WO2013045934A2 - Compositions comprising agents that inhibit neuropilin and tolloid like 2 - Google Patents

Compositions comprising agents that inhibit neuropilin and tolloid like 2 Download PDF

Info

Publication number
WO2013045934A2
WO2013045934A2 PCT/GB2012/052400 GB2012052400W WO2013045934A2 WO 2013045934 A2 WO2013045934 A2 WO 2013045934A2 GB 2012052400 W GB2012052400 W GB 2012052400W WO 2013045934 A2 WO2013045934 A2 WO 2013045934A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
composition according
neto
cells
pharmaceutical composition
Prior art date
Application number
PCT/GB2012/052400
Other languages
French (fr)
Other versions
WO2013045934A3 (en
Inventor
Richard Birnie
Norman Maitland
Original Assignee
Procure Therapeutics Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Procure Therapeutics Limited filed Critical Procure Therapeutics Limited
Priority to CA2847826A priority Critical patent/CA2847826A1/en
Priority to EP12780520.8A priority patent/EP2732036A2/en
Publication of WO2013045934A2 publication Critical patent/WO2013045934A2/en
Publication of WO2013045934A3 publication Critical patent/WO2013045934A3/en
Priority to US14/227,538 priority patent/US20140199310A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1203Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
    • C07K16/1228Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • compositions Comprising Agents that Inhibit Neuropilin and Tolloid Like 2 Field of the Invention
  • the disclosure relates to agents that inhibit the expression or activity of neuropilin and tolloid like 2 [NETO-2] which has elevated expression in cancer stem cells; the monitoring of expression of NETO-2 as a diagnostic or prognostic marker of tumour initiation; the use NETO-2 polypeptide in the identification of agents that inhibit activity; and including vaccines comprising NETO-2 polypeptides and antibodies that binds NETO-2.
  • NETO-2 neuropilin and tolloid like 2
  • stem cell represents a generic group of undifferentiated cells that possess the capacity for self-renewal while retaining varying potential to form differentiated cells and tissues.
  • Stem cells can be pluripotent or multipotent.
  • a pluripotent stem cell is a cell that has the ability to form all tissues found in an intact organism although the pluripotent stem cell cannot form an intact organism.
  • a multipotent cell has a restricted ability to form differentiated cells and tissues.
  • adult stem cells are multipotent stem cells and are the precursor stem cells or lineage restricted stem cells that have the ability to form some cells or tissues and replenish senescing or damaged cells/tissues. Generally they cannot form all tissues found in an organism although some reports have claimed a greater potential for such 'adult' stem cells than originally thought.
  • tumour stem cells are clonal and are therefore derived from a single cell.
  • cancer stem cells There are few studies that identify and characterize those cells types that are responsible for maintaining tumour cell growth. Some have searched for these so called “cancer stem cells”.
  • tumour cells for example, in leukaemia, the ability to initiate new tumour growth resides in a rare phenotypically distinct subset of tumour cells [Bonnet D, Dick J.E. Human acute myeloid leukaemia is organized as a hierarchy that originates from a primitive hematopoietic cell Nat. Med. 1997, 3: 730737] which are defined by the expression of CD34 and CD38 surface antigens and have been termed leukaemia stem cells.
  • tumour-initiating cells have also been found in 'solid' cancers such as prostate [Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ: Prospective Identification of Tumorigenic Prostate Cancer Stem Cells. Cancer Res. 2005, 65: 1094610951 ], breast [Al Hajj M, Wicha MS, BenitoHernandez A, Morrison SJ, Clarke MF: Prospective identification of tumorigenic breast cancer cells.
  • colon [O'Brien CA, Pollett A, Gallinger S, Dick JE: A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007, 445: 1061 10; RicciVitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R: Identification and expansion of human colon cancer initiating cells. Nature 2007, 445: 1 1 1 1 15]; and gastric cancers [Houghton J, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, Cai X, Fox JG, Goldenring JR, Wang TC: Gastric cancer originating from bone marrow derived cells. Science 2004, 306: 15681571 ].
  • This disclosure relates to the identification of NETO-2 which has enhanced expression in cancer stem cells and in particular prostate cancer stem cells and wherein expression is correlated with tumour cell initiation.
  • WO2005/089043 we describe the isolation of prostate stem cells which have been directly isolated from lymph node and prostate glands from a series of patient samples. These stem cells express markers that characterise the cells with stem cell properties. The following markers are typically expressed as prostate stem cell markers; human epithelial antigen (HEA), CD44, a 2 3i hl and CD133.
  • HAA human epithelial antigen
  • CD44 CD44
  • This disclosure identifies NETO-2 the expression of which is critical for colony formation which is the in initiating step in the formation of a tumour.
  • NETO-2 NETO-2-binds to NETO-2.
  • the expression of NETO-2 therefore represents the early stages of tumour formation and allows an early therapeutic intervention with consequent inhibition of tumour formation.
  • the detection of NETO-2 also serves as a diagnostic or prognostic marker of the early stages of tumour formation.
  • NETO-2 exists as two isoforms. The full length isoform is 525 amino acids in length. A second isoform is a shorter version and is missing amino acid residues 1 -324 and has a different amino acid sequence between amino acid residues 325-333.
  • NETO-2 is a single span transmembrane receptor and is known interact with glutamate receptor which is primarily expressed in the brain.
  • an agent that inhibits the expression of NETO-2 or the activity of NETO-2 wherein said expression/activity is enhanced in a cancer stem cell and wherein the agent inhibits tumour initiation.
  • NETO-2 comprises or consists of the nucleotide sequence as represented in Figure 1 A.
  • said agent is an antisense oligonucleotide or RNA.
  • composition comprising an agent that inhibits tumour initiation wherein the agent is selected from the group consisting of:
  • an agent comprising one or more antisense oligonucleotide or antisense RNA molecules wherein said oligonucleotide or antisense RNA molecule comprise a nucleotide sequence adapted to anneal to a sense nucleotide sequence derived from at least one gene represented by the sense sequence presented in Figure 1 a; or
  • an agent comprising an antibody, or active antibody binding fragment thereof, that binds to the extracellular domain of NETO 2.
  • compositions comprising one or more antisense oligonucleotide or RNA molecules wherein said oligonucleotide or antisense RNA molecule comprise a nucleotide sequence adapted to anneal to a sense nucleotide sequence derived from at least one gene represented by the sense sequence presented in Figure 1 A.
  • said composition is a pharmaceutical composition.
  • said composition comprises, one, two, three or four antisense oligonucleotides or RNA molecules.
  • said composition consists essentially of one or more oligonucleotides or antisense RNA molecules and physiologically compatible excipients and/or adjuvants.
  • said antisense RNA molecule is part of a siRNA or shRNA molecule.
  • siRNA small inhibitory or interfering RNA
  • the siRNA molecule comprises two complementary strands of RNA (a sense strand and an antisense strand) annealed to each other to form a double stranded RNA molecule.
  • the siRNA molecule is typically derived from exons of the gene which is to be ablated. The mechanism of RNA interference is being elucidated. Many organisms respond to the presence of double stranded RNA by activating a cascade that leads to the formation of siRNA.
  • RNA double stranded RNA activates a protein complex comprising RNase III which processes the double stranded RNA into smaller fragments (siRNAs, approximately 21 -29 nucleotides in length) which become part of a ribonucleoprotein complex.
  • the siRNA acts as a guide for the RNase complex to cleave mRNA complementary to the antisense strand of the siRNA thereby resulting in destruction of the mRNA.
  • said antisense RNA molecule is between 19 nucleotides [nt] and 29nt in length. More preferably still said antisense RNA molecule is between 21 nt and 27nt in length. Preferably said antisense RNA molecule is about 21 nt in length.
  • said antisense RNA consists of 21 nt.
  • said siRNA is selected from the nucleotide sequences set forth in SEQ ID NO: 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52, 53, 54 or 55.
  • said antisense, siRNA or shRNA includes modified nucleotides.
  • modified as used herein describes a nucleic acid molecule in which;
  • i) at least two of its nucleotides are covalently linked via a synthetic internucleoside linkage (i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide).
  • a synthetic internucleoside linkage i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide.
  • said linkage may be the 5' end of one nucleotide linked to the 5' end of another nucleotide or the 3' end of one nucleotide with the 3' end of another nucleotide; and/or ii) a chemical group, such as cholesterol, not normally associated with nucleic acids has been covalently attached to the double stranded nucleic acid.
  • Preferred synthetic internucleoside linkages are phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, phosphate triesters, acetamidates, peptides, and carboxymethyl esters.
  • modified nucleotides also encompasses nucleotides with a covalently modified base and/or sugar.
  • modified nucleotides include nucleotides having sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3' position and other than a phosphate group at the 5' position.
  • modified nucleotides may also include 2' substituted sugars such as 2'-0-methyl-; 2-O-alkyl; 2-O-allyl; 2'-S-alkyl; 2'-S-allyl; 2'- fluoro-; 2'-halo or 2;azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, and sedoheptulose.
  • 2' substituted sugars such as 2'-0-methyl-; 2-O-alkyl; 2-O-allyl; 2'-S-alkyl; 2'-S-allyl; 2'- fluoro-; 2'-halo or 2;azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses
  • Modified nucleotides include, by example and not by way of limitation, alkylated purines and/or pyrimidines; acylated purines and/or pyrimidines; or other heterocycles. These classes of pyrimidines and purines are known in the art and include, pseudoisocytosine; N4, N4-ethanocytosine; 8-hydroxy-N6-methyladenine; 4- acetylcytosine, 5-(carboxyhydroxylmethyl) uracil; 5-fluorouracil; 5-bromouracil;5- carboxymethylaminomethyl-2-thiouracil; 5 carboxymethylaminomethyl uracil; dihydrouracil; inosine; N6-isopentyl-adenine; l-methyladenine; 1 -methylpseudouracil; 1 - methylguanine; 2,2-dimethylguanine; 2-methyladenine; 2-methylguanine; 3- methyl
  • Modified double stranded nucleic acids also can include base analogs such as C-5 propyne modified bases (see Wagner et al., Nature Biotechnology 14:840-844, 1996).
  • said pharmaceutical composition includes a carrier adapted to deliver said antisense RNA to a cell or tissue.
  • siRNA can be chemically modified and conjugated to a lipophilic cholesterol moiety at the 3' end of the sense strand.
  • Cationic delivery systems can also be employed in the delivery of siRNA. These include cationic lipids and liposomes, cationic polymers, cationic dendrimers and cationic cell penetrating peptides.
  • the cationic delivery vehicles have a common positive charge which facilitates complex formation with negatively charged siRNA.
  • liposome based delivery vehicles include Lipofectin, RNAifect, Oligofectamine, Lipofectamine and TransIT TKO have been used in vitro.
  • DOTAP N [1 -(2, 3- dioleoyloxy)]-N, N, N-trimethyl ammonium propane
  • Oligfectamine Oligfectamine
  • Other liposome based delivery vehicle includes solid nucleic acid lipid particles [SNALPs] which are also conjugated with polyethylene glycol.
  • Peptide delivery vehicles have also been successful in delivering siRNA.
  • Pegylated polyethyleneimine [PEI] comprising RGD peptides have been used to target siRNA to angiogenesis factors such as VEGF.
  • Atelocollagen has been used in the delivery of siRNA to tumours in vivo. Delivery of siRNA has also been demonstrated using cyclodextrin polymers.
  • LPD nanoparticles which have been used to deliver to solid and metastatic tumours.
  • LPD nanoparticles comprise cationic lipids combined with protamine which interacts with negatively charged siRNA.
  • Pegylated versions of LPD nanoparticles are also known which have improved pharmacokinetics.
  • said antibody is a polyclonal antibody.
  • said antibody is a monoclonal antibody.
  • Immunoglobulins are protein molecules which have specificity for foreign molecules (antigens).
  • Immunoglobulins are a class of structurally related proteins consisting of two pairs of polypeptide chains, one pair of light (L) (low molecular weight) chain ( ⁇ or ⁇ ), and one pair of heavy (H) chains ( ⁇ , ⁇ , ⁇ , ⁇ and ⁇ ), all four linked together by disulphide bonds.
  • L light
  • H heavy chains
  • Both H and L chains have regions that contribute to the binding of antigen and that are highly variable from one Ig molecule to another.
  • H and L chains contain regions that are non-variable or constant.
  • the L chains consist of two domains.
  • the carboxy-terminal domain is essentially identical among L chains of a given type and is referred to as the "constant" (C) region.
  • the amino terminal domain varies from L chain to L chain and contributes to the binding site of the antibody. Because of its variability, it is referred to as the "variable” (V) region.
  • the H chains of Ig molecules are of several classes, ⁇ , ⁇ , ⁇ , a, and ⁇ (of which there are several sub-classes).
  • An assembled Ig molecule consisting of one or more units of two identical H and L chains, derives its name from the H chain that it possesses.
  • Ig isotypes there are five Ig isotypes: IgA, IgM, IgD, IgE and IgG (with four sub-classes based on the differences in the H chains, i.e., lgG1 , lgG2, lgG3 and lgG4). Further detail regarding antibody structure and their various functions can be found in, Using Antibodies: A laboratory manual, Cold Spring Harbour Laboratory Press.
  • said fragment is a single chain antibody fragment.
  • a Fab fragment is a multimeric protein consisting of the immunologically active portions of an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region, covalently coupled together and capable of specifically binding to an antigen.
  • Fab fragments are generated via proteolytic cleavage (with, for example, papain) of an intact immunoglobulin molecule.
  • a Fab 2 fragment comprises two joined Fab fragments. When these two fragments are joined by the immunoglobulin hinge region, a F(ab') 2 fragment results.
