US20060100143A1 - Polypeptide - Google Patents

Polypeptide Download PDF

Info

Publication number
US20060100143A1
US20060100143A1 US10/522,043 US52204305A US2006100143A1 US 20060100143 A1 US20060100143 A1 US 20060100143A1 US 52204305 A US52204305 A US 52204305A US 2006100143 A1 US2006100143 A1 US 2006100143A1
Authority
US
United States
Prior art keywords
polypeptide
iaspp
nucleic acid
peptide
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/522,043
Inventor
Xin Lu
Patricia Kuwabara
David Selwood
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ludwig Institute for Cancer Research Ltd
Genome Research Ltd
UCL Cruciform Ltd
Original Assignee
Ludwig Institute for Cancer Research Ltd
Genome Research Ltd
UCL Cruciform Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0223193A external-priority patent/GB0223193D0/en
Priority claimed from GB0306261A external-priority patent/GB0306261D0/en
Application filed by Ludwig Institute for Cancer Research Ltd, Genome Research Ltd, UCL Cruciform Ltd filed Critical Ludwig Institute for Cancer Research Ltd
Assigned to LUDWIG INSTITUTE FOR CANCER RESEARCH reassignment LUDWIG INSTITUTE FOR CANCER RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LU, XIN
Assigned to UCL CRUCIFORM LIMITED reassignment UCL CRUCIFORM LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SELWOOD, DAVID
Assigned to GENOME RESEARCH LIMITED reassignment GENOME RESEARCH LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KUWABARA, PATRICIA
Publication of US20060100143A1 publication Critical patent/US20060100143A1/en
Priority to US12/043,058 priority Critical patent/US7745571B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/50Hydrolases (3) acting on carbon-nitrogen bonds, other than peptide bonds (3.5), e.g. asparaginase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4747Apoptosis related proteins

Definitions

  • the invention relates to a polypeptide, or part thereof, which inhibits the apoptotic activity of the tumour suppressor protein p53; screening methods to identify agents which interfere with the activity of said polypeptide and agents with said activity.
  • Apoptosis or programmed cell death, is a process by which multi-cellular organisms regulate cell number and differentiation. The process is regulated by factors which either induce or prevent apoptosis. Inducers of apoptosis include Bcl-2 family members, caspase family members and their associated factors Apaf-1 and Fadd. Caspases are synthesised as proenzymes which become activated after proteolytic cleavage. The active caspase then induces many of the morphological and biochemical changes associated with apoptosis. Mitochondria play a pivotal role in the activation process through the release of pro-apoptotic factors such as cytochrome c, AIF and Diablo. The release from mitochondria is controlled by the Bcl-2 family of proteins; (e.g. Bcl-2 and Bcl-xl inhibit release; Bax and Bak induce release).
  • Bcl-2 family of proteins e.g. Bcl-2 and Bcl-xl inhibit release; Bax and Bak induce
  • WO9953051 discloses a cytokine dependent protein p21 which has pro-apoptotic activity.
  • p21 is expressed in a cytokine dependent manner in myeloid/erthyroid cells. These cells are dependent on IL-3 for growth and in the absence of IL-3 the translation of p21 is induced resulting in apoptosis and cell death.
  • p21 is a cytoplasmic protein which translocates to the outer mitochondrial membrane to induce pro-apoptotic activities.
  • Tumour suppressor proteins also have pro-apoptotic activities.
  • Tumour suppressor genes encode proteins which function to inhibit cell growth or division and are therefore important with respect to maintaining proliferation, growth and differentiation of normal cells. Mutations in tumour suppressor genes result in abnormal cell-cycle progression whereby the normal cell-cycle check points which arrest the cell-cycle, when, for example, DNA is damaged, are ignored and damaged cells divide uncontrollably.
  • the products of tumour suppressor genes function in all parts of the cell (e.g. cell surface, cytoplasm, nucleus) to prevent the passage of damaged cells through the cell- cycle (i.e. G1, S, G2, M and cytokinesis).
  • the tumour suppressor gene which has been the subject of the most intense research is p53.
  • p53 encodes a protein which functions as a transcription factor and is a key regulator of the cell division cycle. It was discovered in 1978 as a protein shown to bind with affinity to the SV40 large T antigen.
  • the p53 gene encodes a 393 amino acid polypeptide with a molecular weight of 53 kDa.
  • Genes regulated by the transcriptional activity of p53 contain a p53 recognition sequence in their 5′ regions. These genes are activated when the cellular levels of p53 are elevated due to, for example DNA damage. Examples of genes which respond to p53 include, mdm2, Bax and PIG-3. Bax and PIG-3 are involved in one of the most important functions of p53, the induction of apoptosis.
  • ASPP serotonin-1 phosphatidylcholine
  • iASPP an inhibitor of ASPP family members referred to as iASPP.
  • iASPP is an oncogene and is the most conserved member of the ASPP family.
  • iASPP is the only ASPP-like protein found in C. elegans. Similar to human iASPP, the C. elegans homologue functions as a key inhibitor of p53.
