlneo and failure of the expressed receptor to
localise to focal adhesions in Chinese hamster ovary cells has been reported (Cone et al,
1994). Facscan analysis revealed comparable levels of surface expression of mutant βό
to that seen for the full length wild-type receptor (see Fig. 26).
Equal protein loads of cell lysates prepared from SW480 cells were
immunoprecipitated with either anti-β6 monoclonal antibody (mAb R6G9) or matched
isotype control antibody. Surface biotinylation prior to immunoprecipitation confirmed
equal surface expression of mutant and wild-type β6 (see Fig. 27 (A). Aliquots of the
immunoprecipitates were electrophoresed and transferred to nitrocellulose for Western
blotting using monoclonal antibody ElO which recognises ERK1/2. As seen in Fig.
27(B), loss of the RSKAKWQTGTNPLYR sequence in the βό cytoplasmic domain reduced levels of βό-bound ERK by greater than approximately 75% of that observed for
the wild type receptor.
Example 6
6.1 Growth inhibition study.
HT29 and SW480 βό-expressing colon cancer cell lines were seeded into wells of
96-well microtitre plates (Nunclon) in Dulbecco's Modified Eagle's Medium (DMEM)
supplemented with 10% foetal bovine serum, glutamine, Hepes, and antibiotics. Seeding
cell densities were 3 x 10 cells per triplicate well for each condition tested and after 24
hours incubation of cell cultures in 5% CO2, 100% humidity at 37°C, the culture
medium was exchanged for serum-free DMEM medium supplemented with insulin,
transferrin, selenous acid, hydrocortisone, non-essential amino acids, glutamine, Hepes
and antibiotics containing either peptide RSKAKWQTGTNPLYR alone or penetratin-
peptide complex at a concentration of lOμm for HT29 cells or 30μm for SW480 βό-
expressing cells. Cell cultures were incubated for a further 24 hours following which
cultures were photographed (Kodak Techpan Film at 100 ASA setting) and the
experiments terminated by addition of the cell proliferation reagent WST-1 (Boehringer
Mannheim) to monitor effects of the peptide on cell growth. The cell proliferation
reagent WST-1 is designed to be used for the non-radioactive, spectrophotometric
quantification of cell growth and viability in proliferation and chemosensitivity assays.
The colourmetric assay is based on the cleavage of the tetrazolium salt WST-1 by
mitochondrial dehydrogenase in viable cells. In particular, at the termination of experiments, 30μl of WST-1 was added to
270μl culture medium volume in each microtitre well and the colour change quantitated
in an ELISA plate reader by measuring absorbance of the formazan product at 450mn
(using a reference wavelength of more than 600nm). The mean absorbance readings
from triplicate wells (± standard error of the means) after subtraction of background
control wells (culture medium without cells)was determined. Only the carrier
penetratin-peptide complex was effective in inhibiting cell proliferation in contrast to
either peptide or penetratin alone as shown in Figs. 28 and 29 indicating that the
penetratin-peptide complex was internalised by both the HT29 and SW480 cells
resulting in the observed suppression of colon cancer growth. Photographs of the
SW480 cells treated with penetratin alone or the penetratin-peptide complex are shown
in Fig. 30 (A) to (C).
Example 7
SW480 mock and S W480 βό transfectants were cultured in DMEM medium
supplemented with 1% foetal bovine serum in the presence of 20 μM seq. 4 coupled to
penetratin. Percentage inhibition was assessed by the WST-1 colorimetric
dehydrogenase assay described in Example 6. The percentage inhibition of growth
observed for the -βό and +βό expressing cells was 17% and 50%, respectively as
indicated in Fig 31 A. The -β6 and +β cultured cells are shown in Fig 3 IB. Example 8
The proliferation of SW480 cells expressing a β6 Δ746-764 deletion mutant was
compared with non-β6 expressing SW480 cells and SW480 cells expressing full length
wild-type βό. Cells were cultured for 10 days within a 3-dimensional collagen type I
matrix. Collagen gels were prepared as bilayers (upper layer cell-containing and lower
layer minus cells) in 24 well culture plates as previously described (Agrez, 1989; Agrez,
1994) except for the use of 5% foetal bovine serum as the supplement for DMEM.
Colonies were photographed in the gel Fig.32(A) at 10 days (bar represents 200μ) and
following dissolution of the collagen with collagenase Fig.32(B) prior to visual colony
counting of all colonies exceeding 200μ in diameter within each well Fig.32(C). As can
be seen, significant proliferation of the SW480 cells expressing the full length wild-type
β6 was observed compared to the non-βό expressing cells and cells expressing the β6
Δ746-764 deletion mutant.
Example 9
Growth inhibition of S W480 cells expressing full length wild-type βό exposed to
seq.4 coupled to penetratin or RSKAKWQTGTNPLYR peptide coupled to penetratin (5,
10, 20, 30 μM in DMEM minus foetal bovine serum) but which peptide contained
alanine substitutions at the four positions indicated was assessed. As shown in
Fig.33(A) and Fig 33(B), progressive inhibition of proliferation in a dose-response
manner was observed for the seq.4 penetratin complex compared with the alanine
substituted peptide-penetratin complex which was without effect at all doses tested. Example 10
Binding of ERK2 to the seq.4 peptide (RSKAKWQTGTNPLYR) was compared
with peptides corresponding to regions of the cytoplasmic domain of integrin subunits
βl, β2, β3 and β5. The amino acid sequences for those peptides is shown below:
βl KFEKEKMNAKWDTGENPIYK
β2 KEKLKSQWNNDNPLFK
β3 RARAKWDTANNPLYK
β5 RSRARYEMASNPLYR
As shown in Fig.34, significant binding of ERK2 to the seq. 4 peptide was observed.
Binding of ERK2 to the β5 and β3 peptides was also found. The results have been
corrected for non-specific binding and indicate a hierarchy of binding of ERK2 to
integrin subunits.
Although the present invention has been described hereinbefore with reference to a
number of preferred embodiments, the skilled addressee will understand that numerous
variations and modifications are possible without departing from the scope of the
invention.
References Cited:
Agrez, M.V. and Bates R.C., Colorectal cancer and the integrin family of cell
adhesion receptors: current status and future directions. European Journal of Cancer
30A, 2166-2170 (1994).
Agrez, M.V., Human colon cancer and fibroblast cell lines cultured in and on
collagen gels. Aust. N. Z. J. Surg. 59, 415-420 (1989).
Agrez, M.V., Bates R.C., Mitchell, D., Wilson, N., Ferguson, N., Anseline, P.
and Sheppard. D., Multiplicity of fibronectin-binding αv integrin receptors in
colorectal cancer. Br. J. Cancer 73, 887-892 (1996).
Agrez, M.V., Cell adhesion molecules and colon cancer. A.N.Z.J. Surgery. 66,
789-796 (1996).
Agrez, M.V., Chen, A., Cone, R.I., Pytela, R. and Sheppard, D., The
αvβόintegrin promotes proliferation of colon carcinoma cells through a unique region of
the βό cytoplasmic domain. J. Cell Biol. 127, 547-556 (1994).
Agrez, M.V.? Gu, X., Turton, J., Meldrum, C, Niu, J., Antalis, T. and
Howard, E.W., The αvβό integrin induces gelatinase B secretion in colon cancer cells.
Int. J. Cancer 81, 90-97 (1999).
Bachmann, A.S., Surovoy, A., Jung, G. and Moelling, K., Integrin receptor-
targeted transfer peptides for efficient delivery of antisense oligodeoxynucleotides. J.
Mol. Med. 76, 126-132 (1998).
Berkner, Curr. Top. Microbiol. Immunology 158, 39-61 (1992).
Bookstein, R., Demers, W., Gregory, R., et al, p53 gene therapy in vivo for
hepatocellular and liver metastatic colorectal cancer. Seminars Oncol. 23, 66-77 (1996). Boudreau, N., Sympson, C.J., Werb, Z. and Bissell, M., Suppression of ICE and
apoptosis in mammary epithelial cells by extracellular matrix. Science 267, 891-893