  • An Fv fragment is multimeric protein consisting of the immunologically active portions of an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region covalently coupled together and capable of specifically binding to an antigen.
  • a fragment could also be a single chain polypeptide containing only one light chain variable region, or a fragment thereof that contains the three CDRs of the light chain variable region, without an associated heavy chain variable region, or a fragment thereof containing the three CDRs of the heavy chain variable region, without an associated light chain moiety; and multi specific antibodies formed from antibody fragments, this has for example been described in US patent No 6,248,516.
  • Fv fragments or single region (domain) fragments are typically generated by expression in host cell lines of the relevant identified regions.
  • a fragment of an antibody or immunoglobulin can also have bispecific function as described above.
  • said antibody is a chimeric antibody.
  • said antibody is a humanized or human antibody.
  • Chimeric antibodies are recombinant antibodies in which all of the V-regions of a mouse or rat antibody are combined with human antibody C-regions.
  • Humanised antibodies are recombinant hybrid antibodies which fuse the complementarity determining regions from a rodent antibody V-region with the framework regions from the human antibody V- regions. The C-regions from the human antibody are also used.
  • the complementarity determining regions (CDRs) are the regions within the N-terminal domain of both the heavy and light chain of the antibody to where the majority of the variation of the V- region is restricted. These regions form loops at the surface of the antibody molecule. These loops provide the binding surface between the antibody and antigen. Antibodies from non-human animals provoke an immune response to the foreign antibody and its removal from the circulation.
  • Both chimeric and humanised antibodies have reduced antigenicity when injected to a human subject because there is a reduced amount of rodent (i.e. foreign) antibody within the recombinant hybrid antibody, while the human antibody regions do not ellicit an immune response. This results in a weaker immune response and a decrease in the clearance of the antibody. This is clearly desirable when using therapeutic antibodies in the treatment of human diseases. Humanised antibodies are designed to have less "foreign" antibody regions and are therefore thought to be less immunogenic than chimeric antibodies.
  • compositions of the present invention are administered in pharmaceutically acceptable preparations.
  • Such preparations may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers and supplementary anti-cancer agents.
  • compositions of the invention can be administered by any conventional route, including injection or by gradual infusion over time.
  • Treatment may be topical or systemic.
  • the administration may, for example, be oral, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, transdermal, transepithelial or intra bone marrow administration or by direct injection into the tumour mass.
  • compositions of the invention are administered in effective amounts.
  • An "effective amount” is that amount of a composition that alone, or together with further doses, produces the desired response.
  • the desired response is inhibiting the progression of the disease. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods.
  • Such amounts will depend, of course, on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
  • compositions used in the foregoing methods preferably are sterile and contain an effective amount of an agent according to the invention for producing the desired response in a unit of weight or volume suitable for administration to a patient.
  • the doses of the antisense RNA according to the invention administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. Other factors include the desired period of treatment. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
  • doses of antisense RNA e.g., siRNA
  • doses can range from 1 nM-500nM, 5nM-200nM, and 10nM-100nM.
  • Other protocols for the administration of compositions will be known to one of ordinary skill in the art, in which the dose amount, schedule of injections, sites of injections, mode of administration and the like vary from the foregoing.
  • the administration of compositions to mammals other than humans, is carried out under substantially the same conditions as described above.
  • a subject, as used herein, is a mammal, preferably a human, and including a non-human primate, cow, horse, pig, sheep, goat, dog, cat or rodent.
  • the pharmaceutical preparations of the invention When administered, the pharmaceutical preparations of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptable compositions.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients.
  • Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents' (e.g. anti-inflammatory agents such as steroids, non-steroidal anti-inflammatory agents, chemotherapeutic agents).
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention.
  • Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like.
  • pharmaceutically- acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
  • compositions may be combined, if desired, with a pharmaceutically-acceptable carrier.
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration into a human.
  • carrier in this context denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application, (e.g. liposome or immuno-liposome).
  • the components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
  • the pharmaceutical compositions may contain suitable buffering agents, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • suitable buffering agents including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • suitable preservatives such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active compound.
  • Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as syrup, elixir or an emulsion or as a gel.
  • Compositions may be administered as aerosols and inhaled.
  • compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of agent, which is preferably isotonic with the blood of the recipient.
  • This preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation also may be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example, as a solution in 1 , 3-butane diol.
  • the acceptable solvents that may be employed are water, Ringer's solution , and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono-or di-glycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectables.
  • Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA.
  • said composition includes an additional chemotherapeutic agent.
  • chemotherapeutic agent is an agent that typical is a small chemical compound that kills cells in particular diseased cells, for example cancer cells.
  • the agents can be divided with respect to their structure or mode of action.
  • chemotherapeutic agents include alkylating agents, anti-metabolites, anthracyclines, alkaloids, plant terpenoids and toposisomerase inhibitors.
  • Chemotherapeutic agents typically produce their effects on cell division or DNA synthesis.
  • said chemotherapeutic agent is an alkylating agent.
  • said alkylating agent is selected from the group consisting of: cisplatin, carboplatin or oxaliplatin.
  • said chemotherapuetic agent is an anti- metabolic drug.
  • said drug is a purine analogue. In an alternative preferred embodiment of the invention said drug is a pyrimidine analogue.
  • Purine analogues are known in the art; for example thioguanine is used to treat acute leukaemia; fludarabine inhibits the function of DNA polymerases, DNA primases and DNA ligases and is specific for cell-cycle S-phase; pentostatin and cladribine are adenosine analogues and are effective against hairy cell leukaemias.
  • mecrcaptopurine which is an adenine analogue.
  • Pyrimidine analogues are similarly known in the art.
  • 5-fluorouracil (5-FU) floxuridine and cytosine arabinoside. 5-FU has been used for many years in the treatment of breast, colorectal cancer, pancreatic and other cancers. 5-FU can also been formed from the pro-drug capecitabine which is converted to 5-FU in the tumour.
  • said chemotherapeutic agent is 5- fluorouracil.
  • said anti-metabolic drug is administered with leucovorin.
  • Leucovorin also known as folinic acid
  • folinic acid is administered as an adjuvant in cancer chemotherapy and which enhances the inhibitory effects of 5-FU on thymidylate synthase.
  • said chemotherapeutic agent is an alkaloid; preferably said alkaloid is a vinca alkaloid, for example vincristine or vinblastine.
  • said chemotherapeutic agent is a terpenoid; preferably a taxane e.g. palitaxel.
  • a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence as represented in Figure 1 a for the identification of agents that modulate the activity of said polypeptide.
  • a screening method for the identification of an agent that inhibits the activity of a cancer stem cell gene expression product comprising:
  • a modelling method to determine the association of an agent with a cancer stem cell gene expression product comprising:
  • the Molecular Similarity application permits comparisons between different structures, different conformations of the same structure, and different parts of the same structure.
  • Each structure is identified by a name.
  • One structure is identified as the target (i.e., the fixed structure); all remaining structures are working structures (i.e. moving structures).
  • the working structure is translated and rotated to obtain an optimum fit with the target structure.
  • the person skilled in the art may use one of several methods to screen chemical entities or fragments for their ability to associate with a target. The screening process may begin by visual inspection of the target on the computer screen, generated from a machine-readable storage medium. Selected fragments or chemical entities may then be positioned in a variety of orientations, or docked, within the binding pocket.
  • CAVEAT P. A. Bartlett et al, "CAVEAT: A Program to Facilitate the Structure- Derived Design of Biologically Active Molecules". In Molecular Recognition in Chemical and Biological Problems", Special Pub., Royal Chem. Soc, 78, pp. 182-196 (1989)).
  • CAVEAT is available from the University of California, Berkeley, California.
  • 3D Database systems such as MACCS-3D (MDL Information Systems, San Leandro, California). This is reviewed in Y. C. Martin, "3D Database Searching in Drug Design", J. Med. Chem., 35, pp. 2145-2154 (1992); and HOOK (available from Molecular Simulations, Burlington, Mass.).
  • a vaccine composition comprising a polypeptide selected from the group consisting of: i) polypeptide encoded by a nucleotide sequence as represented
  • Figure 1 a or an antigenic fragment thereof
  • polypeptide or antigenic fragment comprising an amino acid sequence wherein said sequence is modified by addition deletion or substitution of at least one amino acid residue as represented in Figures 1 b or 1 c, wherein said composition optionally includes an adjuvant and/or carrier.
  • said antigenic fragment is an extracellular domain of said polypeptide.
  • said extracellular domain comprises or consists of the amino acid sequence as represented in Figure 1 C.
  • said composition includes an adjuvant and/or carrier.
  • said adjuvant is selected from the group consisting of: cytokines selected from the group consisting of GMCSF, interferon gamma, interferon alpha, interferon beta, interleukin 12, interleukin 23, interleukin 17, interleukin 2, interleukin 1 , TGF, TNFa, and TNF3.
  • said adjuvant is a TLR agonist such as CpG oligonucleotides, flagellin, monophosphoryl lipid A, poly l:C and derivatives thereof.
  • said adjuvant is a bacterial cell wall derivative such as muramyl dipeptide (MDP) and/or trehalose dicorynomycolate (TDM).
  • MDP muramyl dipeptide
  • TDM trehalose dicorynomycolate
  • An adjuvant is a substance or procedure which augments specific immune responses to antigens by modulating the activity of immune cells.
  • adjuvants include, by example only, Freunds adjuvant, muramyl dipeptides, liposomes.
  • An adjuvant is therefore an immunomodulator.
  • a carrier is an immunogenic molecule which, when bound to a second molecule augments immune responses to the latter.
  • the term carrier is construed in the following manner.
  • a carrier is an immunogenic molecule which, when bound to a second molecule augments immune responses to the latter.
  • Such antigens contain B-cell epitopes, but no T cell epitopes.
  • the protein moiety of such a conjugate (the "carrier” protein) provides T-cell epitopes which stimulate helper T-cells that in turn stimulate antigen-specific B-cells to differentiate into plasma cells and produce antibody against the antigen.
  • a vaccine according to the invention for use in the treatment of cancer.
  • cancer refers to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth.
  • the term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness.
  • cancer includes malignancies of the various organ systems, such as those affecting, for example, lung, breast, thyroid, lymphoid, gastrointestinal, and genito-urinary tract, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumours, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • carcinoma is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary.
  • carcinosarcomas also includes carcinosarcomas, e.g., which include malignant tumours composed of carcinomatous and sarcomatous tissues.
  • An "adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
  • sarcoma is art recognized and refers to malignant tumors of mesenchymal derivation. In a preferred embodiment of the invention said cancer is a carcinoma. Preferably said carcinoma is prostate carcinoma.
  • the vaccine compositions of the invention can be administered by any conventional route, including injection, intranasal spray by inhalation of for example an aerosol or nasal drops.
  • the administration may be, for example, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, or intradermal.
  • the vaccine compositions of the invention are administered in effective amounts.
  • An "effective amount" is that amount of a vaccine composition that alone or together with further doses, produces the desired immune response.
  • the amounts of vaccine will depend, of course, on the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used sufficient to provoke immunity; that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
  • the doses of vaccine administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
  • doses of vaccine are formulated and administered in effective immunizing doses according to any standard procedure in the art.
  • Other protocols for the administration of the vaccine compositions will be known to one of ordinary skill in the art, in which the dose amount, schedule of injections, sites of injections, mode of administration and the like vary from the foregoing.
  • Administration of the vaccine compositions to mammals other than humans, is carried out under substantially the same conditions as described above.