  • the C. elegans iASPP is capable of substituting for human iASPP in all of the assays performed in human cells. Moreover, reciprocal substitution studies reveal that the apoptotic function of C. elegans p53 is enhanced or inhibited by human ASPP and iASPP, respectively. Using RNAi we further demonstrate that iASPP is a key inhibitor of p53 mediated apoptosis in C. elegans. All of these observations show that the regulation of p53 by ASPP family members is evolutionarily conserved. Control of p53 activity plays a pivotal role in development and tumourigenesis. Hence, inhibiting the oncogenic function of iASPP could provide an important new strategy for treating tumours expressing wild type p53. Sequence comparision between C. elegans and human iASPP reveals a conserved domain between the nematode and human sequence which likely explains the functional conservation between the proteins.
  • an isolated nucleic acid molecule which encodes a polypeptide, or sequence variant thereof, wherein said polypeptide is a fragment of the polypeptide sequence represented in FIG. 1 a or 1 b, which fragment is selected from the group consisting of:
  • said nucleic acid molecule encodes a polypeptide fragment consisting of amino acid residues from about 128-224 of the sequence represented in FIG. 1 a.
  • said nucleic acid molecule is isolated from a human.
  • said nucleic acid molecule encodes a polypeptide fragment consisting of amino acid residues from about 128-224 of the sequence represented in FIG. 1 b.
  • said nucleic acid molecule is isolated from a nematode.
  • said nematode is of the genus Caenorhabditis spp.
  • nucleic acid molecule encodes a polypeptide, or sequence variant thereof, which polypeptide inhibits the activity of a polypeptide represented by the amino acid sequence represented in FIG. 1 a or 1 b.
  • said nucleic acid molecule is a cDNA.
  • said nucleic acid molecule is genomic DNA.
  • a polypeptide fragment or sequence variant thereof encoded by a nucleic acid molecule according to the invention.
  • fragments which are sequence variants may retain the biological activity of the full length polypeptide or alternatively have antagonistic activity by competing for binding sites in p53.
  • specificity of polypeptides according to the invention with respect to binding to p53 is shown by binding equilibrium constants.
  • Polypeptides which are capable of selectively binding p53 preferably have binding equilibrium constants of at least about 10 7 M ⁇ 1 , more preferably at least about 10 8 M ⁇ 1 , and most preferably at least about 10 9 M ⁇ 1 .
  • a sequence variant i.e. a fragment polypeptide and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, truncations which may be present in any combination.
  • preferred variants are those that vary from a reference polypeptide by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid by another amino acid of like characteristics.
  • amino acids are considered conservative replacements (similar): a) alanine, serine, and threonine; b) glutamic acid and asparatic acid; c) asparagine and glutamine d) arginine and lysine; e) isoleucine, leucine, methionine and valine and f) phenylalaine, tyrosine and tryptophan.
  • a functionally equivalent polypeptide according to the invention is a variant wherein one or more amino acid residues are substituted with conserved or non-conserved amino acid residues, or one in which one or more amino acid residues includes a substituent group.
  • Conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu and Ile; interchange of the hydroxl residues Ser and Thr; exchange of the acidic residues Asp and Glu; substitution between amide residues Asn and Gln; exchange of the basic residues Lys and Arg; and replacements among aromatic residues Phe and Tyr.
  • the invention features polypeptide sequences having at least 75% identity with the polypeptide sequences as hereindisclosed, or fragments and functionally equivalent polypeptides thereof.
  • the polypeptides have at least 85% identity, more preferably at least 90% identity, even more preferably at least 95% identity, still more preferably at least 97% identity, and most preferably at least 99% identity with the amino acid sequences illustrated herein.
  • the invention also provides, in certain embodiments, “dominant negative” polypeptides derived from the polypeptides hereindisclosed.
  • a dominant negative polypeptide is an inactive variant of a protein, which, by interacting with the cellular machinery, displaces an active protein from its interaction with the cellular machinery or competes with the active protein, thereby reducing the effect of the active protein.
  • a dominant negative receptor which binds a ligand but does not transmit a signal in response to binding of the ligand can reduce the biological effect of expression of the ligand.
  • a dominant negative catalytically-inactive kinase which interacts normally with target proteins but does not phosphorylate the target proteins can reduce phosphorylation of the target proteins in response to a cellular signal.
  • a dominant negative transcription factor which binds to another transcription factor or to a promoter site in the control region of a gene but does not increase gene transcription can reduce the effect of a normal transcription factor by occupying promoter binding sites without increasing transcription.
  • the end result of the expression of a dominant negative polypeptide in a cell is a reduction in function of active proteins.
  • One of ordinary skill in the art can assess the potential for a dominant negative variant of a protein, and using standard mutagenesis techniques to create one or more dominant negative variant polypeptides.
  • one of ordinary skill in the art can modify the sequence of iASPP polypeptides by site-specific mutagenesis, scanning mutagenesis, partial gene deletion or truncation, and the like. See, e.g., U.S. Pat. No. 5,580,723 and Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, 1989.
  • the skilled artisan then can test the population of mutagenized polypeptides for diminution in a selected activity (e.g., p53 binding, modulation of apoptosis) and/or for retention of such an activity.