(1995).
Boulton, T.G., Nye, S.H. and Robbins, D. J., ERKs: a family of protein-
serine/threonine kinases that are activated and tyrosine phosphorylated in response to
insulin and NGF. Cell 65, 663-675 (1991).
Breakfield and Geller, Mol. Neurobiol. 1, 337-371 (1987).
Breuss, J.M., Gallo, J., De Lisser, H.M., Klimanskaya, IN., Folkesson, H.G.,
Pittet, J.F., Νishimura, S.L., Aldape, K., Landers, D.V., Carrenter, W., Gillet, Ν.,
Sheppard, D., Mathay, M., Albeda, S.M., Kramer, R.H., and Pytela, R., Expression
of the β6 integrin in development, neoplasia and tissue repair suggests a role in epithelial
remodelling. J. Cell Sci. 108, 2241-2251 (1995).
Breuss, J.M., Gillet, Ν., Lu, L., Sheppard, D. and Pytela, R., Restricted
distribution of integrin beta 6 mRΝA in primate epithelial tissues. J. Histochem.
Cytochem. 41, 1521-1527 (1993).
Brooks, P.C., Montgomery, A.M.P., Rosenfeld, M., Reisfeld, R.A., Hu, T.,
Klier, G. and Cheresh, D.A., Integrin alpha v beta 3 antagonists promote tumor
regression by inducing apoptosis of angiogenic blood vessels. Cell 79, 1157-1164
(1994).
Brooks, P.C., Stromblad, S., sanders, L.C., von Schalscha, T.L., Aimes, R.T.,
Stetler-stevenson, W.G., Quigley, J.P. and Cheresh, D.A., Localization of matrix
metal loproteinase MMP-2 to the surface of invasive cells by interaction with integrin
αvβ3. Cell 85, 683-693 (1996). Busk, M., Pytela, R. and Sheppard, D., Characterisation of the αvβό as a
fibronectin-binding protein. J. Biol. Chem. 267, 5790-5796 (1992).
Chen, Q., Kinch, M.S. and Lin, T.H., Integrin-mediated cell adhesion activates
mitogen-activated protein kinases. J. Biol. Chem. 269, 26602-26605 (1994).
Chomczynski, P. and Sacchi, N., Single-step method of RNA isolation by acid
guanidium thiocyanate-phenol-chloroform extraction. Anal. Biochem. 162, 156-159
(1987).
Clark, R.A., Ashcroft, G.S., Spencer, M.J., Larjava, H. and Ferguson, M.W.,
Re-epithelialization of normal human excisional wounds is associated with a switch
from alpha v beta 5 to alpha v beta 6 integrins. Br. J. Dermatol. 135, 46-51 (1996).
Cone, R.I., Weinacker, A., Chen, A. and Sheppard, D., Effects of beta subunit
cytoplasmic domain deletions on the recruitment of the integrin alpha v beta 6 to focal
contacts. Cell Adhes. Comm. 2, 101-113 (1994).
Coppolino, M., Leung-Hagesteijn, C. Dedhar, S. and Wilkins, J., Inducible
interaction of integrin alpha 2 beta 1 with calreticulin. Dependence of the activation state
of the integrin. J. Biol. Chem. 270, 23132-23138 (1995).
Dedhar, S. and Hannigan, G.E., Integrin cytoplasmic interactions and
bidirectional transmembrane signalling. Curr. Opin. Cell Biol. 8, 657-669 1996).
Dedhar et al., J. Cell. Biol. 104, 585 (1997).
Derossi, D., Calvet, S., Trembleau, A., et al, Cell internalization of the third
helix of the Antennapedia homeo domain is receptor-independent. J. Biol. Chem. 271,
18188-18193 (1996). Derossi, D., Joliot, A.H., Chassaing, G. and Prochiantz, A., The third helix of
the Antennapedia homeodomain translocates through biological membranes. J. Biol.
Chem. 269, 10444-10450 (1994).
Felding-Habermann, B., Mueller, B.M., Romerdahl, CA. and Cheresh, D.,
Involvement of integrin av gene expression in human melanoma tumorigenicity. J. Clin.
Invest. 89, 2018-2022 (1992).
Fink et al., Hum. Gene Ther. 3, 11-19 (1992).
Freese et al., Biochem. Pharmacol. 40, 2189-2199 (1990).
Gamble, J.R., Matthias, L.J., Meyer, G., Kaur, P., Russ, G., Faull, R., Berndt,
M.C. and Vadas, M.A., Regulation of in vitro capillary tube formation by anti-integrin
antibodies. J. Cell Biol. 121, 931-43 (1993).
Garrington, T.P., and Johnson, G.L., Organization and regulation of mitogen-
activated protein kinase signaling pathways. Curr. Opin. Cell Biol. 11, 211-218 (1999).
Giancotti, F.G. and Ruoslahti, E., Integrin signalling. Science 285, 1028-1032
(1999).
Gorgziglia and Kapikian, J. Virol. 66, 4407-4412 (1992).
Grammer, T.C. and Blenis, J., Evidence for MEK-independent pathways
regulating the prolonged activation of the ERK-MAP kinases. Oncogene 14, 1635-1642
(1997). Guan, J.L. and Shalloway, D., Regulation of focal adhesion-associated protein
tyrosine kinase by both cellular adhesion and oncogenic transformation. Nature 358,
690-692 (1992). Haapasalmi, K., Zhang, K., Tonneson, M., Olerud, J., Sheppard, D., Salo, T.,
Kramer, R., Clark, R.A.F., Uitlo, V-J. and Larjava, H., Keratinocytes in human
wounds express avbό integrin. J. Invest. Dermatol. 106, 42-48 (1996).
Hannigan, G.E., Leung-Hagesteijn, C, Fitz-Gibbon, L., Coppolino, M.G.,
Radeva, G., Filmus, J., Bell, J.C. and Dedhar, S., Regulation of cell adhesion and
anchorage-dependent growth by a new beta 1 -integrin-linked protein kinase. Nature
379, 91-96 (1996).
He, T-C, Zhou, S., Da Costa, L.T., Yu, J., Kinzler, K.W. and Vogelstein, B. A
simplified system for generating recombinant adenoviruses. Proc, Natl. Acad. Sci. USA
95, 2509-2514 (1998).
Hemler, M.E., Integrin associated proteins. Curr. Opin. Cell Biol. 10, 578-585
(1998).
Hergott, G.J., Nagai, H. and Kalnins, V.I., Inhibition of retinal pigment
epithelial cell migration and proliferation with monoclonal antibodies against the beta 1
integrin subunit during wound healing in organ culture. Invest. Ophthalmol. Vis. Sci. 34,
2761-2768.(1993).
Horwitz, A., Duggan, K., Buck, C, Beckerle, M.C. and Burridge, K.,
Interaction of plasma membrane fibronectin receptor with talin-a transmembrane
linkage. Nature 320, 531 -533 (1986). Howe, A., Aplin, A.E., Alahari, S.K. and Juliano, R.L., Integrin signaling and
cell growth control. Curr. Opin. Cell Biol. 10, 220-231 (1998)
Hynes, R. O., Integrins: versatility, modulation and signaling in cell adhesion.
Cell 69, 11-25 (1992). Johnson et al., J. Virol. 66, 2952-2965 (1992).
Jones, J., Watt, F.M. and Speight, P.M., Changes in the expression of alpha v
integrins in oral squamous cell carcinomas. J. Oral Path. & Med. 26, 63-68 (1997).
Klemke, R,L., Cai, S. Giannini, A.L., Gallagher, P.J., de Lanerolle, P. and
Cheresh, D.A., Regulation of cell motility by mitogen-activated protein kinase. J. Cell
Biol. 137, 481-492 (1997).
Knezevic, I., Leisner, T.M. and Lam, SC-T., Direct binding of the platelet
integrin alpha lib beta 3 (GPIIb-IIIa) to talin. Evidence that interaction is mediated
through the cytoplasmic domains of both alpha lib and beta 3. J. Biol. Chem. 271,
16416-16421 (1996).
Kohler G and Milstein C. Continuous cultures of fused cells secreting antibody
of predefined specificity. Nature 256, 495-497 (1975).
Kornberg, L., Earp, H.S., Parsons, J.T., Schaller, M. and Juliano, R.L., Cell
adhesion or integrin clustering increases phosphorylation of a focal adhesion-associated
tyrosine kinase. J. Biol. Chem. 267, 23439-23442 (1992).
Kraft, S., Dienfenbach, B., Mehta, R., Jonczyk, A., Luckenbach, G.A. and
Goodman, S.L., Definition of an unexpected ligand recognition motif for αVβό
integrin. J. Biol. Chem. 274 (4) 1979-1985 (1999).
Lengauer, C, Kinzler, K.W. and Vogelstein, B., Genetic instability in colorectal
cancers. Nature 386, 623-7 (1997).
Licato, L.L., Keku, T.O. and Wurzelmann, J.I., In vivo activation of mitogen-
activated protein kinases in rat intestinal neoplasia. Gastroenterology 113, 1589-1598
(1997). Lindberg, F.P., Gresham, H.D., Reinhold, M.I. and Brown, E.J., Integrin-
associated protein immunoglobulin domain is necessary for efficient vitronectin bead
binding . J. Cell Biol. 134, 1313-1322 (1996).
Liu, S., Thomas, S.M., Woodside, D.G., Rose, D.M., Klosses, W.B., Pfaff, M.
and Ginsberg, M.H., Binding of paxillin to alpha 4 integrins modifies integrin-
dependent biological responses. Nature 402, 676-680 (1999).
Lo C.W., Transformation by iontophoretic microinjection of DNA: Multiple
integrations without tandem insertions. Mol. Cell. Biol. 3,1803 (1983).