  • a subject, as used herein, is a mammal, preferably a human, and including a non-human primate, cow, horse, pig, sheep or goat.
  • a vaccine composition according to the invention that includes at least one additional anti-cancer agent.
  • said ant-cancer agent is a chemotherapeutic agent.
  • a diagnostic or prognostic method for the detection of cancer cells isolated from a subject comprising determining the expression of NETO-2, wherein over-expression of said gene is indicative of cancer or a predisposition to cancer in said subject.
  • said method comprises:
  • said method comprises:
  • kits comprising oligonucleotide primer pairs adapted to amplify one or more nucleic acid molecules comprising the nucleotide sequences as represented in Figure 1 A.
  • kit further includes components required for polymerase chain reaction.
  • kits comprising one or more antibodies adapted to bind one or more polypeptides as represented by the amino acid sequences presented in Figure 1 B.
  • said kit further includes components required for the detection of bound antibody in an immunoassay.
  • a composition comprising or consisting essentially of the extracellular domain of NETO-2 for use in the screening of antibody phage display libraries for the identification of antagonistic NETO-2 antibodies.
  • said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
  • a method to screen a combinatorial antibody phage display library wherein said library comprises antibody variable heavy and variable light chain nucleic acids comprising the steps:
  • said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
  • Figure 1 illustrates the mRNA and amino acid sequence of NETO-2 Nucleotide sequence of NETO-2 mRNA (3653bp) based on the published sequence (Accession ID: NM_018092.3) (A). Full length 525 amino acid sequence of NETO-2 (B) and the 347 amino acid sequence of the extracellular domain (C);
  • Figure 2 illustrates differential expression of NETO-2 by microarray Differential gene expression for NETO-2 in stem cells versus committed basal in cancer versus benign cells and cancer versus benign stem cells as detected by Affymetrix microarray;
  • Figure 3 illustrates NETO-2 mRNA expression in prostate cells
  • NETO-2 mRNA expression level was determined by qRT-PCR in PNT2, P4E6 and PC3 prostate cell lines.
  • Graph shows comparison between cell lines expressed relative to PNT2. All values are the mean ⁇ standard deviation of at least three independent experiments;
  • Figure 4 illustrates Inhibition of NETO-2 using siRNA. Effects of target specific siRNA on mRNA levels of NETO-2 in PNT2, P4E6, and PC3. Graphs show comparison of untransfected (UNT) and target-specific siRNA transfected relative to a non specific control siRNA (NEG: negative, scrambled siRNA).
  • the mRNA knockdown data is an average of three independent experiments ⁇ SD, taken from the colony forming efficiency assays shown in Figure 5;
  • Figure 5 illustrates the effect of NETO-2 siRNA on colony forming efficiency
  • Colony forming efficiency was measured in PNT2 cells and PC3 transfected for 72hrs with media alone (UNT), non-specific siRNA (NEG:negative) or NETO-2 specific siRNA.
  • the graphs show the mean percentage colony forming efficiency from three independent experiments ⁇ SD;
  • Figure 6 illustrates the effect of NETO-2 siRNA on cell viability (WST assay).
  • Cell viability was measured by WST assay in PNT2 cells, P4E6 cells and PC3 cells transfected for 72hrs with non-specific siRNA (NEG:negative) or NETO-2 specific siRNA.
  • the graphs show the mean fold change from at least three independent experiments ⁇ SD;
  • Figure 7 illustrates the effect of NETO-2 siRNA on cell viability (apoptosis assay)
  • the loss of the number of viable cells i.e. cell death
  • the graphs show the mean fold change from three independent experiments ⁇ SD
  • Purified His and Fc tagged versions of the ECD (extracellular domain) of NET02 are prepared for immunisation and screening ( ⁇ 2mg each). Mice are immunised with selected NETO-2 ECD antigen. Splenic B-lymphocytes are immortalised by fusion to myeloma cells. Hybridomas plated and cloned in one step and screened for affinity, FACS binding and inhibition of cfu and proliferation in relation to cancer/cancer stem cell. Selected clones (up to 4) are expanded and mAbs tested in a mouse tumour xenograft models (PC3 cells and human prostate cancer xenograft. Lead candidate selected for humanisation
  • mice Immunisation of mice is carried out with the NET02 ECD, followed by fusion of splenocytes with a suitable immortalised cell line to form a hybridoma.
  • Anti-NET02 mAbs produced by the hybridomas undergo primary screening for antigen binding and affinity. Cloning will form part of the fusion and plating process and sequencing will confirm clonality.
  • Four to 6 hybridomas are selected based on optimal in vitro assay criteria and expanded in tissue culture medium and banked in liquid nitrogen as well as other backup clones. The final lead (and back-up) selection is based on comparative activity in the primary and secondary in vitro screening tests and antitumour efficacy against prostate tumour xenografts in immunocompromised mice with or without cytotoxic drugs.
  • ADCC antibody-dependent cellular cytotoxicity
  • NET02 mAbs in in vivo test systems alone and in combination with known chemotherapeutic agents (e.g Docetaxel)
  • xenograft models PC3 cells and human prostate cancer xenograft
  • Lead candidate selected for humanisation.
  • ADCC Antibody dependent cellular cytotoxicity
  • CDC complement dependent cytotoxicity
  • PC3 prostate cancer cell lines
  • WST water-soluble tetrazolium
  • PC3 cells In addition to human xenograft models, we routinely use several different models involving the implantation into mice of prostate cancer cells grown in culture, including PC3 cells.
  • the PC3 cell line was established from a bone metastasis of prostate cancer, it expresses NET02 and is tumourigenic in mice.
  • NET02 The PC3 cell line was established from a bone metastasis of prostate cancer, it expresses NET02 and is tumourigenic in mice.
  • NET02 NET02
  • PC3 cells transfected with anti-NET02 siRNA are incapable of initiating tumours in mice.
  • tumour response will be evaluated for tumour response as single agents and in combination with (e.g.) Docetaxel to assess synergy and to measure the effects on time to relapse.
  • Tumours are initiated by grafting selected cell phenotypes orthotopically into the prostate or under the kidney capsule and mice are then randomly assigned to treatment groups (including a placebo group). Treatment is either initiated on the day of grafting or once tumours have become established. End points are reached once tumours reach 1 .5cm and include any observed adverse effects from each therapy.
  • Tumour response will be determined by measurement of tumours when the mice are killed, as well as examination for metastatic spread. The tumours will be retrieved, measured and the fate of the stem cell population determined using our proprietary assays.
  • mice Immunisation of mice is carried out with the NET02 ECD, followed by fusion of splenocytes with a suitable immortalised cell line to form a hybridoma.
  • Anti-NET02 mAbs produced by the hybridomas undergo primary screening for antigen binding and affinity. Cloning forms part of the fusion and plating process and sequencing will confirm clonality.
  • hybridomas Four to 6 hybridomas are selected based on optimal in vitro assay criteria and expanded in tissue culture medium and banked in liquid nitrogen as well as other back-up clones.
  • Prostate cell lines were maintained under standard culture conditions in a humidified incubator at 37 ⁇ C in 5%C0 2 .
  • PNT2 cells were maintained in RPMI 1640 media (Invitrogen, Paisley, UK) with the addition of 10% foetal calf serum (FCS; PAA Laboratories Ltd. Yeovil, UK) and 1 % L-Glutamine (Invitrogen, Paisley, UK).
  • PC3 cells were maintained in HAMS F12 (Invitrogen, Paisley, UK) supplemented with 7% foetal calf serum and 1 % L-Glutamine and P4E6 cells were grown in keratinocyte serum free medium (Invitrogen, Paisley, UK) with bovine pituitary extract (BPE), epidermal growth factor (EGF), 2% FCS and 1 % L-Glutamine.
  • BPE bovine pituitary extract
  • EGF epidermal growth factor
  • FCS 1 % L-Glutamine
  • Reverse transcription was carried out on 50-500ng of fractionated cell RNA to generate cDNA.
  • Real Time PCR was carried out using the Taqman gene expression system (Applied Biosystems, Warrington, UK) according to the manufacturer's protocol with the exception that a reduced total reaction volume of 10 ⁇ was used. All reactions were carried out in triplicate in 96-well PCR plates on an ABI Prism 7300 sequence detection system (Applied Biosystems). Standard thermal cycling conditions included a hot start of 2 minutes at 50 °C, 10 minutes at 95 ⁇ C, followed by 40 cycles of: 95 ⁇ C 15 s, 60 °C for 1 minute. Data analysis was carried out using ABI SDS software and Microsoft Excel. 18S was used as endogenous control gene and for normalizing all expression values. For the measurement of RNA knockdown by siRNA differential RNA expression in response to siRNA was calculated by the ⁇ method (according to manufacturer, Applied Biosystems).
  • PNT2, P4E6 and PC3 cells were seeded at 5 * 10 4 cells per well of a 24 well plate and incubated overnight prior to transfection.
  • PNT2 and PC3 cells were transfected with Nanofectin (PAA Laboratories Ltd. Yeovil, UK) according to the manufacturer's protocols.
  • P4E6 cells were transfected with Oligofectamine (Invitrogen, Paisley, UK) according to the manufacturer's protocols. In all cases cells were incubated for 72 hours before being assayed for RNA knockdown by qRT-PCR and clonogenicity.
  • Colony forming assays in prostate epithelial cells After treatment with siRNA for 72 hours, cells were plated at 200 cells/well on 24-well plates. Medium was changed every 2-3 days and colony formation was monitored throughout. The endpoint was determined based on the observed proliferation of the negative siRNA control cells ( ⁇ 7 days for PNT2 and PC3 cells, ⁇ 10days for P4E6 cells). Colony forming efficiency (CFE) was calculated as the number of colonies >32 cells divided by the number of cells initially plated x 100.
  • CFE Colony forming efficiency
  • cell proliferation was measured using a WST assay. Briefly, cells were treated for 72 hrs with siRNA in triplicate in standard tissue culture media. The media was removed and replaced with a 1 :10 dilution of WST-1 reagent in tissue culture media according to the manufacturer's instructions (Roche, Burgess Hill, UK). Cells were subsequently incubated for four hours at 37 ⁇ C. The absorbance was read at 450nm on a Fluostar Optima plate reader (BMG Labtech). Results are expressed as relative absorbance with respect to cells transfected with nonspecific siRNA after background subtraction.
  • Apoptosis assays in prostate epithelial cells After treatment with siRNA for 72 hrs, cells were imaged and then cell death was investigated using an apoptosis assay.
  • Cells were washed in MACS buffer (2mM EDTA, 0.5% FCS, PBS) and incubated with CD133-APC antibody for 10 minutes on a circular mixer in the fridge (clone 293C3, Miltenyi Biotec, Bergisch Gladbach, Germany). The cells were rinsed with MACS buffer and incubated with a fluoroisothiocyanate (FITC) conjugate of the cell-permeable caspase inhibitor VAD-FMK (In Situ Caspace Assay, Promega, Southampton, UK) for 20 minutes at 37°C on a rotating mixer.
  • FITC fluoroisothiocyanate
  • PC3 cells were plated at 4x10 6 per 150cm 3 tissue culture flasks and left overnight to adhere. Cells were treated with either NET02 siRNA, a scrambled control siRNA, or were untransfected and left for 72 hours. Cells were trypsinised and washed, then resuspended in media and counted. Cells were centrifuged and re-suspended at 1 x10 s cells per 100 ⁇ matrigel (BD MatrigelTM Basement Membrane Matrix). Cells were administered subcutaneously into the rear flank of BALB/c Nude under anaesthetic, with ten mice per treatment group. Formation of a bulla indicated satisfactory injection.
  • NET02 siRNA a scrambled control siRNA
  • tumours were measured every two days in terms of length (L), width (W), and height (H) and their volumes calculated according to the formula LxWxHxO.5236. Animals were culled when the tumour reached a size of no more than 1 .5cm, or if the animal showed any signs of distress.
  • NETO-2 expression was measured in cell lines representing benign prostate epithelium (PNT2), early stage prostate cancer (P4E6) and advanced metastatic prostate cancer (PC3). Analysis of NETO-2 expression by qRT-PCR showed that NETO-2 is decreased (0.25 fold) ( Figure 3). Similar levels of mRNA expression of NETO-2 were observed in P4E6 cells relative to PNT2 cells.
  • Clonogenic recovery assays were carried out to determine the ability of the prostate cells treated with NETO-2 siRNA (Figure 5) to form colonies. Results are presented as percent colony forming efficiency (CFE) calculated as follows: (No. of colonies ⁇ 32 cells/no. of cells plated) X 100. Treatment with NETO-2 siRNA showed a small but significant decrease in CFE of 28% (p ⁇ 0.001 ) in PNT2 cells. Treatment of P4E6 cells with NET02 siRNA caused a significant decrease in CFE of 71 % (p ⁇ 0.001 ). Treatment of PC3 cells with NETO-2 siRNA for 72 hrs resulted in a significant decrease in CFE of 70%, respectively (p ⁇ 0.05) ( Figure 5).
  • CFE colony forming efficiency
  • Example 6 Effect of NETO-2 inhibition on viability of prostate cells as measured by an apoptosis assay
  • NETO-2 inhibition was also determined using a FACS based apoptosis assay.
  • Cell death was monitored by determining the expression of caspase proteins, which are involved in the apoptosis cell signalling pathway.
  • caspase inhibitor In addition to the caspase inhibitor, DAPI uptake was used as an indicator of compromised integrity of the plasma membrane which is a feature of necrosis.
  • NETO-2 specific siRNA treatment did not show a change in cell death for PNT2 or P4E6 cells ( Figure 7). However, there was an increase in cell death in PC3 cells after NET02 siRNA knockdown (34%, p ⁇ 0.01 )
  • mice in this group were culled by day 42 post initiation.
  • pre-treatment of PC3 cells with NET02 siRNA caused a significant decrease in the size and formation of tumours, with only 30% of mice forming small tumours, and only one mouse being culled at day 67 post initiation, having reached the maximum tumour size permissible.
  • NET02 siRNA pre-treatment of PC3 cells caused a significant increase in survival proportion compared to both untransfected and scrambled siRNA pre-treated cells, as shown by Kaplan-Meier survival curves. Median survival for untransfected, scrambled siRNA and NET02 were 28 days, 39 days and undefined respectively.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Oncology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Toxicology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Plant Pathology (AREA)
  • Gynecology & Obstetrics (AREA)