  • a selected activity e.g., p53 binding, modulation of apoptosis
  • Other similar methods for creating and testing dominant negative variants of a protein will be apparent to one of ordinary skill in the art.
  • a vector comprising a nucleic acid according to the invention.
  • said vector is an expression vector conventionally adapted for gene expression.
  • said adaptation includes, by example and not by way of limitation, the provision of transcription control sequences (promoter sequences) which mediate cell/tissue specific expression.
  • promoter sequences may be cell/tissue specific, inducible or constitutive.
  • Enhancer elements are cis acting nucleic acid sequences often found 5′ to the transcription initiation site of a gene (enhancers can also be found 3′ to a gene sequence or even located in intronic sequences and is therefore position independent). Enhancers function to increase the rate of transcription of the gene to which the enhancer is linked. Enhancer activity is responsive to trans acting transcription factors (polypeptides) which have been shown to bind specifically to enhancer elements.
  • transcription factors are responsive to a number of environmental cues which include, by example and not by way of limitation, intermediary metabolites (eg glucose, lipids), environmental effectors (eg light, heat,).
  • intermediary metabolites eg glucose, lipids
  • environmental effectors eg light, heat,
  • Promoter elements also include so called TATA box and RNA polymerase initiation selection (RIS) sequences which function to select a site of transcription initiation. These sequences also bind polypeptides which function, inter alia, to facilitate transcription initiation selection by RNA polymerase.
  • RIS RNA polymerase initiation selection
  • Adaptations also include the provision of selectable markers and autonomous replication sequences which both facilitate the maintenance of said vector in either the eukaryotic cell or prokaryotic host.
  • Vectors which are maintained autonomously are referred to as episomal vectors.
  • Episomal vectors are desirable since these molecules can incorporate large DNA fragments (30-50 kb DNA). Episomal vectors of this type are described in WO98/07876.
  • Adaptations which facilitate the expression of vector encoded genes include the provision of transcription termination/polyadenylation sequences. This also includes the provision of internal ribosome entry sites (IRES) which function to maximise expression of vector encoded genes arranged in bicistronic or multi-cistronic expression cassettes.
  • IRS internal ribosome entry sites
  • LCRs Locus Control Regions
  • a cell transformed or transfected with a nucleic acid molecule or vector according to the invention is provided.
  • said host cells are eukaryotic cells, for example, insect cells such as cells from a species Spodoptera frugiperda using a baculovirus expression system.
  • This expression system is favoured in the instance where post-translational modification of the polypeptide is required.
  • Host cells and cell lines can be prokaryotic (e.g., E. coli ), or eukaryotic (e.g., CHO cells, COS cells, yeast expression systems and recombinant baculovirus expression in insect cells).
  • mammalian cells such as human, mouse, hamster, pig, goat, primate, etc. They may be of a wide variety of tissue types, and include primary cells and cell lines.
  • keratinocytes include peripheral blood leukocytes, fibroblasts, bone marrow stem cells and embryonic stem cells.
  • the expression vectors require that the pertinent sequence, i.e., those nucleic acids described above, be operably linked to a promoter.
  • the invention there is provided a polypeptide according to the invention for use as a pharmaceutical.
  • nucleic acid according to the invention for use as a pharmaceutical.
  • said pharmaceutical further comprises a a diluent, carrier or excipient.
  • 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, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents, such as chemotherapeutic agents.
  • the therapeutics of the invention can be administered by any conventional route, including injection or by gradual infusion over time.
  • the administration may, for example, be oral, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, or transdermal.
  • a preferred route of administration is by pulmonary aerosol.
  • Techniques for preparing aerosol delivery systems containing antibodies are well known to those of skill in the art. Generally, such systems should utilize components which will not significantly impair the biological properties of the antibodies, such as the paratope binding capacity (see, for example, Sciarra and Cutie, “Aerosols,” in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp 1694-1712; incorporated by reference).
  • Those of skill in the art can readily determine the various parameters and conditions for producing antibody aerosols without resort to undue experimentation. When using antisense preparations of the invention, slow intravenous administration is preferred.
  • 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 or can be monitored according to diagnostic methods of the invention discussed herein.
  • 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 dominant negative iASPP or nucleic acid encoding a dominant negative iASPP, for producing the desired response in a unit of weight or volume suitable for administration to a patient.
  • the response can, for example, be measured by determining the signal transduction inhibited by the dominant negative iASPP-1, composition via a reporter system as described herein, by measuring downstream effects such as gene expression, or by measuring the physiological effects of the iASPP composition, such as regression of a tumour, decrease of disease symptoms, modulation of apoptosis, etc.
  • the doses of dominant negative iASPP polypeptide or nucleic acid 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 dominant negative iASPP are formulated and administered in doses between 1 ng and about 500 mg, and between 10 ng and 100 mg, according to any standard procedure in the art. Where nucleic acids encoding dominant negative iASPP are employed, doses of between 1 ng and 0.1 mg generally will be formulated and administered according to standard procedures. Other protocols for the administration of iASPP 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 (e.g., intra-tumoral) and the like vary from the foregoing. Administration of iASPP 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.
  • 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.
  • the salts 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.
  • pharmaceutically-acceptable salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts.