Loftus, J.C, Smith, J.W. and Ginsberg, M.H., Integrin-mediated cell adhesion:
the extracellular face. J. Biol. Chem. 269, 25235-25238 (1994).
Madzak et al., J.Gen. Virol. 73, 1533-1536 (1992).
Mainiero, F. Murgia, C, Wary, K.K., Curatoia, A.M., Pepe, A., Blumemberg,
M., Westwick, J.K., Der, C.J. and Giancotti, F.G., The coupling of alpha 6 beta 4
integrin to Ras-MAP kinase pathways mediated by She controls keratinocyte
proliferation. EMBO J. 16, 2365-2375 (1997).
Maniero, F., Pepe, A., Wary, K.K., Spinardi, L., Mohammadi, M.,
Schlessinger, J. and Giancotti F.G., Signal transduction by the alpha 6 beta 4 integrin:
distinct beta 4 subunits mediate recruitment of Shc/Grb2 and association with the
cytoskeleton of hemidesmosomes. EMBO J 14, 4470-4481 (1995).
Mansour, S.J., Matten, W.T., Hermann, A.S., Candia, J.M., Rong, S.,
Fukasawa, K., Vande-Woude, G.F. and Ahn, N.G., Transformation of mammalian
cells by constitutively active MAP kinase kinase. Science 265, 966-670 (1994).
Margolskee, Curr. Top. Microbiol. Immunol. 158, 67-90 (1992). Meredith, J.E. Jr, Fazeli, B. and Schwartz, M.A., The extracellular matrix as a
cell survival factor. Mol. Biol. Cell 4, 953-961 (1993).
Miller, Current. Top. Microbiol. Immunol. 158, 1-24 (1992).
Miranti, C.K., Ohno, S. and Brugge, J.S., Protein Kinase C regulates integrin-
induced activation of the extracellular regulated kinase pathway upstream of She. J.
Biol. Chem. 274, 10571-10581 (1999).
Montgomery, A.M.P., Reisfeld, R.A. and Cheresh, D.A., Integrin αvβ3 rescues
melanoma cells form apoptosis in three-dimensional dermal collagen. Proc. Natl. Acad.
Sci. USA 91, 8856-8860 (1994).
Moss, Curr. Top. Microbiol. Immunol. 158, 25-38 (1992).
Munger, J.S., Huang, X., Kawakatsu, H., Griffiths, M.J.D., Dalton, S.L., Wu,
J., Pittet, J.F., Kaminski, N., Garat, C, Matthoy, M.A., Rifkin, D.B. and Sheppard,
D., The integrin avbό binds and activates latent TGFbl : a mechanism for regulating
pulmonary inflammation and fibrosis. Cell 96, 319-328 (1999).
Muzyczka, Curr. Top. Microbiol. Immunol. 158, 97-123 (1992).
Otey, C.A., Pavalko, F.M. and Burridge, K., An interaction between alpha-
actinin and the beta 1 integrin subunit in vitro. J. Cell Biol. 111,721-729 (1990).
Payne, D.M., Rossomando, A.J., Martino, P., et al, Identification of the
regulatory phosphorylation sites in pp42/mitogen-activated protein kinase (MAP kinase).
EMBOJ 10, 885-892 (1991).
Petropoulos et al., J. Virol. 66, 3391-3397 (1992).
Pfaff, M., Liu, S., Erie, D.J., Ginsberg, M.H., Integrin beta cytoplasmic domains
differentially bind to cytoskeletal proteins. J. Biol. Chem. 273, 6104-6109 (1998). Reszka, A.A., Hayashi, Y. and Horwitz, A.F., Identification of amino acid
sequences in the integrin beta 1 cytoplasmic domain implicated in cytoskeletal association. J. Cell Biol. 1 17, 1321-1330 (1992).
Rojiani, M.V., Finlay, B.B., Gray, V. and Dedhar, S., In vitro interaction of a
polypeptide homologous to human Ro/SS-A antigen (calreticulin) with a highly
conserved amino acid sequence in the cytoplasmic domain of integrin alpha subunits.
Biochemistry 30, 9859-9866 (1991).
Schaller, M.D., Otey, C.A., Hilderbrand, J.D. and parsons, J.T., Focal
adhesion kinase and paxillin bind to peptides mimicking beta integrin cytoplasmic
domains. J. Cell Biol. 130, 1181-1187 (1995).
Schiller et al. Int. J. Pept. Prot. Res. 25, 175 (1985).
Schlaepfer, D.D., Hanks, S.K., Hunter, T. and van der Geer, P., Integrin-
mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesion
kinase. Natural 372, 786-791 (1994).
Schneller, M., Vuori, K. and Ruoslahti, E., alpha v beta 3 integrin associates
with activated insulin and PDGF beta receptors and potentiates the biological activity of
PDGF. EMBO J. 16, 5600-5607 (1997).
Sebolt-Leopold, J.S., Dudley, D.T. and Herrera, R., Blockade of the MAP
kinase pathway suppresses growth of colon tumors in vivo. Nature Med. 5, 810-816
(1999).
Sheppard, D., Rozzo, C, Starr, L., Quaranta, V., Erie, D.J. and Pytela, R.,
Complete amino acid sequence of a novel integrin β subunit (β6) identified in epithelial
cells using the polymerase chain reaction. J. Biol. Chem. 265, 11502-11507 (1990). Shimada, J. Clin. Invest. 88, 1043-1047 (1991).
Smythe, W.R., Lebel, E., Bavaria, J.E., Kaiser, L.R. and Albelda, S.M.,
Integrin expression in non-small cell carcinoma of the lung. Cancer & Metastasis
Reviews 14, 229-239 (1995).
Spitz, F.R., Nguyen, D., Skibber, J.M., et al, In vivo adenovirus-mediated p53
tumor suppresor gene therapy for colorectal cancer. Anticancer Res. 16, 3415-3422
(1996).
Stein CA., and Cohen J.S., Oligodeoxynucleotides as inhibitors of gene expression: A review. Cancer Res. 48(10), 2659-2668 (1998).
Takayama, K., Ueno, H., Pei, X.H., et al, The levels of integrin alpha v beta 5
may predict the susceptibility to adenovirus-mediated gene transfer in human lung
cancer cells. Gene Ther. 5, 361.368 (1998).
Takiuchi, H., Kanokogi, M., Fujimoto, N., Hanafusa, T., Kyo, M., Ichikawa,
Y., Nagano, S., Fukunishi, T., Yabumoto, H. and Ihara, H., Expression of integrin
molecule in urological tumour cell lines by using RT-PCR method. Jap. J. Urology 85,
584-588 (1994).
Thomas, G.J., Jones, J. and Speight, P.M., Integrins and oral cancer. Oral
Oncology 33, 381-388 (1997).
Thomas, J.W., Mathias, P., Cheresh, D.A. and Nemeron, G., Integrins alpha v
beta 3 and alpha v beta 5 promote adenovirus internalisation but not virus attachment.
Cell 73, 309-319 (1993). Thompson S., Clark A.R., Pow A.M., Hooper M.L., and Melton D.W., Germ
line transmission and expression of a corrected HPRT gene produced by gene targeting
in embryonic stem cells. Cell 27, 56 (2) 313-321 (1989).
Van der Krol A.R., Mol J.N., and Stuitje A.R., Modulation of eukaryotic gene
expression by complementary RNA or DNA sequences. Biotechniques 6, 958-976
(1998).
Van der Putten H., Botteri F.M., Miller A.D., Rosenfeld M.G., Fan H., Evans
R.M., and Verma I.M., Efficient insertion of genes into the mouse germ line via
retroviral vectors. Proc. Natl. Acad. Sci. USA. 82, 6148 (1988).
Varner, J.A., Emerson, D.A. and Juliano, R.L., Integrin alpha 5 beta 3
expression negatively regulates cell growth: reversal by attachment to fibronectin. Mol.
Biol. Cell o, 725-740 (1995),
Wang, A., Yokosaki, Y., Ferrando, R., Balmes, J. and Sheppard, D.,
Differential regulation of airway epithelial integrins by growth factors. Am. J. Respira.
Cell & Mol. Biol. 15, 664-672 (1996).
Wary, K.K., Mainiero, F., Isakoff, S.J., Marcantonio, E.E. and Giancotti,
F.G., The adaptor protein She couples a class of integrins to the control of cell cycle
progression. Cell 87, 733-743 (1996).
Weinacker, A., Ferrando, R., Elliott, M. Hogg, J., Balmes, J. and Sheppard,
D., Distribution of integrins alpha v beta 6 and alpha 9 beta 1 and their known ligands,
fibronectin and tenascin, in human airways. Am. J. Respira. Cell & Mol. Biol. 12, 547-
556 (1995). Widmann, C, Gibson, S., Jarpe, B and Johnson, G.L., Mitogen-activated
protein kinase: conservation of a three-kinase module from yeast to human. Phys. Rev.
79, 143-180 (1999).
Williams, E.J., Dunican, D.J., Green, P.J., Howell, F.V., Derossi, D., Walsh,
F.S. and Doherty, P., Selective inhibition of growth factor-stimulated mitogenesis by a
cell-permeable Grb2-binding peptide. J. Biol. Chem. 272, 22349-22354 (1997).
Zambruno, G., Marchisio, P.C, Marconi, A., Vaschieri, C, Melchiori, A.,
Giannetti, A. and De Luca, M., Transforming growth factor - βl modulates βl and β5
integrin receptors and induces the de novo expression of the αvβό heterodimer in normal
human keratinocytes: implications for wound healing. J. Cell Biol. 129, 853-865 (1995).
Zhu, X. and Assoian, R.K., Integrin-dependent activation of MAP kinase: a link
to shape-dependent cell proliferation. Mol. Biol. Cell 6, 273-282 (1995).