Abstract

We disclose agents that inhibit the expression of NETO-2 which has elevated expression in cancer stem cells; the use of NETO- 2 as a diagnostic or prognostic marker of tumour initiation; the use NETO-2 polypeptides in the identification of agents that inhibit activity; and including antibodies that bind NETO-2 and vaccines comprising NETO-2 polypeptides.

Description

Compositions Comprising Agents that Inhibit Neuropilin and Tolloid Like 2 Field of the Invention The disclosure relates to agents that inhibit the expression or activity of neuropilin and tolloid like 2 [NETO-2] which has elevated expression in cancer stem cells; the monitoring of expression of NETO-2 as a diagnostic or prognostic marker of tumour initiation; the use NETO-2 polypeptide in the identification of agents that inhibit activity; and including vaccines comprising NETO-2 polypeptides and antibodies that binds NETO-2.
Background to the Invention
The term "stem cell" represents a generic group of undifferentiated cells that possess the capacity for self-renewal while retaining varying potential to form differentiated cells and tissues. Stem cells can be pluripotent or multipotent. A pluripotent stem cell is a cell that has the ability to form all tissues found in an intact organism although the pluripotent stem cell cannot form an intact organism. A multipotent cell has a restricted ability to form differentiated cells and tissues. Typically adult stem cells are multipotent stem cells and are the precursor stem cells or lineage restricted stem cells that have the ability to form some cells or tissues and replenish senescing or damaged cells/tissues. Generally they cannot form all tissues found in an organism although some reports have claimed a greater potential for such 'adult' stem cells than originally thought. Evidence suggests that tumours are clonal and are therefore derived from a single cell. However, there are few studies that identify and characterize those cells types that are responsible for maintaining tumour cell growth. Some have searched for these so called "cancer stem cells". The concept of a cancer stem cell within a more differentiated tumour mass, as an aberrant form of normal differentiation, is now gaining acceptance over the current model of oncogenesis in which all tumour cells are equivalent both in growth and tumour-initiating capacity [Hamburger AW, Salmon SE: Primary bioassay of human tumor stem cells. Science 1977, 197: 461463; Pardal R, Clarke MF, Morrison SJ: Applying the principles of stem cell biology to cancer. Nat. Rev. Cancer 2003, 3: 895902.] For example, in leukaemia, the ability to initiate new tumour growth resides in a rare phenotypically distinct subset of tumour cells [Bonnet D, Dick J.E. Human acute myeloid leukaemia is organized as a hierarchy that originates from a primitive hematopoietic cell Nat. Med. 1997, 3: 730737] which are defined by the expression of CD34 and CD38 surface antigens and have been termed leukaemia stem cells.
Similar tumour-initiating cells have also been found in 'solid' cancers such as prostate [Collins AT, Berry PA, Hyde C, Stower MJ, Maitland NJ: Prospective Identification of Tumorigenic Prostate Cancer Stem Cells. Cancer Res. 2005, 65: 1094610951 ], breast [Al Hajj M, Wicha MS, BenitoHernandez A, Morrison SJ, Clarke MF: Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003, 100: 39833988], brain [Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB: Identification of human brain tumour initiating cells. Nature 2004, 432: 396401 ], lung [Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I., Vogel S, Crowley D, Bronson RT, Jacks T: Identification of bronchioalveolar stem cells in normal lung and lung cancer. Ce// 2005, 121 : 823-835] colon [O'Brien CA, Pollett A, Gallinger S, Dick JE: A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007, 445: 1061 10; RicciVitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R: Identification and expansion of human colon cancer initiating cells. Nature 2007, 445: 1 1 1 1 15]; and gastric cancers [Houghton J, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, Cai X, Fox JG, Goldenring JR, Wang TC: Gastric cancer originating from bone marrow derived cells. Science 2004, 306: 15681571 ].
This disclosure relates to the identification of NETO-2 which has enhanced expression in cancer stem cells and in particular prostate cancer stem cells and wherein expression is correlated with tumour cell initiation. In WO2005/089043 we describe the isolation of prostate stem cells which have been directly isolated from lymph node and prostate glands from a series of patient samples. These stem cells express markers that characterise the cells with stem cell properties. The following markers are typically expressed as prostate stem cell markers; human epithelial antigen (HEA), CD44, a23ihl and CD133. This disclosure identifies NETO-2 the expression of which is critical for colony formation which is the in initiating step in the formation of a tumour. We disclose that inhibition of expression of NETO-2 results in a failure to form colonies and a subsequent failure to form a tumour in vivo. The expression of NETO-2 therefore represents the early stages of tumour formation and allows an early therapeutic intervention with consequent inhibition of tumour formation. The detection of NETO-2 also serves as a diagnostic or prognostic marker of the early stages of tumour formation. NETO-2 exists as two isoforms. The full length isoform is 525 amino acids in length. A second isoform is a shorter version and is missing amino acid residues 1 -324 and has a different amino acid sequence between amino acid residues 325-333. NETO-2 is a single span transmembrane receptor and is known interact with glutamate receptor which is primarily expressed in the brain.
Statements of Invention
According to an aspect of the invention there is provided an agent that inhibits the expression of NETO-2 or the activity of NETO-2 wherein said expression/activity is enhanced in a cancer stem cell and wherein the agent inhibits tumour initiation.
Preferably NETO-2 comprises or consists of the nucleotide sequence as represented in Figure 1 A.
In a preferred embodiment of the invention said agent is an antisense oligonucleotide or RNA.
According to an aspect of the invention there is provided a pharmaceutical composition comprising an agent that inhibits tumour initiation wherein the agent is selected from the group consisting of:
i) an agent comprising one or more antisense oligonucleotide or antisense RNA molecules wherein said oligonucleotide or antisense RNA molecule comprise a nucleotide sequence adapted to anneal to a sense nucleotide sequence derived from at least one gene represented by the sense sequence presented in Figure 1 a; or
ii) an agent comprising an antibody, or active antibody binding fragment thereof, that binds to the extracellular domain of NETO 2.
According to a further aspect of the invention there is provided a composition comprising one or more antisense oligonucleotide or RNA molecules wherein said oligonucleotide or antisense RNA molecule comprise a nucleotide sequence adapted to anneal to a sense nucleotide sequence derived from at least one gene represented by the sense sequence presented in Figure 1 A. In a preferred embodiment of the invention said composition is a pharmaceutical composition. In a preferred embodiment of the invention said composition comprises, one, two, three or four antisense oligonucleotides or RNA molecules. In a preferred embodiment of the invention said composition consists essentially of one or more oligonucleotides or antisense RNA molecules and physiologically compatible excipients and/or adjuvants.
In a preferred embodiment of the invention said antisense RNA molecule is part of a siRNA or shRNA molecule.
A technique to specifically ablate gene function is through the introduction of double stranded RNA, also referred to as small inhibitory or interfering RNA (siRNA), into a cell which results in the destruction of mRNA complementary to the sequence included in the siRNA molecule. The siRNA molecule comprises two complementary strands of RNA (a sense strand and an antisense strand) annealed to each other to form a double stranded RNA molecule. The siRNA molecule is typically derived from exons of the gene which is to be ablated. The mechanism of RNA interference is being elucidated. Many organisms respond to the presence of double stranded RNA by activating a cascade that leads to the formation of siRNA. The presence of double stranded RNA activates a protein complex comprising RNase III which processes the double stranded RNA into smaller fragments (siRNAs, approximately 21 -29 nucleotides in length) which become part of a ribonucleoprotein complex. The siRNA acts as a guide for the RNase complex to cleave mRNA complementary to the antisense strand of the siRNA thereby resulting in destruction of the mRNA.
In a preferred embodiment of the invention said antisense RNA molecule is between 19 nucleotides [nt] and 29nt in length. More preferably still said antisense RNA molecule is between 21 nt and 27nt in length. Preferably said antisense RNA molecule is about 21 nt in length.
In a preferred embodiment of the invention said antisense RNA consists of 21 nt.
In an alternative preferred embodiment of the invention said siRNA is selected from the nucleotide sequences set forth in SEQ ID NO: 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52, 53, 54 or 55.
In a preferred embodiment of the invention said antisense, siRNA or shRNA includes modified nucleotides.
The term "modified" as used herein describes a nucleic acid molecule in which;
i) at least two of its nucleotides are covalently linked via a synthetic internucleoside linkage (i.e., a linkage other than a phosphodiester linkage between the 5' end of one nucleotide and the 3' end of another nucleotide). Alternatively or preferably said linkage may be the 5' end of one nucleotide linked to the 5' end of another nucleotide or the 3' end of one nucleotide with the 3' end of another nucleotide; and/or ii) a chemical group, such as cholesterol, not normally associated with nucleic acids has been covalently attached to the double stranded nucleic acid. iii) Preferred synthetic internucleoside linkages are phosphorothioates, alkylphosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, phosphate triesters, acetamidates, peptides, and carboxymethyl esters.
The term "modified" also encompasses nucleotides with a covalently modified base and/or sugar. For example, modified nucleotides include nucleotides having sugars which are covalently attached to low molecular weight organic groups other than a hydroxyl group at the 3' position and other than a phosphate group at the 5' position. Thus modified nucleotides may also include 2' substituted sugars such as 2'-0-methyl-; 2-O-alkyl; 2-O-allyl; 2'-S-alkyl; 2'-S-allyl; 2'- fluoro-; 2'-halo or 2;azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, and sedoheptulose.
Modified nucleotides are known in the art and include, by example and not by way of limitation, alkylated purines and/or pyrimidines; acylated purines and/or pyrimidines; or other heterocycles. These classes of pyrimidines and purines are known in the art and include, pseudoisocytosine; N4, N4-ethanocytosine; 8-hydroxy-N6-methyladenine; 4- acetylcytosine, 5-(carboxyhydroxylmethyl) uracil; 5-fluorouracil; 5-bromouracil;5- carboxymethylaminomethyl-2-thiouracil; 5 carboxymethylaminomethyl uracil; dihydrouracil; inosine; N6-isopentyl-adenine; l-methyladenine; 1 -methylpseudouracil; 1 - methylguanine; 2,2-dimethylguanine; 2-methyladenine; 2-methylguanine; 3- methylcytosine; 5-methylcytosine; N6-methyladenine; 7-methylguanine; 5- methylaminomethyl uracil; 5-methoxy amino methyl-2-thiouracil; β-D-mannosylqueosine; 5-methoxycarbonylmethyluracil; 5-methoxyuracil; 2 methylthio-N6-isopentenyladenine; uracil-5-oxyacetic acid methyl ester; psueouracil; 2-thiocytosine; 5-methyl-2 thiouracil, 2- thiouracil; 4-thiouracil; 5-methyluracil; N-uracil-5-oxyacetic acid methylester; uracil 5— oxyacetic acid; queosine; 2-thiocytosine; 5-propyluracil; 5-propylcytosine; 5-ethyluracil; 5-ethylcytosine; 5-butyluracil; 5-pentyluracil; 5-pentylcytosine; and 2,6,-diaminopurine; methylpsuedouracil; 1 -methylguanine; 1 -methylcytosine. Modified double stranded nucleic acids also can include base analogs such as C-5 propyne modified bases (see Wagner et al., Nature Biotechnology 14:840-844, 1996). In an alternative preferred embodiment of the invention said pharmaceutical composition includes a carrier adapted to deliver said antisense RNA to a cell or tissue.
The delivery of antisense oligonucleotide, siRNA or shRNA is achieved using delivery vehicles known in the art. For example siRNA can be chemically modified and conjugated to a lipophilic cholesterol moiety at the 3' end of the sense strand. Cationic delivery systems can also be employed in the delivery of siRNA. These include cationic lipids and liposomes, cationic polymers, cationic dendrimers and cationic cell penetrating peptides. The cationic delivery vehicles have a common positive charge which facilitates complex formation with negatively charged siRNA. Commercially available examples of liposome based delivery vehicles include Lipofectin, RNAifect, Oligofectamine, Lipofectamine and TransIT TKO have been used in vitro. DOTAP (N [1 -(2, 3- dioleoyloxy)]-N, N, N-trimethyl ammonium propane) and Oligfectamine have been utilised in vivo. Other liposome based delivery vehicle includes solid nucleic acid lipid particles [SNALPs] which are also conjugated with polyethylene glycol. Peptide delivery vehicles have also been successful in delivering siRNA. Pegylated polyethyleneimine [PEI] comprising RGD peptides have been used to target siRNA to angiogenesis factors such as VEGF. Atelocollagen has been used in the delivery of siRNA to tumours in vivo. Delivery of siRNA has also been demonstrated using cyclodextrin polymers. A yet further example of a siRNA delivery vehicle are self assembled LPD nanoparticles which have been used to deliver to solid and metastatic tumours. LPD nanoparticles comprise cationic lipids combined with protamine which interacts with negatively charged siRNA. Pegylated versions of LPD nanoparticles are also known which have improved pharmacokinetics.
In a preferred embodiment of the invention said antibody is a polyclonal antibody.
In an alternative preferred embodiment of the invention said antibody is a monoclonal antibody.
Antibodies, also known as immunoglobulins, are protein molecules which have specificity for foreign molecules (antigens). Immunoglobulins (Ig) are a class of structurally related proteins consisting of two pairs of polypeptide chains, one pair of light (L) (low molecular weight) chain (κ or λ), and one pair of heavy (H) chains (γ, α, μ, δ and ε), all four linked together by disulphide bonds. Both H and L chains have regions that contribute to the binding of antigen and that are highly variable from one Ig molecule to another. In addition, H and L chains contain regions that are non-variable or constant. The L chains consist of two domains. The carboxy-terminal domain is essentially identical among L chains of a given type and is referred to as the "constant" (C) region. The amino terminal domain varies from L chain to L chain and contributes to the binding site of the antibody. Because of its variability, it is referred to as the "variable" (V) region. The H chains of Ig molecules are of several classes, α, μ, σ, a, and γ (of which there are several sub-classes). An assembled Ig molecule consisting of one or more units of two identical H and L chains, derives its name from the H chain that it possesses. Thus, there are five Ig isotypes: IgA, IgM, IgD, IgE and IgG (with four sub-classes based on the differences in the H chains, i.e., lgG1 , lgG2, lgG3 and lgG4). Further detail regarding antibody structure and their various functions can be found in, Using Antibodies: A laboratory manual, Cold Spring Harbour Laboratory Press.