  • iASPP 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 denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • 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.
  • compositions also may contain, optionally, suitable preservatives, such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
  • 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 a syrup, elixir or an emulsion.
  • compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of iASPP polypeptides or nucleic acids, 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 non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol.
  • acceptable vehicles and 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.
  • transgenic non-human animal comprising a nucleic acid according to the invention.
  • transgenic non-human animals includes non-human animals having one or more exogenous nucleic acid molecules incorporated in germ line cells and/or somatic cells.
  • the transgenic animal include “knockout” animals having a homozygous or heterozygous gene disruption by homologous recombination, animals having episomal or chromosomally incorporated expression vectors, etc.
  • Knockout animals can be prepared by homologous recombination using embryonic stem cells as is well known in the art. The recombination can be facilitated by the cre/lox system or other recombinase systems known to one of ordinary skill in the art.
  • the recombinase system itself is expressed conditionally, for example, in certain tissues or cell types, at certain embryonic or post-embryonic developmental stages, inducibly by the addition of a compound which increases or decreases expression, and the like.
  • conditional expression vectors used in such systems use a variety of promoters which confer the desired gene expression pattern (e.g., temporal or spatial).
  • Conditional promoters also can be operably linked to iASPP family nucleic acid molecules to increase expression of these nucleic acid molecules in a regulated or conditional manner.
  • Trans-acting negative regulators of iASPP activity or expression also can be operably linked to a conditional promoter as described above.
  • trans-acting regulators include antisense nucleic acids molecules, nucleic acid molecules which encode dominant negative molecules, ribozyme molecules specific for iASPP nucleic acids, and the like.
  • the transgenic non-human animals are useful in experiments directed toward testing biochemical or physiological effects of diagnostics or therapeutics for conditions characterized by increased or decrease iASPP expression. Other uses will be apparent to one of ordinary skill in the art.
  • polypeptide or fragment thereof, in a screening method for the identification of agents which inhibit the binding of said polypeptide to p53.
  • a screening method to identify agents which inhibit the binding of a polypeptide or fragment thereof to p53 comprising:
  • said agent is a polypeptide, preferably a peptide.
  • said peptide comprises an amino acid sequence selected from the group consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • said peptide is at least 6 amino acid residues in length.
  • the length of said peptide is selected from the group consisting of: at least 7 amino acid residues; 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues in length.
  • the length of said peptide is at least 20 amino acid residues; 30; 40; 50; 60; 70; 80; 90; or 100 amino acid residues in length.
  • said peptide consists of an amino acid sequence consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • modification to the amino acid sequence of peptides agents could enhance the binding and/or stability of the peptide with respect to its target sequence.
  • modification of the peptide may also increase the in vivo stability of the peptide thereby reducing the effective amount of peptide. necessary to inhibit p53 binding of iASPP. This would advantageously reduce undesirable side effects which may result in vivo.
  • Modifications include, by example and not by way of limitation, acetylation and amidation.
  • said modification includes the use of modified amino acids in the production of recombinant or synthetic forms of peptides.
  • modified amino acids include, by way of example and not by way of limitation, 4-hydroxyproline, 5-hydroxylysine, N 6 -acetyllysine, N 6 -methyllysine, N 6 ,N 6 -dimethyllysine, N 6 ,N 6 ,N 6 -trimethyllysine, cyclohexyalanine, D-amino acids, ornithine.
  • Other modifications include amino acids with a C 2 , C 3 or C 4 alkyl R group optionally substituted by 1, 2 or 3 substituents selected from halo (eg F, Br, I), hydroxy or C 1 -C 4 alkoxy.
  • Cyclisation is known in the art, (see Scott et al Chem Biol (2001), 8:801-815; Gellerman et al J. Peptide Res (2001), 57: 277-291; Dutta et al J. Peptide Res (2000), 8: 398-412; Ngoka and Gross J Amer Soc Mass Spec (1999), 10:360-363.
  • said antagonist is an antibody or antibody binding part.
  • said antibody is a monoclonal antibody or binding part thereof.
  • Immunoglobulins are protein molecules which usually 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, ⁇ , ⁇ , ⁇ , ⁇ , 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 IgA, IgM, IgD, IgE and IgG (with four sub-classes based on the differences in the ‘constant’ regions of the H chains, i.e., IgG1, IgG2, IgG3 and IgG4).
  • IgG1, IgG2, IgG3 and IgG4 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 Fab fragment.
  • said antibody is selected from the group consisting of: F(ab′) 2 , Fab, Fv and Fd fragments; and antibodies comprising CDR3 regions.
  • said fragments are single chain antibody variable regions (scFV's) or domain antibodies. If a hybidoma exists for a specific monoclonal antibody it is well within the knowledge of the skilled person to isolate scFv's from mRNA extracted from said hybridoma via RT PCR. Alternatively, phage display screening can be undertaken to identify clones expressing scFv's. Domain antibodies are the smallest binding part of an antibody (approximately 13 kDa). Examples of this technology is disclosed in U.S. Pat. No. 6,248,516, U.S. Pat. No. 6,291,158, U.S. Pat. No. 6,127,197 and EP0368684 which are all incorporated by reference in their entirety.