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS :-
1. An isolated polypeptide capable of binding with a binding site on a MAP kinase
which binding site binds with a binding domain of an integrin for the MAP kinase, or a
homolog, analog, variant or derivative of the polypeptide, with the proviso that the
polypeptide is other than a full length integrin subunit or a βό(770t) or βό(777t) deletion
mutant.
2. A polypeptide according to claim 1 wherein the polypeptide comprises a binding
domain of an integrin subunit for the MAP kinase.
3. A polypeptide according to claim 1 wherein the polypeptide is other than a
fragment of an integrin subunit.
4. A polypeptide according to claim 3 consisting of or incoφorating the amino acid
sequence RSKAKNPLYR.
5. A polypeptide according to claim 1 or 2 wherein the polypeptide comprises one or
both of amino acid sequences RSKAK and NPL YR.
6. A polypeptide according to any one of claims 1 to 5 wherein the polypeptide has a
length of about 150 amino acids or less.
7. A polypeptide according to claim 6 wherein the polypeptide has a length of about
100 amino acids or less.
8. A polypeptide according to claim 7 wherein the polypeptide has a length of about
50 amino acids or less.
9. A polypeptide according to claim 8 wherein the polypeptide has a length of about
40 amino acids or less.
10. A polypeptide according to claim 9 wherein the polypeptide has a length of about
30 amino acids or less.
11. A polypeptide according to claim 10 wherein the polypeptide has a length of about
20 amino acids or less.
12. A polypeptide according to claim 11 wherein the polypeptide has a length of about
15 amino acids or less.
13. A polypeptide according to claim 12 wherein the polypeptide has a length of about
10 amino acids or less.
14. A polypeptide according to any one of claims 1 to 13 or an analog or derivative of
the polypeptide.
15. A polypeptide according to any one of claims 1 to 14 wherein the MAP kinase is
an ERK family member or a JNK family member.
16. A polypeptide according to claim 15 wherein the MAP kinase is ERK2.
17. A fragment of an integrin subunit wherein the fragment is capable of binding with
a MAP kinase, or a homolog, analog variant or derivative of the fragment, with the
proviso that the integrin subunit is other than a βό(770t) or βό(777t) deletion mutant.
18. A fragment according to claim 17 wherein the fragment comprises a binding
domain of the integrin subunit for the MAP kinase.
19. A fragment according to claim 18 wherein the fragment has a length of about 150
amino acids or less.
20. A fragment according to claim 19 wherein the fragment has a length of about 100
amino acids or less.
21. A fragment according to claim 20 wherein the fragment has a length of about 50 amino acids or less.
22. A fragment according to claim 21 wherein the fragment has a length of about 40
amino acids or less.
23. A fragment according to claim 22 wherein the fragment has a length of about 30
amino acids or less.
24. A fragment according to claim 23 wherein the fragment has a length of about 25
amino acids or less.
25. A fragment according to claim 24 wherein the fragment has a length of about 20
amino acids or less.
26. A fragment according to any one of claims 17 to 25 comprising the amino acid
sequence RSKAKWQTGTNPLYRGSTST or a contiguous partial amino acid sequence
thereof.
27. A fragment according to any one of claims 17 to 25 wherein the fragment has a
length of about 15 amino acids or less.
28. A fragment according to claim 27 wherein the fragment comprises amino acid
sequence RSKAKWQTGTNPLYR.
29. A fragment according to any one of claims 17 to 28 wherein the MAP kinase is an
ERK family member or a JNK family member.
30. A fragment according to any one of claims 17 to 29 wherein the MAP kinase is
ERK2.
31. A fragment according to any one of claims 17 to 30 wherein the integrin subunit is
βό.
32. An agent capable of inhibiting binding of a MAP kinase with an integrin.
33. An agent according to claim 32 wherein the agent is capable of binding to a
binding domain on the integrin for the MAP kinase.
34. An agent according to claim 33 wherein the agent is capable of binding with a
binding site on the MAP kinase which binding site binds to a binding domain on the
integrin for the MAP kinase.
35. An agent according to any one of claims 32 to 34 wherein the agent is adapted to
be capable of passing across a cell membrane.
36. An agent according to claim 35 comprising a carrier moiety for facilitating passage
of the agent across the cell membrane and an inhibitor moiety for inhibiting the binding
of the MAP kinase with the integrin.
37. An agent according to claim 36 wherein the agent is adapted for release of the
inhibitor moiety from the carrier moiety when the agent has passed across the cell
membrane.
38. An agent according to claim 32 comprising:
a targeting moiety for targeting cells expressing the integrin;
an inhibitor moiety for inhibiting binding of the MAP kinase with the integrin; and
a carrier moiety for facilitating passage of the inhibitor moiety across the cell
membrane of a said cell;
wherein the inhibitor and carrier moieties are capable of being released from the
targeting moiety at the cell.
39. An agent according to claim 38 wherein the agent comprises an enzyme cleavage
site for being cleaved to thereby release the carrier moiety and the inhibitor moiety at the
cell.
40. An agent according to claim 39 wherein the enzyme cleavage site is a cleavage site
for matrix metalloproteinase-9 (MMP-9).
41. An agent according to any one of claims 38 to 40 wherein the targeting moiety is
an antibody or a binding fragment of an antibody.
42. An agent according to claim 41 wherein the antibody or the binding fragment is
specific for an extracellular region of the integrin.
43. An agent according to any one of claims 38 to 40 wherein the targeting moiety is
an integrin receptor targeted peptide for binding to the integrin.
44. An agent according to any one of claims 38 to 43 wherein the inhibitor moiety is
capable of binding with a binding site on the MAP kinase which binding site binds to a
binding domain on the integrin for the MAP kinase.
45. An agent according to any one of claims 38 to 43 wherein the inhibitor moiety is
capable of binding to a binding domain on the integrin for the MAP kinase.
46. An agent according to any one of claims 36 to 45 wherein the inhibitor moiety
comprises a polypeptide.
47. An agent according to claim 46 wherein the inhibitor moiety is a polypeptide as
defined in any one of claims 1 to 16, or a homolog. analog, variant or derivative of the
polypeptide.
48. An agent according to any one of claims 36 to 45 wherein the inhibitor moiety is a
fragment as defined in any one of claims 17 to 31 or a homolog, analog, variant or
derivative of the fragment.
49. An agent according to any one of claims 36 to 48 wherein the carrier moiety is a
polypeptide.
50. An agent according to claim 49 wherein the carrier moiety is penetratin.
51. An agent according to any one of claims 32 to 48 wherein the agent is a fusion
protein.
52. An integrin subunit with a mutagenised binding domain for a MAP kinase or in
which the binding domain is deleted, or a homolog, analog, variant or derivative of the
integrin subunit, wherein capability to bind with the MAP kinase is thereby reduced with
the proviso that the integrin subunit is other than a βό Δ746-764 deletion mutant.
53. An integrin subunit according to claim 52 wherein the MAP kinase is an ERK
family membrane or a JNK family member.
54. An integrin subunit according to claim 53 wherein the MAP kinase is ERK2.
55. An integrin subunit according to any one of claims 52 to 55, wherein the integrin
subunit is βό.
56. An integrin subunit according to any one of claims 52 to55, wherein the integrin
subunit is a truncated integrin subunit.
57. An isolated nucleic acid sequence encoding a polypeptide as defined in any one of
claims 1 to 16, or a homolog, analog, variant or derivative of the polypeptide.
58. An isolated nucleic acid sequence encoding a fragment of an integrin subunit as
defined in any one of claims 17 to 31, or a homolog, analog, variant or derivative of the fragment.
59. An isolated nucleic acid sequence encoding an integrin subunit as defined in any
one of claims 52 to 56.
60. An isolated nucleic acid sequence encoding a fusion protein incoφorating a
polypeptide as defined in any one of claims 1 to 16, or a homolog, analog, variant or
derivative of the polypeptide.
61. An isolated nucleic acid sequence encoding a fusion protein incoφorating a
fragment as defined in any one of claims 17 to 31 . or a homolog, analog or variant of the
fragment.