In a preferred embodiment of the invention said fragment is a single chain antibody fragment.
Various fragments of antibodies are known in the art, e.g. Fab, Fab2, F(ab')2, Fv, Fc, Fd,, etc. A Fab fragment is a multimeric protein consisting of the immunologically active portions of an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region, covalently coupled together and capable of specifically binding to an antigen. Fab fragments are generated via proteolytic cleavage (with, for example, papain) of an intact immunoglobulin molecule. A Fab2 fragment comprises two joined Fab fragments. When these two fragments are joined by the immunoglobulin hinge region, a F(ab')2 fragment results. An Fv fragment is multimeric protein consisting of the immunologically active portions of an immunoglobulin heavy chain variable region and an immunoglobulin light chain variable region covalently coupled together and capable of specifically binding to an antigen. A fragment could also be a single chain polypeptide containing only one light chain variable region, or a fragment thereof that contains the three CDRs of the light chain variable region, without an associated heavy chain variable region, or a fragment thereof containing the three CDRs of the heavy chain variable region, without an associated light chain moiety; and multi specific antibodies formed from antibody fragments, this has for example been described in US patent No 6,248,516. Fv fragments or single region (domain) fragments are typically generated by expression in host cell lines of the relevant identified regions. These and other immunoglobulin or antibody fragments are within the scope of the invention and are described in standard immunology textbooks such as Paul, Fundamental Immunology or Janeway's Immunobiology, Murphy, K., Travers, P. & Walport P.
Molecular biology now allows direct synthesis (via expression in cells or chemically) of these fragments, as well as synthesis of combinations thereof. A fragment of an antibody or immunoglobulin can also have bispecific function as described above.
In a preferred embodiment of the invention said antibody is a chimeric antibody.
In an alternative preferred embodiment of the invention said antibody is a humanized or human antibody.
Chimeric antibodies are recombinant antibodies in which all of the V-regions of a mouse or rat antibody are combined with human antibody C-regions. Humanised antibodies are recombinant hybrid antibodies which fuse the complementarity determining regions from a rodent antibody V-region with the framework regions from the human antibody V- regions. The C-regions from the human antibody are also used. The complementarity determining regions (CDRs) are the regions within the N-terminal domain of both the heavy and light chain of the antibody to where the majority of the variation of the V- region is restricted. These regions form loops at the surface of the antibody molecule. These loops provide the binding surface between the antibody and antigen. Antibodies from non-human animals provoke an immune response to the foreign antibody and its removal from the circulation. Both chimeric and humanised antibodies have reduced antigenicity when injected to a human subject because there is a reduced amount of rodent (i.e. foreign) antibody within the recombinant hybrid antibody, while the human antibody regions do not ellicit an immune response. This results in a weaker immune response and a decrease in the clearance of the antibody. This is clearly desirable when using therapeutic antibodies in the treatment of human diseases. Humanised antibodies are designed to have less "foreign" antibody regions and are therefore thought to be less immunogenic than chimeric antibodies.
Reviews of current delivery vehicles can be found in Molecular Pharmaceutics 2008 Vol 6[3] p651 -658; The AAPS Journal 2009 Vol 1 1 [4] p639; Pharmaceutical Research 2009, Vol 26[3] p657; and Nature Reviews 2009 Vol 8, p129.
When administered the compositions of the present invention are administered in pharmaceutically acceptable preparations. Such preparations may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers and supplementary anti-cancer agents.
The compositions of the invention can be administered by any conventional route, including injection or by gradual infusion over time. Treatment may be topical or systemic. The administration may, for example, be oral, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, transdermal, transepithelial or intra bone marrow administration or by direct injection into the tumour mass.
The compositions of the invention are administered in effective amounts. An "effective amount" is that amount of a composition that alone, or together with further doses, produces the desired response. In the case of treating a particular disease, such as cancer, the desired response is inhibiting the progression of the disease. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods.
Such amounts will depend, of course, on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
The pharmaceutical compositions used in the foregoing methods preferably are sterile and contain an effective amount of an agent according to the invention for producing the desired response in a unit of weight or volume suitable for administration to a patient. The doses of the antisense RNA according to the invention administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. Other factors include the desired period of treatment. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
In general, doses of antisense RNA [e.g., siRNA] of between 1 nM - 1 μΜ generally will be formulated and administered according to standard procedures. Preferably doses can range from 1 nM-500nM, 5nM-200nM, and 10nM-100nM. Other protocols for the administration of compositions will be known to one of ordinary skill in the art, in which the dose amount, schedule of injections, sites of injections, mode of administration and the like vary from the foregoing. The administration of compositions to mammals other than humans, (e.g. for testing purposes or veterinary therapeutic purposes), is carried out under substantially the same conditions as described above. A subject, as used herein, is a mammal, preferably a human, and including a non-human primate, cow, horse, pig, sheep, goat, dog, cat or rodent.
When administered, the pharmaceutical preparations of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptable compositions. The term "pharmaceutically acceptable" means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients. Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents' (e.g. anti-inflammatory agents such as steroids, non-steroidal anti-inflammatory agents, chemotherapeutic agents). When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-acceptable salts thereof and are not excluded from the scope of the invention. Such pharmacologically and pharmaceutically-acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic, and the like. Also, pharmaceutically- acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
Compositions may be combined, if desired, with a pharmaceutically-acceptable carrier. The term "pharmaceutically-acceptable carrier" as used herein means one or more compatible solid or liquid fillers, diluents or encapsulating substances which are suitable for administration into a human. The term "carrier" in this context denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application, (e.g. liposome or immuno-liposome). The components of the pharmaceutical compositions also are capable of being co-mingled with the molecules of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficacy.
The pharmaceutical compositions may contain suitable buffering agents, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt. The pharmaceutical compositions also may contain, optionally, suitable preservatives, such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
The pharmaceutical compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active compound. Other compositions include suspensions in aqueous liquids or non-aqueous liquids such as syrup, elixir or an emulsion or as a gel. Compositions may be administered as aerosols and inhaled.
Compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of agent, which is preferably isotonic with the blood of the recipient. This preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation also may be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example, as a solution in 1 , 3-butane diol. Among the acceptable solvents that may be employed are water, Ringer's solution , and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono-or di-glycerides. In addition, fatty acids such as oleic acid may be used in the preparation of injectables. Carrier formulation suitable for oral, subcutaneous, intravenous, intramuscular, etc. administrations can be found in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, PA. In a preferred embodiment of the invention said composition includes an additional chemotherapeutic agent.
A general definition of "chemotherapeutic agent" is an agent that typical is a small chemical compound that kills cells in particular diseased cells, for example cancer cells. The agents can be divided with respect to their structure or mode of action. For example, chemotherapeutic agents include alkylating agents, anti-metabolites, anthracyclines, alkaloids, plant terpenoids and toposisomerase inhibitors. Chemotherapeutic agents typically produce their effects on cell division or DNA synthesis. In a preferred embodiment of the invention said chemotherapeutic agent is an alkylating agent.
Preferably said alkylating agent is selected from the group consisting of: cisplatin, carboplatin or oxaliplatin. In a preferred embodiment of the invention said chemotherapuetic agent is an anti- metabolic drug.
In a preferred embodiment of the invention said drug is a purine analogue. In an alternative preferred embodiment of the invention said drug is a pyrimidine analogue.
Purine analogues are known in the art; for example thioguanine is used to treat acute leukaemia; fludarabine inhibits the function of DNA polymerases, DNA primases and DNA ligases and is specific for cell-cycle S-phase; pentostatin and cladribine are adenosine analogues and are effective against hairy cell leukaemias. A further example is mecrcaptopurine which is an adenine analogue. Pyrimidine analogues are similarly known in the art. For example, 5-fluorouracil (5-FU), floxuridine and cytosine arabinoside. 5-FU has been used for many years in the treatment of breast, colorectal cancer, pancreatic and other cancers. 5-FU can also been formed from the pro-drug capecitabine which is converted to 5-FU in the tumour.
In a preferred embodiment of the invention said chemotherapeutic agent is 5- fluorouracil. In a preferred embodiment of the invention said anti-metabolic drug is administered with leucovorin.
Leucovorin, also known as folinic acid, is administered as an adjuvant in cancer chemotherapy and which enhances the inhibitory effects of 5-FU on thymidylate synthase.
In a further preferred embodiment of the invention said chemotherapeutic agent is an alkaloid; preferably said alkaloid is a vinca alkaloid, for example vincristine or vinblastine. In a yet further preferred embodiment of the invention said chemotherapeutic agent is a terpenoid; preferably a taxane e.g. palitaxel.
According to an aspect of the invention there is provided the use of a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence as represented in Figure 1 a for the identification of agents that modulate the activity of said polypeptide. According to an aspect of the invention there is provided a screening method for the identification of an agent that inhibits the activity of a cancer stem cell gene expression product comprising:
i) providing a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence as represented in Figure 1 a;
ii) providing at least one candidate agent to be tested;
iii) forming a preparation that is a combination of (i) and (ii) above; and iv) testing the effect of said agent on the activity of said polypeptide. According to a further aspect of the invention there is provided a modelling method to determine the association of an agent with a cancer stem cell gene expression product comprising:
i) providing computational means to perform a fitting operation between an agent and a polypeptide comprising or consisting of the amino acid sequence in Figure 1 b or 1 c; and
ii) analysing the results of said fitting operation to quantify the association between the agent and the polypeptide.
The rational design of binding entities for proteins is known in the art and there are a large number of computer programs that can be utilised in the modelling of 3- dimensional protein structures to determine the binding of chemical entities to functional regions of proteins and also to determine the effects of mutation on protein structure. This may be applied to binding entities and also to the binding sites for such entities. The computational design of proteins and/or protein ligands demands various computational analyses which are necessary to determine whether a molecule is sufficiently similar to the target protein or polypeptide. Such analyses may be carried out in current software applications, such as the Molecular Similarity application of QUANTA (Molecular Simulations Inc., Waltham, Mass.) version 3.3, and as described in the accompanying User's Guide, Volume 3 pages. 134-135. The Molecular Similarity application permits comparisons between different structures, different conformations of the same structure, and different parts of the same structure. Each structure is identified by a name. One structure is identified as the target (i.e., the fixed structure); all remaining structures are working structures (i.e. moving structures). When a rigid fitting method is used, the working structure is translated and rotated to obtain an optimum fit with the target structure. The person skilled in the art may use one of several methods to screen chemical entities or fragments for their ability to associate with a target. The screening process may begin by visual inspection of the target on the computer screen, generated from a machine-readable storage medium. Selected fragments or chemical entities may then be positioned in a variety of orientations, or docked, within the binding pocket.
Useful programs to aid the person skilled in the art in connecting the individual chemical entities or fragments include: CAVEAT (P. A. Bartlett et al, "CAVEAT: A Program to Facilitate the Structure- Derived Design of Biologically Active Molecules". In Molecular Recognition in Chemical and Biological Problems", Special Pub., Royal Chem. Soc, 78, pp. 182-196 (1989)). CAVEAT is available from the University of California, Berkeley, California. 3D Database systems such as MACCS-3D (MDL Information Systems, San Leandro, California). This is reviewed in Y. C. Martin, "3D Database Searching in Drug Design", J. Med. Chem., 35, pp. 2145-2154 (1992); and HOOK (available from Molecular Simulations, Burlington, Mass.).