  • a modified antibody, or variant antibody, and reference antibody may differ in amino acid sequence by one or more substitutions, additions, deletions, truncations which may be present in any combination.
  • preferred variants are those that vary from a reference polypeptide by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid by another amino acid of like characteristics.
  • amino acids are considered conservative replacements (similar): a) alanine, serine, and threonine; b) glutamic acid and asparatic acid; c) asparagine and glutamine d) arginine and lysine; e) isoleucine, leucine, methionine and valine and f) phenylalanine, tyrosine and tryptophan. Most highly preferred are variants which show enhanced biological activity.
  • said antibody is a humanised or chimeric antibody.
  • a chimeric antibody is produced by recombinant methods to contain the variable region of an antibody with an invariant or constant region of a human antibody.
  • a humanised antibody is produced by recombinant methods to combine the complementarity determining regions (CDRs) of an antibody with both the constant (C) regions and the framework regions from the variable (V) regions of a human antibody.
  • CDRs complementarity determining regions
  • 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 complimentarity 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 complimentarity 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 elicit 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.
  • an isolated nucleic acid molecule wherein said molecule is isolated from a nematode worm which nucleic acid molecule hybridises a nucleic acid sequence as represented by FIG 1 b.
  • nucleic acid molecule hybridises under stringent hybridisation condtions.
  • said nematode worm is of the genus Caenorhabditis spp.
  • an isolated polypeptide comprising the amino acid as represented in FIG. 1 b which polyeptide is modified by addition, deletion or substitution of at least one amino acid residue.
  • a method of treatment of an animal comprising administering an effective amount of a polypeptide or nucleic acid or vector according to the invention wherein said effective amount induces the apopoptic activity of p53.
  • said treatment is of cancer.
  • a peptide comprising an amino acid sequence selected from the group consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • said peptide is at least 6 amino acid residues in length.
  • the length of said peptide is selected from the group consisting of: at least 7 amino acid residues; 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues in length.
  • the length of said peptide is at least 20 amino acid residues; 30; 40; 50; 60; 70; 80; 90; or 100 amino acid residues in length.
  • said peptide consists of an amino acid sequence consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • composition comprising at least one peptide according to the invention and at least one anti-cancer agent
  • said said anticancer agent is selected from the group consisting of: cisplatin; carboplatin; cyclosphosphamide; melphalan; carmusline; methotrexate; 5-fluorouracil; cytarabine; mercaptopurine; daunorubicin; doxorubicin; epirubicin; vinblastine; vincristine; dactinomycin; mitomycin C; taxol; L-asparaginase; G-CSF; etoposide; colchicine; derferoxamine mesylate; and camptothecin.
  • a complex comprising a peptide according to the invention and at least one antibody, or active binding part thereof.
  • said antibody or fragment is a cell specific antibody.
  • said cell is a cancer cell.
  • a complex of a peptide according to the invention and an antibody which specifically binds a cell, preferably a cancer cell, enables the targeting of said peptide to cell.
  • the complex would be internalised releasing the peptide intracellularly to deliver the peptide to its target.
  • Means to form a complex of antibody and peptide are known in the art and include the use of, for example bi-functional crosslinking agents, these may be hetero-bifunctional or homo-bifunctional.
  • the crosslinkers may be reducible thereby facilitating release of said peptide(s).
  • Cancer specific cell markers are known in the art. For example, tumour rejection antigen precursors.
  • a method of treatment of an animal preferably a human, wherein said animal would benefit from the induction of apoptosis comprising administering an effective amount of a peptide according to the invention.
  • a method of treatment of an animal preferably a human, wherein said animal would benefit from the induction of apoptosis comprising administering an effective amount of a composition according to the invention
  • said treatment is cancer treatment.
  • FIG. 1 a is the nucleic acid sequence of human iASPP
  • FIG. 1 b is the C. elegans nucleic acid sequence of iASPP
  • FIG. 2 a is the amino acid sequence of human iASPP
  • FIG. 2 b is the C. elegans amino acid sequence of iASPP
  • FIG. 3 illustrates that FITC labelled peptide ( 3 a ) DGPEETD and ( 3 b ) TTLSDG can penetrate cells;
  • FIG. 4 illustrates the stimulation of the Bax promoter by p53 after incubation with various peptides, in particular DGPEETD;
  • FIG. 5 illustrates the stimulation of p53 transactivation in a human tumour cell line U2SO after UV damage of DNA in the presence of peptide DGPEETD;
  • FIG. 6 illustrates various experiments showing the interaction of C. elegans iASPP and p53
  • FIG. 7 illustrates a homology comparison between C. elegans iASPP and human iASPP
  • FIG. 8 illustrates further experiments showing the interaction of C. elegans iASPP and p53
  • FIG. 9 illustrates the effect of RNAi on C. elegans iASPP expression
  • FIG. 10 illustrates p53 contact sites in iASPP
  • FIG. 11 Influence of peptides on transactivation level of Bax-Luc reporter.