62. An isolated nucleic acid sequence according to claim 60 or claim 61 wherein the
fusion protein further comprises a carrier polypeptide for facilitating passage of the
fusion protein across a cell membrane.
63. An isolated antisense nucleic acid sequence complementary to a nucleic acid
sequence as defined in claim 57 or 58.
64. An isolated antisense nucleic acid sequence according to claim 63 wherein the
antisense nucleic acid sequence is labelled.
65. A vector incoφorating a nucleic acid sequence as defined in any one of claims 57
to 59.
66. A vector incoφorating a nucleic acid sequence as defined in any one of claims 60
to 62.
67. A vector according to claim 65 or claim 66 wherein the vector is an expression
vector.
68. A host cell transformed with a vector as defined in any one of claims 65 to 67.
69. A host cell transformed with a vector as defined in claim 65 wherein the host cell
is selected from the group consisting of a mammalian cell, an epithelial cell, a neoplastic
cell and a cancer cell.
70. A host cell according to claim 69 wherein the host cell is a colon cancer cell.
71. A pharmaceutical composition comprising a polypeptide as defined in any one of
claims 1 to 16 or a homolog, analog, variant or derivative thereof, together with a
pharmaceutically acceptable carrier or diluent.
72. A pharmaceutical composition comprising a fragment as defined in any one of
claims 17 to 31 or a homolog, analog, variant or derivative thereof, together with a
pharmaceutically acceptable carrier or diluent.
73. A pharmaceutical composition comprising an agent as defined in any one of claims
32 to 51 together with a pharmaceutically acceptable carrier or diluent.
74. A pharmaceutical composition comprising a vector as defined in claim 65 together
with a pharmaceutically acceptable carrier or dil ent.
75. A pharmaceutical composition comprising an antisense nucleic acid sequence as
defined in claim 63 together with a pharmaceutically acceptable carrier or diluent.
76. An antibody capable of binding to a binding domain of an integrin for a MAP
kinase, or a binding fragment of the antibody.
77. An antibody capable of binding to a polypeptide as defined in any one of claims 1
to 16 or a homolog, analog, variant or derivative of the polypeptide.
78. An antibody capable of binding to a fragment as defined in any one of claims 17 to
31 or a homolog, analog, variant or derivative of the polypeptide.
79. An antibody according to any one of claims 76 to 78 wherein the antibody is a
monoclonal antibody.
80 A method of screening for an agent capable of inhibiting binding of a MAP kinase
to a binding domain of an integrin for the MAP kinase, comprising:
(a) testing a number of agents for ability to bind to either the MAP kinase or the
integrin; and
(b) determining if any said agent is capable of inhibiting binding of the MAP
kinase to the binding domain of the integrin on the basis of the testing.
81. A method of screening for an agent capable of inhibiting binding of a MAP kinase
to a binding domain of an integrin for the MAP kinase, comprising:
(a) testing a number of agents for ability to bind to either the MAP kinase or the
integrin;
(b) selecting an agent or agents identified as being able to bind to the MAP kinase
or the integrin on the basis of the testing; and
(c) utilising the selected said agent or agents in an assay for indicating whether the
or any of the selected said agents is capable of inhibiting the binding of the MAP kinase
to the binding domain of the integrin.
82. A method of evaluating whether an agenl i s capable of inhibiting binding of a
MAP kinase to a binding domain of an integrin for the MAP kinase, comprising:
(a) selecting the agent; (b) utilising the agent in an assay for indicating whether the agent is capable of
inhibiting the binding of the MAP kinase to the binding domain of the integrin; and
(c) determining if the agent is capable oϊ inhibiting the binding of the MAP kinase
to the binding domain of the integrin on the basis of the assay.
83. A method of screening for an agent capable of binding to a binding domain of an
integrin for a MAP kinase, comprising:
(a) testing a number of agents for ability to bind to the binding domain of the
integrin for the MAP kinase; and
(b) determining if any said agent is capable of binding to the binding domain of the
integrin on the basis of the testing.
84. A method of screening for an agent capable of binding to a binding domain of an
integrin for a MAP kinase, comprising:
(a) testing a number of agents for ability to bi d to the integrin;
(b) selecting an agent or agents identified as being able to bind to the integrin on
the basis of the testing; and
(c) utilising the selected said agent or agents in an assay for indicating whether the
or any of the selected said agents is capable of binding to the binding domain of the
integrin for the MAP kinase.
85. A method of evaluating whether an agent is capable of binding to a binding
domain of an integrin for a MAP kinase, comprising:
(a) testing the agent for ability to bind to the binding domain of the integrin for the
MAP kinase; and (b) determining if the agent is capable of binding to the binding domain on the basis of the testing.
86. A method according to any one of claims 83 to 85 wherein the testing comprises
utilising a polypeptide consisting of the binding domain of the integrin, or a homolog,
analog, variant or derivative of the polypeptide.
87. A method according to any one of claims 83 to 85 wherein the testing comprises
utilising a polypeptide comprising core amino acid sequence of the binding domain
wherein binding of the agent to the polypeptide indicates the agent is capable of binding
to the binding domain of the integrin, or a homolog, analog variant or derivative of the
polypeptide.
88. A method according to any one of claims 80 to 87 wherein the integrin comprises
β6.
89. A method according to any one of claims 80 to 88 wherein the MAP kinase is an
ERK family member of a JNK family member.
90. A method according to claim 89 wherein the MAP kinase is ERK2.
91. An agent identified to be capable of binding to a binding domain of an integrin for
a MAP kinase by a method as defined in any one of claims 80 to 82.
92. An agent identified to be capable of inhibiting binding of a MAP kinase to a
binding domain of an integrin for the MAP kinase by a method as defined in any one of
claims 83 to 85.
93. A method of isolating a MAP kinase from a sample utilising a molecule
immobilised on a solid support and which is capable of binding to a binding site on the MAP kinase which binding site binds with a binding domain of an integrin for the MAP kinase, comprising:
(a) contacting the molecule immobilised on the solid support with the sample
under conditions suitable for binding of the MAP kinase to the molecule;
(b) eluting the MAP kinase from the solid support; and
(c) collecting the eluted MAP kinase.
94. A method according to claim 93 wherein ihc molecule is an integrin subunit of the
integrin which integrin subunit incoφorates the binding domain, or a homolog, analog,
variant or derivative of the integrin subunit.
95. A method according to claim 93 wherein the molecule is a fragment of an integrin
subunit of the integrin which integrin subunit incorporates the binding domain, or a
homolog, analog, variant or derivative of the fragment.
96. A method according to claim 93 wherein the molecule is a polypeptide.
97. A method according to claim 96 wherein the polypeptide is other than an integrin
subunit incoφorating the binding domain or a fragment of the integrin subunit.
98. A method according to claim 97 wherein the polypeptide comprises amino acid
sequence RSKAKNPLYR.
99. A method according to claim 93 wherein the molecule is an antibody capable of
binding to the binding domain of the integrin for the MAP kinase.
100. A method according to claim 93 wherein the integrin comprises β6.
101. A method according to any one of claims 93 to 100 wherein the MAP kinase is an
ERK family member or a JKN family member.
102. A method according to claim 101 wherein the MAP kinase is ERK2.
103. A MAP kinase isolated from a sample by a method as defined in any one of claims 93 to 103.
104. A kit for determining if a test agent is capable of binding to a binding domain of an
integrin for a MAP kinase, wherein the kit comprises:
a molecule for forming with a test agenl a complex for being detected to indicate
the test agent is capable of binding to the binding domain of the integrin; and
instructions for use.
105. A kit for determining if a test agent is capable of inhibiting binding of a MAP
kinase to a binding domain of an integrin for the MAP kinase, wherein the kit comprises:
a molecule for forming with a test agent a complex for being detected to indicate
the test agent is capable of inhibiting binding of the \ A kinase to the binding domain
of the integrin; and
instructions for use.
106. A kit according to claim 104 or 105 wherein the molecule is a polypeptide
consisting of the binding domain of the integrin or a homolog. analog or variant or
derivative of the polypeptide.
107. A kit according to claim 104 or 105 wherein the molecule is a polypeptide
comprising core amino acid sequence of the binding domain, or a homolog, analog
variant or derivative of the polypeptide.
108. A kit according to any one of claims 104 to 107 wherein the integrin comprises βό.
109. A kit according to any one of claims 104 to 108 wherein the MAP kinase is an
ERK family member or a JNK family member.
110. A kit according to claim 109 wherein the MAP kinase is ERK2.
111. A method of modulating activity of a cell, comprising causing the expression of an
integrin to be down-regulated, wherein the integrin has a binding domain for a MAP
kinase.
112. A method according to claim 111 wherein the down-regulation of the expression
of the integrin is provided by an antisense nucleic acid sequence that inhibits expression
of a gene encoding an integrin subunit of the integrin.
113. A method according to claim 1 12 wherein the integrin subunit is βό.
114. A method according to any one of claims 1 1 1 to 1 13 wherein the antisense nucleic
acid sequence is administered to the cell or is transcribed from an expression vector
introduced into the cell.
115. A method according to any one of claims 1 1 1 to 1 14 wherein the antisense nucleic
acid sequence specifically hybridises with sense nucleic acid encoding for at least part of
the binding domain for the integrin.
116. A method according to any one of claims 1 1 1 to 115 wherein the cell is a cancer
cell.
117. A method according to any one of claims 1 1 1 to 1 1 6 wherein the MAP kinase is an
ERK family member or a JNK family member.
118. A method according to claim 117 wherein the MAP kinase is ERK2.
119. A method of modulating activity of a cell expressing an integrin, comprising:
transfecting the cell with a nucleic acid sequence encoding an integrin subunit or a
homolog, analog or variant thereof for being expressed by the cell, and which has a
mutagenised binding domain for a MAP kinase or in which the binding domain has been deleted whereby binding of the MAP kinase is thereby reduced, with the proviso that the
integrin subunit is other than a βό Δ746-764 deletion mutant.
120. A method according to claim 119 wherein the transfecting comprises transfecting
the cell with an expression vector incoφorating the nucleic acid sequence.
121. A method according to claim 119 or 120 wherein the MAP kinase is an ERK
family member or a JNK family member.
122. A method according to claim 121 wherein the MAP kinase is ERK2.
123. A method according to any one of claims 1 19 to 1 12 wherein the integrin subunit
is βό.
124. A method according to any one of claims 1 19 to 123 wherein the integrin subunit
is truncated.
125. A method of modulating activity of a cell expressing an integrin, comprising:
transfecting the cell with a nucleic acid sequence encoding a polypeptide for being
expressed by the cell wherein the polypeptide is capable of inhibiting binding of a MAP
kinase with a binding domain of the integrin f r the MAP kinase.
126. A method according to claim 125 wherein the polypeptide is capable of binding
with the binding site on the MAP kinase which binding site binds to the binding domain
of the integrin.
127. A method according to claim 125 or 126 wherein the polypeptide comprises the
binding domain of the integrin.
128. A method according to claim 126 or 127 wherein the polypeptide comprises the
amino acid sequence RSKAKWQTGTNPLYR., or a homolog, variant or analog of the
polypeptide.
129. A method according to claim 125 wherein the polypeptide comprises sufficient
core amino acid sequence of the binding domain of the integrin to enable the polypeptide
to bind to the binding site on the MAP kinase.
130. A method according to claim 129 wherein the polypeptide comprises the amino
acid sequence RSKAKNPLYR, or a homolog. analog, variant or derivative of the
polypeptide.
131. A method according to any one of claims 125 to 1 0 wherein the nucleic acid
sequence is incorporated in an expression vector.
132. A method according to any one of claims 125 to 13 1 wherein the integrin
comprises β6.
133. A method according to any one of claims 1 25 to 1 32 w herein the MAP kinase is an
ERK family member or a JNK family member.
134. A method according to claim 133 wherein the MAP kinase is ERK2.
135. A method according to any one of claims 125 to 134 wherein the cell is a cancer
cell.
136. A method of modulating activity of a cell comprising:
contacting the cell with an effective amount of an agent capable of inhibiting
binding of a MAP kinase to a binding domain of an integrin expressed by the cell.
137. A method according to claim 136 wherein the agent is an agent as defined in any
one of claims 32 to 51.
138. A method according to claim 136 wherein the agent is a polypeptide as defined in
any one of claims 1 to 16 or a homolog, analog, variant or derivative thereof.
139. A method according to claim 136 wherein ihc ageni is a fragment as defined in any
one of claims 17 to 31 or a homolog, analog, variant or derivative thereof.
140. A method according to any one of claims 136 to 139 wherein the MAP kinase is an
ERK family member or a JNK family member.
141. A method according to claim 140 wherein the MAP kinase is ERK2.
142. A method according to any one of claims 136 to 141 wherein the integrin
comprises β6.
143. A method according to any one of claims 136 to 142 wherein the activity of the
cell is growth of the cell.
144. A method according to any one of claims 136 to 143 wherein the cell is a cancer
cell.
145. A method according to claim 144 wherein the cancer cell is a colon cancer cell.
146. A method of modulating activity of a cell, comprising contacting the cell with an
effective amount of an agent for down regulating functional activity of an integrin
expressed by the cell and thereby the activity of ihc ceil
147. A method according to claim 146 wherein ihe agent inhibits binding of a MAP
kinase to a binding domain of the integrin for the MAP kinase.
148. A method according to claim 146 wherein the agent is a polypeptide as defined in
any one of claims 1 to 16 or a homolog, analog, variant or derivative thereof.
149. A method according to claim 136 wherein the agent is a fragment as defined in any
one of claims 17 to 31 or a homolog, analog, variant or derivative thereof.
150. A method according to claim 146 wherein the agent is an agent as defined in any
one of claims 32 to 51.
151. A method according to any one of claims 146 to 150 wherein the MAP kinase is an
ERK or a JNK family member.
152. A method according to claim 151 wherein the MAP kinase is ERK2.
153. A method according to any one of claims 146 to 152 wherein the integrin
comprises β6.
154. A method according to any one of claims 146 to 1 53 wherein the activity of the
cell is growth of the cell.
PCT/AU2000/000729 1999-06-26 2000-06-28 A method of modulating integrin mediated cellular activity and agents useful for same WO2001000677A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU55106/00A AU781540B2 (en) 1999-06-28 2000-06-28 A method of modulating integrin mediated cellular activity and agents useful for same
US10/019,816 US8119594B1 (en) 1999-06-28 2000-06-28 Method of modulating integrin mediated cellular activity and agents useful for same
EP00940024A EP1196449B1 (en) 1999-06-28 2000-06-28 A method of modulating integrin mediated cellular activity and agents useful for same
DE60037970T DE60037970T2 (en) 1999-06-28 2000-06-28 PROCESS FOR MODULATING INTEGRIN-MEDIATED CELL ACTIVITY AND SUITABLE REAGENTS
AU2005204275A AU2005204275B2 (en) 1999-06-28 2005-08-26 A method of modulating integrin mediated cellular activity and agents useful for same
US13/349,129 US8618062B2 (en) 1999-06-26 2012-01-12 Method of modulating integrin mediated cellular activity and agents useful for same
US13/832,796 US20140187612A1 (en) 1999-06-28 2013-03-15 Method of modulating integrin mediated cellular activity and agents useful for same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
AUPQ1248A AUPQ124899A0 (en) 1999-06-28 1999-06-28 A method of treatment and agents useful for same
AUPQ1248 1999-06-28
AUPQ8003 2000-06-06
AUPQ8003A AUPQ800300A0 (en) 2000-06-06 2000-06-06 A method of treatment and agents useful for same