Once the agent has been optimally selected or designed, as described above, substitutions may then be made in some of its atoms or side groups in order to improve or modify its binding properties. Generally, initial substitutions are conservative, i.e., the replacement group will have approximately the same size, shape, hydrophobicity and charge as the original group. The computational analysis and design of molecules, as well as software and computer systems are described in US Patent No 5,978,740 which is included herein by reference. According to a further aspect of the invention there is provided a vaccine composition comprising a polypeptide selected from the group consisting of: i) polypeptide encoded by a nucleotide sequence as represented
Figure 1 a, or an antigenic fragment thereof;
a polypeptide encoded by a nucleotide sequence wherein said sequence is degenerate as a result of the genetic code to the nucleotide sequence defined in (i);
a polypeptide or antigenic fragment comprising an amino acid sequence wherein said sequence is modified by addition deletion or substitution of at least one amino acid residue as represented in Figures 1 b or 1 c, wherein said composition optionally includes an adjuvant and/or carrier. In a preferred embodiment of the invention said antigenic fragment is an extracellular domain of said polypeptide. In a preferred embodiment of the invention said extracellular domain comprises or consists of the amino acid sequence as represented in Figure 1 C.
In a preferred embodiment of the invention said composition includes an adjuvant and/or carrier.
In a preferred embodiment of the invention said adjuvant is selected from the group consisting of: cytokines selected from the group consisting of GMCSF, interferon gamma, interferon alpha, interferon beta, interleukin 12, interleukin 23, interleukin 17, interleukin 2, interleukin 1 , TGF, TNFa, and TNF3. In a further alternative embodiment of the invention said adjuvant is a TLR agonist such as CpG oligonucleotides, flagellin, monophosphoryl lipid A, poly l:C and derivatives thereof.
In a preferred embodiment of the invention said adjuvant is a bacterial cell wall derivative such as muramyl dipeptide (MDP) and/or trehalose dicorynomycolate (TDM).
An adjuvant is a substance or procedure which augments specific immune responses to antigens by modulating the activity of immune cells. Examples of adjuvants include, by example only, Freunds adjuvant, muramyl dipeptides, liposomes. An adjuvant is therefore an immunomodulator. A carrier is an immunogenic molecule which, when bound to a second molecule augments immune responses to the latter. The term carrier is construed in the following manner. A carrier is an immunogenic molecule which, when bound to a second molecule augments immune responses to the latter. Some antigens are not intrinsically immunogenic yet may be capable of generating antibody responses when associated with a foreign protein molecule such as keyhole-limpet haemocyanin or tetanus toxoid. Such antigens contain B-cell epitopes, but no T cell epitopes. The protein moiety of such a conjugate (the "carrier" protein) provides T-cell epitopes which stimulate helper T-cells that in turn stimulate antigen-specific B-cells to differentiate into plasma cells and produce antibody against the antigen. According to a further aspect of the invention there is provided a vaccine according to the invention for use in the treatment of cancer.
As used herein, the term "cancer" refers to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth. The term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. The term "cancer" includes malignancies of the various organ systems, such as those affecting, for example, lung, breast, thyroid, lymphoid, gastrointestinal, and genito-urinary tract, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumours, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. The term "carcinoma" is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term "carcinoma" also includes carcinosarcomas, e.g., which include malignant tumours composed of carcinomatous and sarcomatous tissues. An "adenocarcinoma" refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures. The term "sarcoma" is art recognized and refers to malignant tumors of mesenchymal derivation. In a preferred embodiment of the invention said cancer is a carcinoma. Preferably said carcinoma is prostate carcinoma.
The vaccine compositions of the invention can be administered by any conventional route, including injection, intranasal spray by inhalation of for example an aerosol or nasal drops. The administration may be, for example, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, or intradermal. The vaccine compositions of the invention are administered in effective amounts. An "effective amount" is that amount of a vaccine composition that alone or together with further doses, produces the desired immune response.
The amounts of vaccine will depend, of course, on the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used sufficient to provoke immunity; that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
The doses of vaccine administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
In general, doses of vaccine are formulated and administered in effective immunizing doses according to any standard procedure in the art. Other protocols for the administration of the vaccine compositions will be known to one of ordinary skill in the art, in which the dose amount, schedule of injections, sites of injections, mode of administration and the like vary from the foregoing. Administration of the vaccine compositions to mammals other than humans, (e.g. for testing purposes or veterinary therapeutic purposes), is carried out under substantially the same conditions as described above. A subject, as used herein, is a mammal, preferably a human, and including a non-human primate, cow, horse, pig, sheep or goat.
In a preferred embodiment of the invention there is provided a vaccine composition according to the invention that includes at least one additional anti-cancer agent. Preferably said ant-cancer agent is a chemotherapeutic agent.
According to an aspect of the invention there is provided a diagnostic or prognostic method for the detection of cancer cells isolated from a subject comprising determining the expression of NETO-2, wherein over-expression of said gene is indicative of cancer or a predisposition to cancer in said subject. In a preferred method of the invention said method comprises:
i) providing an isolated biological sample to be tested;
ii) forming a preparation comprising said sample and an oligonucleotide primer pair adapted to anneal to a nucleic acid molecule comprising a nucleic acid sequence as represented in Figure 1 a; a thermostable DNA polymerase, deoxynucleotide triphosphates and co-factors; iii) providing polymerase chain reaction conditions sufficient to amplify said nucleic acid molecule;
iv) analysing the amplified products of said polymerase chain reaction for the presence or absence of a nucleic acid molecule comprising a nucleotide sequence derived from Figure 1 a; and optionally
v) comparing the amplified product with a normal matched control.
In an alternative preferred method of the invention said method comprises:
i) providing an isolated biological sample to be tested; ii) forming a preparation comprising said sample and an antibody or antibodies that specifically binds one or more polypeptide^] in said sample as represented by the amino acid sequences presented in Figures 1 b to form an antibody/polypeptide complex; iii) detecting the complex or complexes so formed; and iv) comparing the expression of said polypeptide^] with a normal matched control.
According to a further aspect of the invention there is provided a kit comprising oligonucleotide primer pairs adapted to amplify one or more nucleic acid molecules comprising the nucleotide sequences as represented in Figure 1 A.
In a preferred embodiment of the invention said kit further includes components required for polymerase chain reaction.
According to a further aspect of the invention there is provided a kit comprising one or more antibodies adapted to bind one or more polypeptides as represented by the amino acid sequences presented in Figure 1 B. In a preferred embodiment of the invention said kit further includes components required for the detection of bound antibody in an immunoassay. According to an aspect of the invention there is provided a composition comprising or consisting essentially of the extracellular domain of NETO-2 for use in the screening of antibody phage display libraries for the identification of antagonistic NETO-2 antibodies.
In a preferred embodiment of the invention said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
According to an aspect of the invention there is provided a method to screen a combinatorial antibody phage display library wherein said library comprises antibody variable heavy and variable light chain nucleic acids comprising the steps:
i) contacting a phage display library expressing said antibody variable heavy and light chains with a composition comprising or consisting essentially of the NETO-2 extracellular domain;
ii) selecting phage that specifically bind the NETO-2 extracellular domain contained in said composition;
iii) repeating i) and ii) above to identify phage clones that bind said NETO-2 extracellular domain until high affinity binders are selected; and optionally iv) isolating and storing said phage.
In a preferred method of the invention said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
Throughout the description and claims of this specification, the words "comprise" and "contain" and variations of the words, for example "comprising" and "comprises", means "including but not limited to", and is not intended to (and does not) exclude other moieties, additives, components, integers or steps.
Throughout the description and claims of this specification, the singular encompasses the plural unless the context otherwise requires. In particular, where the indefinite article is used, the specification is to be understood as contemplating plurality as well as singularity, unless the context requires otherwise.
Features, integers, characteristics, compounds, chemical moieties or groups described in conjunction with a particular aspect, embodiment or example of the invention are to be understo to be applicable to any other aspect, embodiment or example described herein unless incompatible therewith.
An embodiment of the invention will now be described by example only and with reference to the following figures:
Figure 1 illustrates the mRNA and amino acid sequence of NETO-2 Nucleotide sequence of NETO-2 mRNA (3653bp) based on the published sequence (Accession ID: NM_018092.3) (A). Full length 525 amino acid sequence of NETO-2 (B) and the 347 amino acid sequence of the extracellular domain (C);
Figure 2 illustrates differential expression of NETO-2 by microarray Differential gene expression for NETO-2 in stem cells versus committed basal in cancer versus benign cells and cancer versus benign stem cells as detected by Affymetrix microarray;
Figure 3 illustrates NETO-2 mRNA expression in prostate cells; NETO-2 mRNA expression level was determined by qRT-PCR in PNT2, P4E6 and PC3 prostate cell lines. Graph shows comparison between cell lines expressed relative to PNT2. All values are the mean ± standard deviation of at least three independent experiments;
Figure 4 illustrates Inhibition of NETO-2 using siRNA. Effects of target specific siRNA on mRNA levels of NETO-2 in PNT2, P4E6, and PC3. Graphs show comparison of untransfected (UNT) and target-specific siRNA transfected relative to a non specific control siRNA (NEG: negative, scrambled siRNA). The mRNA knockdown data is an average of three independent experiments ± SD, taken from the colony forming efficiency assays shown in Figure 5;
Figure 5 illustrates the effect of NETO-2 siRNA on colony forming efficiency Colony forming efficiency was measured in PNT2 cells and PC3 transfected for 72hrs with media alone (UNT), non-specific siRNA (NEG:negative) or NETO-2 specific siRNA. The graphs show the mean percentage colony forming efficiency from three independent experiments ± SD;
Figure 6 illustrates the effect of NETO-2 siRNA on cell viability (WST assay). Cell viability was measured by WST assay in PNT2 cells, P4E6 cells and PC3 cells transfected for 72hrs with non-specific siRNA (NEG:negative) or NETO-2 specific siRNA. The graphs show the mean fold change from at least three independent experiments ± SD;
Figure 7 illustrates the effect of NETO-2 siRNA on cell viability (apoptosis assay) The loss of the number of viable cells (i.e. cell death) was measured by a caspase apoptosis assay in PNT2 cells, P4E6 cells and PC3 cells transfected for 72hrs with non-specific siRNA (Neg: negative) or NETO-2 specific siRNA. The graphs show the mean fold change from three independent experiments ± SD; and Figure 8 Effect of NETO-2 on in vivo tumour formation and survival rates Tumour volume was measured using digital calipers every two days (A) and tumour incidence (B) and survival proportions (C) were calculated. Graphs show the mean tumour volume ± SEM. (n=10 mice per group). Materials and Methods
Generation of NETO-2 Antigen for Antibody Production
Purified His and Fc tagged versions of the ECD (extracellular domain) of NET02 are prepared for immunisation and screening (~2mg each). Mice are immunised with selected NETO-2 ECD antigen. Splenic B-lymphocytes are immortalised by fusion to myeloma cells. Hybridomas plated and cloned in one step and screened for affinity, FACS binding and inhibition of cfu and proliferation in relation to cancer/cancer stem cell. Selected clones (up to 4) are expanded and mAbs tested in a mouse tumour xenograft models (PC3 cells and human prostate cancer xenograft. Lead candidate selected for humanisation
Selection and Characterisation of mAbs to NET02
Immunisation of mice is carried out with the NET02 ECD, followed by fusion of splenocytes with a suitable immortalised cell line to form a hybridoma.Anti-NET02 mAbs produced by the hybridomas undergo primary screening for antigen binding and affinity. Cloning will form part of the fusion and plating process and sequencing will confirm clonality. Four to 6 hybridomas are selected based on optimal in vitro assay criteria and expanded in tissue culture medium and banked in liquid nitrogen as well as other backup clones. The final lead (and back-up) selection is based on comparative activity in the primary and secondary in vitro screening tests and antitumour efficacy against prostate tumour xenografts in immunocompromised mice with or without cytotoxic drugs. CANDIDATE IDENTIFICATION AND HUMANISATION
Generation of monoclonal antibodies is well known in the art using established techniques, for example see Antibodies: A Laboratory Manual Ed Harlow, David P Lane 1988 Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.
1 . Generate mAbs that specifically bind to NET02
2. Select mAbs on the basis of their ability to inhibit the clonogenic recovery and/or proliferation of PC3 cells and primary cells in vitro
3. Evaluate antibody-dependent cellular cytotoxicity (ADCC) and
complement mediated cytotoxicity (CDC) enhancement on the activity of anti-NET02 mAbs in in vitro test systems
4. Evaluate apoptosis/cell killing/cell proliferation activity of the anti-
NET02 mAbs in in vivo test systems, alone and in combination with known chemotherapeutic agents (e.g Docetaxel)
5. Identify and characterise a lead hybridoma clone for humanisation. The key steps in the lead identification process are summarised as follows:
1 . Mice immunised with selected antigen
2. Splenic B-lymphocytes immortalised by fusion to myeloma cells
3. Hybridomas plated and cloned in one step and screened for affinity, FACS binding and inhibition of cfu and proliferation
4. Best clones (up to 4) expanded and mAbs tested in a mouse tumour
xenograft models (PC3 cells and human prostate cancer xenograft) 5. Lead candidate (with back-up) selected for humanisation.