  • Cells were transfected with Bax reporter gene and exposed to UV (20J/m2). Peptides were added for 24 hours in medium at two different concentrations as indicated;
  • FIG. 12 illustrates the influence of peptides on Pig3 reporter activity for 12 hours.
  • Transactivated cells were grown with medium containing Arg-tagged peptides at two concentrations.
  • the left bar graph represents the transactivation activity at a concentration of 25 uM and the second is with a concentration of 50 uM.
  • Yap peptide is used as a control peptide here instead of p9. Except for the control, cells were treated with cisplatin (3 ug/ml);
  • FIG. 13 illustrates the effects of various chemotherapeutic drugs on p53 induced apoptosis
  • FIG. 14 a illustrates the effects of antisense or si-RNA of ASPP1, ASPP2 or iASPP, in combination with the activity of the anti-cancer agent doxorubicin
  • FIG. 14 b illustrates the effects of antisense or si-RNA of ASPP1, ASPP2 or iASPP in combination with the activity of the anti-cancer agent cisplatin;
  • FIG. 15 illustrates the activity of inhibitory peptides in combination with cisplatin on ASPP1, ASPP2 and iASPP.
  • Saos-2, MCF-7, and U2OS cells were grown in DMEM supplemented with 10% FCS, 100 IU/ml penicillin-streptomycin and 2 mM glutamine.
  • Anti-p53 antibodies DO-1 and DO-13 are monoclonal antibodies while CM-1 is a rabbit polyclonal antibody specific to p53.
  • the V5 and 9E10 epitopes are recognised by the mouse monoclonal antibodies V5 and 9E10 respectively.
  • the mouse monoclonal PC-10 is specific to the PCNA protein.
  • CD20Leu is an FITC conjugated monoclonal antibody specific for the cell surface marker CD20 (Becton Dickinson). The mouse and rabbit antibodies to ASPP1 and ASPP2 were described previously 1 .
  • iASPP peptide RLQPALPPEAQSVPELEE
  • All expression plasmids used in this study were driven by the CMV immediate early promoter.
  • ASPP1, iASPP and Ce-iASPP are tagged with V5 epitope while Ce-p53 is tagged with 9E10 epitope.
  • the transfection mix included the DNA of interest in 1 ⁇ HBS buffer (280 mM NaCl, 10 mM KCl, 1.4 mM Na 2 HPO 4 .2H 2 O, 12 mM Glucose, 39 mM HEPES, adjusted to pH 6.9-7.3) precipitated with 2.5 M CaCl 2 .
  • the transfection mix was added dropwise to the cells and washed off after 6 hours with DMEM. 16-24 hours following the wash, cells were lysed either in Reporter lysis buffer (Promega) for Luciferase assays and western blots or in NP40 lysis buffer for western or immunoprecipitation procedures.
  • the cells were lysed in Reporter Lysis Buffer (Promega, WI, USA) 16-24 hr post-wash and assayed using the Luciferase Assay kit (Promega, WI, USA).
  • the fold activation of a particular reporter was determined by the activity of the transfected plasmid above the activity of vector alone.
  • the fold increase of p53 transactivation activity by ASPP was obtained by the activity p53 in combination with ASPP divided by the activity of p53 alone on the promoters used in each assay.
  • Rat Embryo Fibroblasts obtained from Biowhittaker were grown in DMEM to 50% confluence in a 90 mm dish. Cells were then transformed as described previously 14 . Briefly, 2 ⁇ g of EJ ras 6.6, 2 ⁇ g of pCE (E1A), 5 ⁇ g of pCB6-16E, 5 ⁇ g of wild type human p53, 1 ⁇ g or 5 ⁇ g of human or C. elegans iASPP were transfected into the REFS as indicated. All the cells were transfected with the same amount of plasmid DNA expressing neo gene. The transfected cells were then selected with 400 ⁇ g/ml of G418 and the morphologically transformed colonies were scored after 3-4 weeks after transfection.
  • both attached and floating cells were harvested using 4 mM EDTA/PBS and stained with FITC conjugated anti-CD20 antibody CD20Leu.
  • one dish of cells was transfected with the control vector only without CD20. These cells were later stained with antibody CD20Leu under the same conditions as those co-transfected with CD20 plasmid to serve as a negative control.
  • the cells lacking expression of CD20 plasmid were used to set the base line to allow the gating of the CD20 positive (and hence transfected) cells.
  • the cells were fixed and stained with propidium iodide. The DNA content of all the cells expressing CD20 was analysed using the flow cytometer (Becton Dickson) as described 15 .
  • NP40 lysis buffer 1% Nonidet P40, 50 mM Tris pH8.0, 150 mM NaCl, 1 mM EDTA pH 8.0
  • luciferase reporter lysis buffer 1% Nonidet P40, 50 mM Tris pH8.0, 150 mM NaCl, 1 mM EDTA pH 8.0
  • the protein concentration of the cell extracts was determined against a standard curve using the BioRad protein assay system (BioRad). Between 15-100 ⁇ g of extract was mixed with 5 ⁇ sample buffer and loaded on SDS-PAGE gels. The gels were wet transferred on to Protran nitrocellulose membrane and the resulting blots blocked in 10% reconstituted milk powder in 1 ⁇ PBS.