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/019,816 A-371-Of-International US8119594B1 (en) 1999-06-28 2000-06-28 Method of modulating integrin mediated cellular activity and agents useful for same
US13/349,129 Division US8618062B2 (en) 1999-06-26 2012-01-12 Method of modulating integrin mediated cellular activity and agents useful for same

Publications (2)

Publication Number Publication Date
WO2001000677A1 true WO2001000677A1 (en) 2001-01-04
WO2001000677A9 WO2001000677A9 (en) 2002-11-07

Family

ID=25646086

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2000/000729 WO2001000677A1 (en) 1999-06-26 2000-06-28 A method of modulating integrin mediated cellular activity and agents useful for same

Country Status (5)

Country Link
US (2) US8119594B1 (en)
EP (1) EP1196449B1 (en)
AT (1) ATE385503T1 (en)
DE (1) DE60037970T2 (en)
WO (1) WO2001000677A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001094570A2 (en) * 2000-06-07 2001-12-13 Otogene Aktiengesellschaft Transfection method
EP1352055A1 (en) * 2000-12-22 2003-10-15 The University Of Newcastle Research Associates Limited Map kinase integrin-binding domain
WO2005037308A1 (en) * 2003-10-17 2005-04-28 Inter-K Pty Limited Methods and agents for the treatment of cancer
WO2010094085A1 (en) 2009-02-23 2010-08-26 Inter-K Pty Limited Inhibition of multiple cell activation pathways
US8119594B1 (en) 1999-06-28 2012-02-21 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
US8563306B2 (en) 2008-03-31 2013-10-22 Council Of Scientific & Industrial Research Tumor model system useful to study multistage cancer
JP2015510393A (en) * 2012-01-24 2015-04-09 インター・ケー・ピーティーワイ・リミテッド Peptide agent for cancer treatment
US9296784B2 (en) 2002-07-12 2016-03-29 The Johns Hopkins University Mesothelin vaccines and model systems

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110105408A1 (en) * 2008-02-22 2011-05-05 Inter-K Pty Limited Therapeutic peptides

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992012236A1 (en) * 1991-01-11 1992-07-23 The Regents Of The University Of California A NOVEL INTEGRIN β SUBUNIT AND USES THEREOF

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE911869A1 (en) 1990-06-01 1991-12-04 Regeneron Pharma A family of map2 protein kinases
US5177188A (en) * 1990-12-03 1993-01-05 The Scripps Research Institute Methods and compositions for diagnosing chronic immune thrombocytopenic purpura
US5962643A (en) 1991-07-11 1999-10-05 The Regents Of The University Of California Integrin β subunit and uses thereof
US6514745B1 (en) 1993-07-19 2003-02-04 The Regents Of The University Of California Oncoprotein protein kinase
US6495518B1 (en) 1994-06-13 2002-12-17 Vanderbilt University Method for importing biologically active molecules into cells
US5807746A (en) 1994-06-13 1998-09-15 Vanderbilt University Method for importing biologically active molecules into cells
US5877282A (en) 1996-09-20 1999-03-02 Bristol-Myers Squibb Company Peptide inhibitors of nuclear protein translocation having nuclear localization sequences and methods of use thereof
AU4815397A (en) * 1996-10-15 1998-05-11 Vanderbilt University Method of disrupting cellular adhesion
US6007991A (en) 1997-03-28 1999-12-28 The Research Foundation Of Suny Antisense oligonucleotides for mitogen-activated protein kinases as therapy for cancer
US6133246A (en) 1997-08-13 2000-10-17 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US5877309A (en) 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US6248558B1 (en) 1998-03-31 2001-06-19 Vanderbilt University Sequence and method for genetic engineering of proteins with cell membrane translocating activity
CA2325817A1 (en) * 1998-04-15 1999-10-21 Cor Therapeutics, Inc. Transgenic mammals expressing mutant gp iiia
ATE385503T1 (en) 1999-06-28 2008-02-15 Inter K Pty Ltd METHOD FOR MODULATING INTEGRIN-MEDIATED CELL ACTIVITY AND REAGENTS SUITABLE FOR THIS
US6312956B1 (en) 1999-10-01 2001-11-06 Vanderbilt University Nuclear targeted peptide nucleic acid oligomer
WO2001097827A1 (en) * 2000-06-16 2001-12-27 Argonex Pharmaceuticals Mhc peptides over-expressed on prostate cancer cells and methods of use
US7198895B2 (en) * 2000-11-14 2007-04-03 Mohanlal Ramon W In vitro cell-based methods for biological validation and pharmacological screening of chemical entities and biologicals

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992012236A1 (en) * 1991-01-11 1992-07-23 The Regents Of The University Of California A NOVEL INTEGRIN β SUBUNIT AND USES THEREOF