Establishment of in vitro Screening Tests The following in vitro tests are used to screen and characterise mAb candidates:
Primary Screen
1 . Human NET02 antigen ELISA
Secondary Screens
2. NHP NET02 and human NET01 binding by ELISA 3 Surface plasmon resonance binding (BIAcore ; NET02 binding kinetics and epitope mapping)
4 rface binding by fluorescence-activated cell sorting (FACS)
5 Antibody dependent cellular cytotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC), as appropriate
6 Cell fate 1 . The effect of the mAb on colony forming (cfu) efficiency of PC3 and primary cells.
Cell fate 2. The effects of the mAb candidates on cell fate in vitro will determined in prostate cancer cell lines (PC3) by water-soluble tetrazolium (WST) assay and by assay for apoptosis.
8. Cell fate 3. The effects of the mAb candidates on cell fate in prostate cancer stem cells in vitro will be measured by apoptosis assays based on flow cytometry using stem cell markers (e.g. CD133) in conjunction with apoptosis markers such as caspases. This will allow the measurement of apoptosis in stem and non-stem cell populations simultaneously.
Establishment of in vivo Screening Tests
In addition to human xenograft models, we routinely use several different models involving the implantation into mice of prostate cancer cells grown in culture, including PC3 cells. The PC3 cell line was established from a bone metastasis of prostate cancer, it expresses NET02 and is tumourigenic in mice. We have shown that inhibition of NET02 by siRNA in these cells in vitro leads to a loss of colony forming potential. We have also shown that PC3 cells transfected with anti-NET02 siRNA are incapable of initiating tumours in mice. Thus we believe this makes the model a suitable preliminary vehicle for in vivo analysis of the effects of mAb candidates prior to moving into the primary xenograft models.
Proposed experiments using the PC3 cell model
Two types of experiment to show the effect of mAbs against NET02 in an animal model • Inject PC3 cells subcutaneously into athymic nude mice, and inject the mAb at t=0 and monitor for growth of tumour relative to a control group treated with a non-specific mAb. Five different doses of antibody will be used, and animals will be sacrificed when tumours reach 1 .5cm (approx t=6 weeks). A variant of this experiment will be to pre-treat the PC3 cells with the mAb before their injection into the mice and monitor for tumour take relative to the control group.
· Generate established tumours (approximately 2 weeks after injection with PC3 cells) and then to treat with different doses of the mAb as above, monitoring for regression of the tumour or a slower rate of growth relative to the control group.
Xenografts
We routinely engraft tumour tissue biopsies from patients undergoing radical prostatectomy for prostate cancer. As the biopsies are small (~2mm) we cannot sort for rare cell populations directly from these samples, and tissue is therefore grafted into immunocompromised mice - either under the kidney capsule or subcutaneously. We have a colony of Rag-2 (-/-) gamma C (-/-) male mice for this purpose because the gamma C knockout renders the mice more susceptible to human tumour engraftment [see also WO2012/101904 which is incorporated by reference in its entirety. This first step also has the advantage of allowing us to select for malignant epithelium relative to normal. The mAbs will be evaluated for tumour response as single agents and in combination with (e.g.) Docetaxel to assess synergy and to measure the effects on time to relapse. Tumours are initiated by grafting selected cell phenotypes orthotopically into the prostate or under the kidney capsule and mice are then randomly assigned to treatment groups (including a placebo group). Treatment is either initiated on the day of grafting or once tumours have become established. End points are reached once tumours reach 1 .5cm and include any observed adverse effects from each therapy. Tumour response will be determined by measurement of tumours when the mice are killed, as well as examination for metastatic spread. The tumours will be retrieved, measured and the fate of the stem cell population determined using our proprietary assays.
These tests will be used to confirm the activity of lead candidates and will also form part of the Non-Clinical Pharmacology package for regulatory submission for the development candidate. Based on positive results of cell fate experiments in PC3 cells in vitro, testing of the candidates in the PC3 animal model will be carried out. Testing of the candidates in primary human cells in vitro will be performed in parallel with this first round of in vivo studies, and will be completed prior to studying the effects in vivo with primary human tumour xenografts.
Selection and Characterisation of mAbs to NET02
Immunisation of mice is carried out with the NET02 ECD, followed by fusion of splenocytes with a suitable immortalised cell line to form a hybridoma. Anti-NET02 mAbs produced by the hybridomas undergo primary screening for antigen binding and affinity. Cloning forms part of the fusion and plating process and sequencing will confirm clonality.
Four to 6 hybridomas are selected based on optimal in vitro assay criteria and expanded in tissue culture medium and banked in liquid nitrogen as well as other back-up clones.
Cell Culture
Prostate cell lines were maintained under standard culture conditions in a humidified incubator at 37<C in 5%C02. PNT2 cells were maintained in RPMI 1640 media (Invitrogen, Paisley, UK) with the addition of 10% foetal calf serum (FCS; PAA Laboratories Ltd. Yeovil, UK) and 1 % L-Glutamine (Invitrogen, Paisley, UK). PC3 cells were maintained in HAMS F12 (Invitrogen, Paisley, UK) supplemented with 7% foetal calf serum and 1 % L-Glutamine and P4E6 cells were grown in keratinocyte serum free medium (Invitrogen, Paisley, UK) with bovine pituitary extract (BPE), epidermal growth factor (EGF), 2% FCS and 1 % L-Glutamine. qRT-PCR
Reverse transcription was carried out on 50-500ng of fractionated cell RNA to generate cDNA. Real Time PCR was carried out using the Taqman gene expression system (Applied Biosystems, Warrington, UK) according to the manufacturer's protocol with the exception that a reduced total reaction volume of 10μΙ was used. All reactions were carried out in triplicate in 96-well PCR plates on an ABI Prism 7300 sequence detection system (Applied Biosystems). Standard thermal cycling conditions included a hot start of 2 minutes at 50 °C, 10 minutes at 95 <C, followed by 40 cycles of: 95 <C 15 s, 60 °C for 1 minute. Data analysis was carried out using ABI SDS software and Microsoft Excel. 18S was used as endogenous control gene and for normalizing all expression values. For the measurement of RNA knockdown by siRNA differential RNA expression in response to siRNA was calculated by the ΔΔΟί method (according to manufacturer, Applied Biosystems).
Transfection of prostate epithelial cells with siRNA
PNT2, P4E6 and PC3 cells were seeded at 5*104 cells per well of a 24 well plate and incubated overnight prior to transfection. PNT2 and PC3 cells were transfected with Nanofectin (PAA Laboratories Ltd. Yeovil, UK) according to the manufacturer's protocols. P4E6 cells were transfected with Oligofectamine (Invitrogen, Paisley, UK) according to the manufacturer's protocols. In all cases cells were incubated for 72 hours before being assayed for RNA knockdown by qRT-PCR and clonogenicity.
Colony forming assays in prostate epithelial cells After treatment with siRNA for 72 hours, cells were plated at 200 cells/well on 24-well plates. Medium was changed every 2-3 days and colony formation was monitored throughout. The endpoint was determined based on the observed proliferation of the negative siRNA control cells (~7 days for PNT2 and PC3 cells, ~10days for P4E6 cells). Colony forming efficiency (CFE) was calculated as the number of colonies >32 cells divided by the number of cells initially plated x 100.
Cell proliferation (WST) assays in prostate epithelial cells
After treatment with siRNA for 72 hrs, cell proliferation was measured using a WST assay. Briefly, cells were treated for 72 hrs with siRNA in triplicate in standard tissue culture media. The media was removed and replaced with a 1 :10 dilution of WST-1 reagent in tissue culture media according to the manufacturer's instructions (Roche, Burgess Hill, UK). Cells were subsequently incubated for four hours at 37<C. The absorbance was read at 450nm on a Fluostar Optima plate reader (BMG Labtech). Results are expressed as relative absorbance with respect to cells transfected with nonspecific siRNA after background subtraction.
Apoptosis assays in prostate epithelial cells After treatment with siRNA for 72 hrs, cells were imaged and then cell death was investigated using an apoptosis assay. Cells were washed in MACS buffer (2mM EDTA, 0.5% FCS, PBS) and incubated with CD133-APC antibody for 10 minutes on a circular mixer in the fridge (clone 293C3, Miltenyi Biotec, Bergisch Gladbach, Germany). The cells were rinsed with MACS buffer and incubated with a fluoroisothiocyanate (FITC) conjugate of the cell-permeable caspase inhibitor VAD-FMK (In Situ Caspace Assay, Promega, Southampton, UK) for 20 minutes at 37°C on a rotating mixer. Finally, cells were washed and resuspended in PBS buffer (0.01 M phosphate buffer, 0.0027 M potassium chloride and 0.137 M sodium chloride, pH 7.4) containing DAPI at 1 :10,000 concentration for 10 minutes prior to analysis by flow cytometry. Data was collected using a DakoCytomation CyAn ADP instrument (Dako UK Ltd, Cambridgeshire, UK). FACS results were analysed with Summit Software, v4.3 (Dako UK Ltd, Cambridgeshire, UK).
In vivo Animal Model PC3 cells were plated at 4x106 per 150cm3 tissue culture flasks and left overnight to adhere. Cells were treated with either NET02 siRNA, a scrambled control siRNA, or were untransfected and left for 72 hours. Cells were trypsinised and washed, then resuspended in media and counted. Cells were centrifuged and re-suspended at 1 x10s cells per 100μΙ matrigel (BD Matrigel™ Basement Membrane Matrix). Cells were administered subcutaneously into the rear flank of BALB/c Nude under anaesthetic, with ten mice per treatment group. Formation of a bulla indicated satisfactory injection. Thereafter, tumours were measured every two days in terms of length (L), width (W), and height (H) and their volumes calculated according to the formula LxWxHxO.5236. Animals were culled when the tumour reached a size of no more than 1 .5cm, or if the animal showed any signs of distress.
Example 1
Identification of NETO-2 as a target
The generation of a cancer stem cell gene expression signature using whole genome microarray analysis has been reported previously (Birnie et al, 2008). When gene expression profiles from stem cells and committed basal cells isolated from primary cultures of benign and malignant prostate epithelial cells were compared NET02 showed an increase in gene expression in stem cells relative to committed basal cells (1 .14 fold) (Figure 2).NET02 was also upregulated in cancer versus benign samples (all cells: 1 .39) as well as cancer stem cells relative to benign stem cells (1 .64 fold) (Figure 2). Example 2
Expression of NETO-2 in prostate cells NETO-2 expression was measured in cell lines representing benign prostate epithelium (PNT2), early stage prostate cancer (P4E6) and advanced metastatic prostate cancer (PC3). Analysis of NETO-2 expression by qRT-PCR showed that NETO-2 is decreased (0.25 fold) (Figure 3). Similar levels of mRNA expression of NETO-2 were observed in P4E6 cells relative to PNT2 cells.
Example 3 Inhibition of NET02 expression using siRNA
Having demonstrated that NETO-2 is expressed in prostate cells, siRNA was used to inhibit the expression in order to investigate the effects on cell fate. Transfection of a NETO-2 specific siRNA reduced NETO-2 mRNA expression by an average of 69% (n=3) in PNT2 cells (Figure 4), by an average of 69% (n=3) in P4E6 cells, and by an average of 89% (n=3) in PC3 cells, relative to a non-specific control siRNA.
Example 4
Effect of NETO-2 inhibition on clonogenicity of prostate cells
Clonogenic recovery assays were carried out to determine the ability of the prostate cells treated with NETO-2 siRNA (Figure 5) to form colonies. Results are presented as percent colony forming efficiency (CFE) calculated as follows: (No. of colonies ≥32 cells/no. of cells plated) X 100. Treatment with NETO-2 siRNA showed a small but significant decrease in CFE of 28% (p<0.001 ) in PNT2 cells. Treatment of P4E6 cells with NET02 siRNA caused a significant decrease in CFE of 71 % (p<0.001 ). Treatment of PC3 cells with NETO-2 siRNA for 72 hrs resulted in a significant decrease in CFE of 70%, respectively (p<0.05) (Figure 5). Example 5
Effect of NETO-2 inhibition on viability of prostate cells as measured by a WST assay
The effect of NETO-2 inhibition on cell viability was determined using a WST assay. This assay is based on the cleavage of a tetrazolium salt that is added to the culture medium and is irreversibly cleaved by metabolically active cells, releasing a product that can measured on a UV-Vis spectrophotometer. After 72 hrs transfection with non-specific siRNA (NEG) or siRNA specific for NETO-2 of primary prostate epithelial cells, the cell viability was determined. Inhibition of NETO-2 mRNA expression showed decreased cell viability for all three cell lines (loss of cell viability: 45%, 35%, and 29% for PNT2, P4E6 and PC3 cells, respectively, Figure 6). The decrease in cell viability is statistically significant for PNT2 and P4E6 (p<0.009) but not for PC3 (p=0.08).
Example 6 Effect of NETO-2 inhibition on viability of prostate cells as measured by an apoptosis assay
The effect of NETO-2 inhibition on cell viability was also determined using a FACS based apoptosis assay. Cell death was monitored by determining the expression of caspase proteins, which are involved in the apoptosis cell signalling pathway. In addition to the caspase inhibitor, DAPI uptake was used as an indicator of compromised integrity of the plasma membrane which is a feature of necrosis. NETO-2 specific siRNA treatment did not show a change in cell death for PNT2 or P4E6 cells (Figure 7). However, there was an increase in cell death in PC3 cells after NET02 siRNA knockdown (34%, p<0.01 )
Example 7
Effect of NET02 on in vivo tumour formation and survival rates The effect of NETO-2 inhibition on the ability to form tumours in vivo was determined by siRNA pre-treatment of PC3 cells, which were then injected into BALB/c Nude mice. Tumour growth was monitored every two days (Figure 8). It was shown that mice given untransfected PC3 cells formed large tumours rapidly as expected, with 100% of mice in the group forming tumours. All mice in this group had to be culled by day 30 post initiation, having reached maximum tumour size allowed. Mice injected with PC3 cells treated with scrambled control siRNA formed tumours in 100% of the mice. These tumours were smaller and took longer to form than the untransfected group, suggesting that siRNA treatment may be affecting tumour growth. All mice in this group were culled by day 42 post initiation. In contrast, pre-treatment of PC3 cells with NET02 siRNA caused a significant decrease in the size and formation of tumours, with only 30% of mice forming small tumours, and only one mouse being culled at day 67 post initiation, having reached the maximum tumour size permissible. NET02 siRNA pre-treatment of PC3 cells caused a significant increase in survival proportion compared to both untransfected and scrambled siRNA pre-treated cells, as shown by Kaplan-Meier survival curves. Median survival for untransfected, scrambled siRNA and NET02 were 28 days, 39 days and undefined respectively.