  • the blots were incubated with primary antibody prepared either in tissue culture medium or as undiluted hybridoma supernatant and incubated with the appropriate secondary HRP conjugated antibody (Dako). In between each stage the blot was washed with repeat changes of TBST (10 mM Tris pH 8.0, 150 mM NaCl, 0.5% Tween 20). The blot was exposed to hyperfiln following the use of ECL substrate solution (Amersham Life Science).
  • NP40 lysis buffer For immunoprecipitation cells were lysed in NP40 lysis buffer on ice for 30 minutes and pre-cleared with protein G beads for 1 hour at 4 C. The protein concentration was determined and 1000 ⁇ g of the extract was incubated with antibody pre-bound to protein G beads for 4 hours at 4 C. The beads were washed twice in NP40 lysis buffer and twice in NET buffer (50 mM Tris pH 8.0, 150 mM NaCl, 1 mM EDTA pH 8.0). The IP beads were mixed with 5 ⁇ sample buffer and loaded onto a SDS-PAGE gel.
  • ASPP family members and p53 were in vitro translated and labelled with 35 S—Methionine using the TNT T7 Quick coupled Transcription/Translation System (Promega).
  • TNT T7 Quick coupled Transcription/Translation System Promega.
  • 30 and 45 ⁇ l of in vitro translated lysates of iASPP were added in addition to p53 and ASPP2.
  • Rabbit anti-p53 antibody CM1 was used to detect the presence of unlabelled p53.
  • the bound proteins were released in SDS gel sample buffer and analysed by 10% SDS-polyacrylamide gel electrophoresis (PAGE). Ce-p53 was detected by autoradiography and Human iASPP was detected by anti-V5 antibody following a western blot. For the rest of the figures, the proteins were labelled with 35 S -Methionine and immunoprecipitated as above using the indicated antibodies. Results were visualised using autoradiography.
  • U20S cells in the absence or presence of transfected plasmids were washed with PBS and then incubated with 250 ⁇ ci/ml of 35 S—methionine and 250 ⁇ ci of 35-cysteine in DMEM lacking both methionine and cysteine for 2 hours at 37C. Cells were then washed with PBS before harvesting. For FIG. 2C , twenty four hours after transfection the cells were in vivo labelled with 35 S-methionine and 35 S-cysteine for 2hrs. Cells expressing CD20 (transfected cells) were stained with FITC-conjugated anti-CD20 antibody.
  • a biotin conjugated anti-FITC antibody was then added to the cell pellet, and after the incubation, the cells were mixed with streptavidin conjugated magnetic beads to isolate the CD20 expressing cells.
  • the cells were then lysed with NP40 lysis buffer and the proteins immunoprecipitated with mouse anti iASPP antibody.
  • the immunoprecipitates were washed with NET buffer and resolved by SDS-PAGE and autoradiography.
  • Monolayers of cells were grown in 30 mm dishes and washed with 1 ⁇ PBS. Cells were fixed with 1 ml of 4% para-formaldehyde for 15 minutes and then washed in 1 ⁇ PBS. 1 ml of 0.2% Triton-X100 in 1 ⁇ PBS was used to permeabilise the cells for 2 minutes and this was washed off with three washes of 1 ⁇ PBS. Primary antibody was prepared in tissue culture medium at the appropriate concentration and added to the dishes for 3 hours.
  • the dishes were washed with 1 ⁇ PBS and the secondary antibody of either anti-rabbit TRITC (Tetramethyl rhodamine isothiocyanate) or anti-mouse FITC (Fluorescein isothiocyanate) prepared in tissue culture medium at the manufacturers recommended dilution (Sigma, UK) and added to the dishes for one hour.
  • the cells were washed in 1 ⁇ PBS and left to air dry.
  • Citifluor shielding agent (Citifluor, UK) was applied as a drop to the surface of the cells and a cover slip placed on top. A drop of immersion oil on top of the cover slip allowed the immunocomplexes to be visualised using a Zeiss Axiophot fluorescence microscope.
  • Antibodies 9E10 and V5 were used to detect the expression of epitope tagged Ce-p53 (9E10), human iASPP (V5) and Ce-iASPP (V5) respectively. Human p53 was detected by DO.1 antibody.
  • cDNAs carrying the complete coding regions of Ce-ape-1 (iASPP) and Ce-cep-1 (p53) were generated by RT-PCR using the Promega Access kit, cloned into the vector pCR4-TOPO (Invitrogen) and sequenced. C. elegans p53 and iASPP were then subcloned into a mammalian expression vector pcDNA3 in frame with the epitope of 9E10 and V5 respectively. The full-length Ce-ape-1 is predicted to be trans-spliced to SL1 (Y. Kohara, unpublished).
  • RNAi was performed by feeding or microinjection using established procedures (Fire et al., 1998; Timmons and Fire, 1998)
  • Ce-iASPP RNAi was first subjected to Ce-iASPP RNAi feeding.
  • 20 F1 animals were removed from the feeding plate, injected with Ce-p53 dsRNA and returned to independent Ce- iASPP dsRNA feeding plates.