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
CHEN ET AL.: "Distinct structural requirements for interaction of the integrins alpha5beta1, alphavbeta5 and alphavbeta6 with the central cell binding domain in fibronectin", CELL ADHESION AND COMMUNICATION, vol. 4, no. 4-5, 1996, pages 237 - 250 *
CONE ET AL.: "Effects of beta subunit cytoplasmic domain deletions on the recruitment of the integrin alphavbeta6 to focal contacts", CELL ADHESION AND COMMUNICATION, vol. 2, 1994, pages 101 - 113 *
COPPOLINO AND DEDHAR: "Bi-directional signal transduction by integrin receptors", THE INTERNATIONAL JOURNAL OF BIOCHEMISTRY & CELL BIOLOGY, vol. 32, 2000, pages 171 - 188 *
DIXIT ET AL.: "Identification of a sequence within the integrin beta6 subunit cytoplasmic domain that is required to support the specific effect of alphavbeta6 on proliferation in three-dimensional culture", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, no. 42, 1996, pages 25976 - 25980 *
GOTOH ET AL.: "Cross-linking of integrins induces tyrosine phosphorylation of the proto-oncogene product Vav and the protein tyrosine kinase Syk in human factor-dependent myeloid cells", CELL GROWTH AND DIFFERENTIATION, vol. 8, 1997, pages 721 - 729 *
LIN ET AL.: "Integrin-mediated activation of MAP kinase is independent of FAK: Evidence for dual integrin signaling pathways in fibroblasts", THE JOURNAL OF CELL BIOLOGY, vol. 136, no. 6, 1997 *
SHEPPARD ET AL.: "Complete amino acid sequence of a novel integrin beta subunit (beta6) identified in epithelial cells using the polymerase chain reaction", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 265, no. 20, 1990, pages 11502 - 11507 *
YOKOSAKI ET AL.: "Differential effects of the integrins alphavbeta3 and alphavbeta6 on cell proliferative responses to tenascin: Roles of the beta subunit extracellular and cytoplasmic domains", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, no. 39, 1996, pages 24144 - 24150 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8618062B2 (en) 1999-06-26 2013-12-31 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
US8119594B1 (en) 1999-06-28 2012-02-21 Inter-K Pty Limited Method of modulating integrin mediated cellular activity and agents useful for same
WO2001094570A3 (en) * 2000-06-07 2002-06-27 Otogene Ag Transfection method
WO2001094570A2 (en) * 2000-06-07 2001-12-13 Otogene Aktiengesellschaft Transfection method
EP1352055A1 (en) * 2000-12-22 2003-10-15 The University Of Newcastle Research Associates Limited Map kinase integrin-binding domain
EP1352055A4 (en) * 2000-12-22 2004-07-07 Univ Newcastle Res Ass Map kinase integrin-binding domain
US7422883B2 (en) 2000-12-22 2008-09-09 Inter-K Pty Limited Map kinase integrin-binding domain
US10350282B2 (en) 2002-07-12 2019-07-16 The Johns Hopkins University Mesothelin vaccines and model systems
US9296784B2 (en) 2002-07-12 2016-03-29 The Johns Hopkins University Mesothelin vaccines and model systems
GB2409679B (en) * 2003-10-17 2005-12-28 Inter K Pty Ltd Anti-cancer agents which inhibit the interaction between integrins and MAP kinases.
EP2418219A1 (en) 2003-10-17 2012-02-15 Inter-K Pty Limited Methods and agents for the treatment of cancer
WO2005037308A1 (en) * 2003-10-17 2005-04-28 Inter-K Pty Limited Methods and agents for the treatment of cancer
AU2011200901B2 (en) * 2003-10-17 2012-02-02 Inter-K Pty Limited Methods and Agents for the Treatment of Cancer
US9765113B2 (en) 2003-10-17 2017-09-19 Interk Peptide Therapeutics Limited Methods and agents for the treatment of cancer
GB2409679A (en) * 2003-10-17 2005-07-06 Inter K Pty Ltd Pharmaceutical preparation comprising an inhibitor of the integrin-MAP kinase interaction
US9131668B2 (en) 2008-03-31 2015-09-15 Council For Scientific And Industrial Research Tumor model system useful to study multistage cancer
US8563306B2 (en) 2008-03-31 2013-10-22 Council Of Scientific & Industrial Research Tumor model system useful to study multistage cancer
JP2012518602A (en) * 2009-02-23 2012-08-16 インター−ケイ ピーティーワイ リミテッド Inhibition of multiple cell activation pathways
WO2010094085A1 (en) 2009-02-23 2010-08-26 Inter-K Pty Limited Inhibition of multiple cell activation pathways
EP2398484A4 (en) * 2009-02-23 2013-07-10 Inter K Pty Ltd Inhibition of multiple cell activation pathways
EP2398484A1 (en) * 2009-02-23 2011-12-28 Inter-K Pty Limited Inhibition of multiple cell activation pathways
US20150110744A1 (en) * 2012-01-24 2015-04-23 Inter-K Pty Limited Peptide agents for cancer therapy
JP2015510393A (en) * 2012-01-24 2015-04-09 インター・ケー・ピーティーワイ・リミテッド Peptide agent for cancer treatment
US9403877B2 (en) 2012-01-24 2016-08-02 Inter-K Pty Limited Peptide agents for cancer therapy

Also Published As

Publication number Publication date
DE60037970D1 (en) 2008-03-20
EP1196449A4 (en) 2004-06-16
US8618062B2 (en) 2013-12-31
EP1196449A1 (en) 2002-04-17
ATE385503T1 (en) 2008-02-15
EP1196449B1 (en) 2008-02-06
WO2001000677A9 (en) 2002-11-07
DE60037970T2 (en) 2008-08-21
US8119594B1 (en) 2012-02-21
US20120232249A1 (en) 2012-09-13

Similar Documents

Publication Publication Date Title
US8618062B2 (en) Method of modulating integrin mediated cellular activity and agents useful for same
Buday et al. Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor
Qin et al. The transcription factor E2F-1 is a downstream target of RB action
Hsia et al. Differential regulation of cell motility and invasion by FAK
EP1352055B1 (en) Map kinase integrin-binding domain
EP0636174B1 (en) Hyaluronan receptor (rhamm = receptor for hyluronan mediated mobility) and hyaluronan binding peptides
US9765113B2 (en) Methods and agents for the treatment of cancer
Xing et al. Characterization of ret oncogenic activation in MEN2 inherited cancer syndromes
US20140187612A1 (en) Method of modulating integrin mediated cellular activity and agents useful for same
AU2004281152B2 (en) Cystatin C as an antagonist of tgf-b and methods related thereto
AU781540B2 (en) A method of modulating integrin mediated cellular activity and agents useful for same
AU2005204275B2 (en) A method of modulating integrin mediated cellular activity and agents useful for same
AU2001297809B2 (en) MAP kinase integrin-binding domain
AU2001297809A1 (en) MAP kinase integrin-binding domain
AU2007231835A1 (en) Screening assay for an inhibitor of MAP kinase?integrin binding
WO1999035283A1 (en) Methods and reagents for modulating cell motility
CA2542608A1 (en) Igf-i responsive gene and use thereof
AU741133B2 (en) SH2 domain-containing peptides
WO2003060075A2 (en) USE OF SOLUBLE TYPE II TGF-β RECEPTOR TO SUPPRESS PANCREATIC CANCER GROWTH AND METASTASIS
Zhanga et al. Caveolin-1 Inhibits Epidermal Growth Factor-stimulated Lamellipod Extension and Cell Migration in Metastatic Mammary Adenocarcinoma Cells (MTLn3)
IE20040691A1 (en) IGF-I responsive gene and use thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ 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 GW ML MR NE SN TD TG

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

Ref document number: 2000940024

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 55106/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 10019816

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 2000940024

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

AK Designated states

Kind code of ref document: C2

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

AL Designated countries for regional patents

Kind code of ref document: C2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ 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 GW ML MR NE SN TD TG

NENP Non-entry into the national phase

Ref country code: JP

WWG Wipo information: grant in national office

Ref document number: 55106/00

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: 2000940024

Country of ref document: EP