Claims

Claims
1 . A pharmaceutical composition comprising an agent that inhibits tumour initiation wherein the agent is selected from the group consisting of:
i) an agent comprising one or more antisense oligonucleotide or antisense RNA molecules wherein said oligonucleotide or antisense RNA molecule comprise a nucleotide sequence adapted to anneal to a sense nucleotide sequence derived from at least one gene represented by the sense sequence presented in Figure 1 a; or
ii) an agent comprising an antibody, or active antibody binding fragment thereof, that binds to the extracellular domain of NETO 2.
2. The pharmaceutical composition according to claim 2 wherein said composition consists essentially of one or more antisense oligonucleotides or antisense RNA molecules and physiologically compatible excipients and/or adjuvants.
3. The pharmaceutical composition according to claim 1 or 2 wherein said antisense RNA molecule is part of a siRNA or shRNA molecule.
4. The pharmaceutical composition according to any of claims 1 to 3 wherein said antisense RNA molecule is between 19 nucleotides [nt] and 29nt in length.
5. The pharmaceutical composition according to claim 3 or 4 wherein said siRNA/shRNA is selected from the group set forth in SEQ ID NO: 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52, 53, 54 or 55.
6. The pharmaceutical composition according to any of claims 3 to 5 wherein said antisense, siRNA or shRNA includes modified nucleotides.
7. The pharmaceutical composition according to any of claims 1 to 6 wherein composition includes a carrier adapted to deliver said antisense oligonucleotide or antisense RNA to a cell or tissue.
8. The pharmaceutical composition according to claim 1 wherein said antibody is polyclonal.
9. The pharmaceutical composition according to claim 1 wherein said antibody is a monoclonal antibody.
10. The pharmaceutical composition according to claim 1 wherein said fragment is a single chain antibody fragment.
1 1 . The pharmaceutical composition according to claim 1 wherein said antibody is a chimeric antibody.
12. The pharmaceutical composition according to claim 1 wherein said antibody is a humanized or human antibody.
13. The pharmaceutical composition according to any one of claims 1 -12 wherein said extracellular domain of NET02 comprises or consists essentially of the amino acid sequence set forth in Figure 1 C.
14. A pharmaceutical composition according to any of claims 1 to 13 wherein said composition includes supplementary anti-cancer agents.
15. A pharmaceutical composition according to claim 14 wherein said anti-cancer agent is a chemotherapeutic agent.
16. The use of a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence as represented in Figure 1 A for the identification of agents that modulate the activity of said polypeptide.
17. A screening method for the identification of an agent that inhibits the activity of a cancer stem cell gene expression product comprising:
i) providing a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence as represented in Figure 1 A; ϋ) providing at least one candidate agent to be tested; iii) forming a preparation that is a combination of (i) and (ii) above; and
testing the effect of said agent on the activity of said polypeptide.
18. A vaccine composition comprising a polypeptide selected from the group consisting of: i) a polypeptide encoded by a nucleotide sequence as represented in Figure 1 A, or an antigenic fragment thereof;
ii) a polypeptide encoded by a nucleotide sequence wherein said sequence is degenerate as a result of the genetic code to the nucleotide sequence defined in (i);
iii) a polypeptide or antigenic fragment comprising an amino acid sequence wherein said sequence is modified by addition deletion or substitution of at least one amino acid residue as represented in Figures 1 B, wherein said composition optionally includes an adjuvant and/or carrier.
19. A vaccine composition according to claim 18 wherein said antigenic fragment is an extracellular domain of said polypeptide.
20. A vaccine composition according to claim 19 wherein said extracellular domain comprises or consists of the amino acid sequence as represented in Figure 1 C.
21 . A vaccine composition according to any of claims 19 to 20 wherein said composition includes an adjuvant.
22. A vaccine according to any of claims 18-21 for use in the treatment of cancer.
23. A vaccine according to claim 22 wherein said cancer is carcinoma.
24. A vaccine composition according to claim 23 wherein said carcinoma is prostate carcinoma.
25. A diagnostic or prognostic method for the detection of cancer cells isolated from a subject comprising determining the expression of NETO-2, wherein over-expression of said gene is indicative of cancer or a predisposition to cancer in said subject.
26. A method according to claim 25 wherein said method comprises:
i) providing an isolated biological sample to be tested; ii) forming a preparation comprising said sample and an oligonucleotide primer pair adapted to anneal to a nucleic acid molecule comprising a nucleic acid sequence as represented in Figure 1 A; a thermostable DNA polymerase, deoxynucleotide triphosphates and co-factors;
iii) providing polymerase chain reaction conditions sufficient to amplify said nucleic acid molecule;
iv) analysing the amplified products of said polymerase chain reaction for the presence or absence of a nucleic acid molecule comprising a nucleotide sequence derived from Figure 1 A; and optionally v) comparing the amplified product with a normal matched control.
27. A method according to claim 25 wherein said method comprises:
i) providing an isolated biological sample to be tested;
ii) forming a preparation comprising said sample and an antibody or antibodies that specifically binds one or more polypeptide^] in said sample as represented by the amino acid sequences presented in Figures 1 B to form an antibody/polypeptide complex;
iii) detecting the complex or complexes so formed; and iv) comparing the expression of said polypeptide^] with a normal matched control.
28. A kit comprising oligonucleotide primer pairs adapted to amplify one or more nucleic acid molecules comprising the nucleotide sequences as represented in Figure 1 A.
29. A kit according to claim 28 wherein said kit further includes components required for polymerase chain reaction.
30. A kit comprising one or more antibodies adapted to bind one or more polypeptides as represented by the amino acid sequences presented in Figures 1 B.
31 . A kit according to claim 30 wherein said kit further includes components required for the detection of bound antibody in an immunoassay.
32. An isolated composition comprising or consisting essentially of the extracellular domain of NETO-2 for use in the screening of antibody phage display libraries for the identification of antagonistic NETO-2 antibodies.
33. The composition according to claim 32 wherein said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
34. A method to screen a combinatorial antibody phage display library wherein said library comprises antibody variable heavy and variable light chain nucleic acids comprising the steps:
i) contacting a phage display library expressing said antibody variable heavy and light chains with a composition comprising or consisting essentially of the NETO-2 extracellular domain;
ii) selecting phage that specifically bind the NETO-2 extracellular domain contained in said composition;
iii) repeating i) and ii) above to identify phage clones that bind said NETO-2 extracellular domain until high affinity binders are selected; and optionally iv) isolating and storing said phage.
35. The method according to claim 34 wherein said extracellular domain consists of the amino acid sequence set forth in Figure 1 C.
PCT/GB2012/052400 2011-09-28 2012-09-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2 WO2013045934A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2847826A CA2847826A1 (en) 2011-09-28 2012-09-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2
EP12780520.8A EP2732036A2 (en) 2011-09-28 2012-09-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2
US14/227,538 US20140199310A1 (en) 2011-09-28 2014-03-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1116702.0A GB201116702D0 (en) 2011-09-28 2011-09-28 Cell surface markers
GB1116702.0 2011-09-28

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/227,538 Continuation-In-Part US20140199310A1 (en) 2011-09-28 2014-03-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2

Publications (2)

Publication Number Publication Date
WO2013045934A2 true WO2013045934A2 (en) 2013-04-04
WO2013045934A3 WO2013045934A3 (en) 2013-05-30

Family

ID=44994103

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2012/052400 WO2013045934A2 (en) 2011-09-28 2012-09-27 Compositions comprising agents that inhibit neuropilin and tolloid like 2

Country Status (5)

Country Link
US (1) US20140199310A1 (en)
EP (2) EP2732036A2 (en)
CA (1) CA2847826A1 (en)
GB (1) GB201116702D0 (en)
WO (1) WO2013045934A2 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5978740A (en) 1995-08-09 1999-11-02 Vertex Pharmaceuticals Incorporated Molecules comprising a calcineurin-like binding pocket and encoded data storage medium capable of graphically displaying them
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
WO2005089043A2 (en) 2004-03-19 2005-09-29 Procure Therapeutics Limited Prostate stem cell
WO2012101904A1 (en) 2011-01-28 2012-08-02 オリンパスメディカルシステムズ株式会社 Illumination device and observation system

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US5978740A (en) 1995-08-09 1999-11-02 Vertex Pharmaceuticals Incorporated Molecules comprising a calcineurin-like binding pocket and encoded data storage medium capable of graphically displaying them
WO2005089043A2 (en) 2004-03-19 2005-09-29 Procure Therapeutics Limited Prostate stem cell
WO2012101904A1 (en) 2011-01-28 2012-08-02 オリンパスメディカルシステムズ株式会社 Illumination device and observation system

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
"Remington's Pharmaceutical Sciences", MACK PUBLISHING CO.
"User's Guide", vol. 3, pages: 134 - 135
"Using Antibodies: A laboratory manual", COLD SPRING HARBOUR LABORATORY PRESS
AI HAJJ M; WICHA MS; BENITOHERNANDEZ A; MORRISON SJ; CLARKE MF: "Prospective identification of tumorigenic breast cancer cells", PROC NATL ACAD SCI USA, vol. 100, 2003, pages 3983 - 3988
BONNET D; DICK J.E.: "Human acute myeloid leukaemia is organized as a hierarchy that originates from a primitive hematopoietic cell", NAT. MED., vol. 3, 1997, pages 730737
COLLINS AT; BERRY PA; HYDE C; STOWER MJ; MAITLAND NJ: "Prospective Identification of Tumorigenic Prostate Cancer Stem Cells", CANCER RES., vol. 65, 2005, pages 10946 - 0951, XP008128778, DOI: doi:10.1158/0008-5472.CAN-05-2018
HAMBURGER AW; SALMON SE: "Primary bioassay of human tumor stem cells", SCIENCE, vol. 197, 1977, pages 461 - 63, XP001094575, DOI: doi:10.1126/science.560061
HOUGHTON J; STOICOV C; NOMURA S; ROGERS AB; CARLSON J; LI H; CAI X; FOX JG; GOLDENRING JR; WANG TC: "Gastric cancer originating from bone marrow derived cells", SCIENCE, vol. 306, 2004, pages 1568 - 1571, XP002374400, DOI: doi:10.1126/science.1099513
KIM CF; JACKSON EL; WOOLFENDEN AE; LAWRENCE S; BABAR I; VOGEL S; CROWLEY D; BRONSON RT; JACKS T: "Identification of bronchioalveolar stem cells in normal lung and lung cancer", CE, vol. 121, 2005, pages 823 - 835
MOLECULAR PHARMACEUTICS, vol. 6, no. 3, 2008, pages 651 - 658
NATURE REVIEWS, vol. 8, 2009, pages 129
O'BRIEN CA; POLLETT A; GALLINGER S; DICK JE: "A human colon cancer cell capable of initiating tumour growth in immunodeficient mice", NATURE, vol. 445, 2007, pages 106 - 110
P. A. BARTLETT ET AL.: "In Molecular Recognition in Chemical and Biological Problems", vol. 78, 1989, SPECIAL PUB., ROYAL CHEM. SOC., article "CAVEAT: A Program to Facilitate the Structure-Derived Design of Biologically Active Molecules", pages: 182 - 196
PARDAL R; CLARKE MF; MORRISON SJ: "Applying the principles of stem cell biology to cancer", NAT. REV. CANCER, vol. 3, 2003, pages 89502
PHARMACEUTICAL RESEARCH, vol. 26, no. 3, 2009, pages 657
RICCIVITIANI L; LOMBARDI DG; PILOZZI E; BIFFONI M; TODARO M; PESCHLE C; DE MARIA R: "Identification and expansion of human colon cancer initiating cells", NATURE, vol. 445, 2007, pages 111 - 115
SINGH SK; HAWKINS C; CLARKE ID; SQUIRE JA; BAYANI J; HIDE T; HENKELMAN RM; CUSIMANO MD; DIRKS PB: "Identification of human brain tumour initiating cells", NATURE, vol. 432, 2004, pages 396 - 401, XP002467853, DOI: doi:10.1038/nature03128
THE AAPS JOURNAL, vol. 11, no. 4, 2009, pages 639
WAGNER ET AL., NATURE BIOTECHNOLOGY, vol. 14, 1996, pages 840 - 844
Y. C. MARTIN: "3D Database Searching in Drug Design", J. MED. CHEM., vol. 35, 1992, pages 2145 - 2154

Also Published As

Publication number Publication date
EP2765141A1 (en) 2014-08-13
CA2847826A1 (en) 2013-04-04
US20140199310A1 (en) 2014-07-17
EP2732036A2 (en) 2014-05-21
GB201116702D0 (en) 2011-11-09
WO2013045934A3 (en) 2013-05-30

Similar Documents

Publication Publication Date Title
US11708412B2 (en) Methods for treating hematologic cancers
RU2756275C2 (en) Antibodies specific to human poliovirus receptor (pvr)
US20220033502A1 (en) METHODS FOR UPREGULATING IMMUNE RESPONSES USING COMBINATIONS OF ANTI-RGMb AND ANTI-PD-1 AGENTS
KR20170061152A (en) Modulation of stimulatory and non-stimulatory myeloid cells
CN106659772B (en) Compositions and methods for modulating renalase in the treatment of diseases and disorders
AU2015335029A1 (en) Combination
EP3362074A2 (en) Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
JP2014526475A5 (en)
JP2014526475A (en) Hs. For inhibition of proliferation, development, or differentiation of stem cells, including cancer stem cells. Antagonist of the 549642 Unigene cluster product
JP2022512901A (en) CD73 antibody that activates B cells
JP2022502062A (en) 2&#39;FANA-modified FOXP3 antisense oligonucleotide and its usage
WO2010079833A1 (en) Novel cancer antigen eef2
AU2019227641A1 (en) Methods for treating cancer using combinations of anti-BTNL2 and immune checkpoint blockade agents
CN115485395A (en) Compositions and methods comprising splice-derived antigens for the treatment of cancer
CN112996504A (en) Methods of treating cancer by inhibiting ubiquitin conjugating enzyme E2K (UBE2K)
EP2765141A1 (en) Compositions comprising agents that inhibit neuroplilin and tolloid like 2
US20210355221A1 (en) Targeting the Non-Canonical NFkB Pathway in Cancer Immunotherapy
US20240201192A1 (en) Folr2+ macrophages and anti-tumor immunity
CA3225139A1 (en) Compositions and methods for inhibiting the expression of tmigd2
JP2022513082A (en) Use of IRE1α-XBP1 signaling pathway biomarkers to regulate immune response
Konga et al. Blockade of Immune Checkpoint B7-H4 and Lysine Demethylase 5B in Esophageal Squamous Cell Carcinoma Confers Protective Immunity against P. gingivalis Infection
WO2011110838A2 (en) Differentiation factor

Legal Events

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

Ref document number: 12780520

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2012780520

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2847826

Country of ref document: CA