  • the F2 progeny of animals fed Ce-iASPP (+/ ⁇ injection of Ce-p53 dsRNA) were stained with SYTO12 and apoptotic corpses scored as described12.
  • Ce-iASSP feeding RNAi any differences in the mean number of apoptotic cell corpses detected in the two populations are likely to result from Ce-p53 dsRNA injection.
  • iASPP is the Most conserveed Member of the ASPP Family
  • Ce-iASPP consists of 769 amino acids; sequence comparisons reveal that the C-terminus of Ce-iASPP is the region most conserved with other ASPP members ( FIG. 7 ). It was previously shown that the C-terminus of ASPP2 interacts with p53. Moreover, all but one (6 out of 7) residue involved in this interaction are conserved in both iASPP and Ce-iASPP. Taken together these results suggested that Ce-iASPP might interact with both human and C. elegans p53. This was tested in vitro by co-immunoprecipitation. As shown in FIG. 8B , Ce-iASPP interacts with both human and Ce-p53. The interaction between Ce-p53 and Ce-iASPP was further confirmed by reciprocal immunoprecipitation ( FIG. 8B , right panel).
  • Ce-iASPP contains the hallmark ankyrin repeats and SH3 domain found in other ASPP family members and is expressed in the cytoplasm and nucleus of human cells ( FIG. 8C ).
  • the expression of human iASPP is predominantly nuclear but cytoplasmic staining is also detectable.
  • Both human and C. elegans p53 are primarily expressed in the nucleus of transfected human cells ( FIG. 8C ). Since ASPP and iASPP can positively and negatively regulate the apoptotic function of p53, respectively, we tested the effect of Ce-iASPP on the activities of p53.
  • Ce-iASPP When Ce-iASPP was co-expressed with human p53 in mammalian cells, it produced a small reduction in the transactivation and apoptotic function of p53, presumably by inhibiting endogenous ASPP function. However, in the presence of ASPP1 or ASPP2, co-expression of Ce-iASPP prevented ASPP1 or ASPP2 from stimulating the transactivation and apoptotic function of p53 to the same extent as human iASPP ( FIG. 8D ). Furthermore, the ability of human and C. elegans iASPP to inhibit apoptosis is p53-dependent since they both failed to inhibit Bax induced apoptosis under the same conditions ( FIG. 8E ).
  • iASPP The ability of iASPP to inhibit the activity of p53 is not due to the reduced expression of p53 ( FIG. 8F ). Like human iASPP, the Ce-iASPP also has oncogenic activity. The expression of Ce-iASPP enhanced the transforming activity of ras and E1A. Moreover, expression of Ce-iASPP inhibited the suppressor function of wild type human p53 ( FIG. 8G ). These results demonstrate that Ce-iASPP is more likely to be an orthologue of human iASPP than of ASPP. Also, the ability of Ce-iASPP to inhibit p53 suggests that the regulation of p53 apoptotic function by the ASPP family of proteins is evolutionarily conserved.
  • Ce-p53 could induce apoptosis in mammalian cells because of the limited sequence similarity between human and C. elegans p53, although most of the Ce-p53 residues that contact ASPP are conserved. If Ce-iASPP inhibits the activities of human p53 in a manner similar to that of human iASPP, this argues that the regulation of p53 by the ASPP family has been evolutionarily conserved. This further suggests that the activities of Ce-p53 could be subject to regulation by the ASPP family of proteins. To address these issues, Ce-p53 was tested for its ability to interact in vitro with members of the human ASPP family by co-immunoprecipitation. As shown in FIG.
  • Ce-p53 interacts with ASPP2 and iASPP.
  • the expression of Ce-p53 induced apoptosis in human cells with an efficiency similar to human p53.
  • expression of human ASPP, ASPP2 in particular, significantly enhanced the ability of Ce-p53 to induce apoptosis to an extent similar to human p53 and the expression of human iASPP also inhibited the apoptotic function of Ce-p53 ( FIG. 6B ).
  • both human and C. elegans iASPP inhibited Ce-p53 induced apoptosis to the same extent ( FIG. 6C ).
  • Ce-p53 to transactivate p53 target gene promoters, such as Bax-luc, was also tested and was found to be much lower than that of human p53.
  • co-expression of ASPP2 and Ce-p53 resulted in a very small but detectable increase in the transactivation function of Ce-p53, indicating that the human ASPP family could regulate Ce-p53 in a manner similar to that of human p53 ( FIG. 6D ).
  • the small increase in the transactivation function of Ce-p53 by ASPP2 is not observed on the mdm2 promoter. All of these results suggest that the residues conserved between human and C. elegans p53 are crucial and sufficient for the apoptotic function of p53 and for p53 to be regulated by the ASPP family.
  • iASPP is an Evolutionarily conserveed Inhibitor of p53 In Vivo
  • RNA mediated interference RNAi 5 . Depletion of endogenous Ce-iASPP increased the number of germ cells undergoing apoptosis, indicating that the normal function of Ce-iASPP is to inhibit apoptosis ( FIG.
  • FIG. 9A compare lanes 5 , 6 and 8 .
  • the depletion of both Ce-iASPP and Ce-p53 by RNAi did not completely eliminate apoptosis, but instead returned the number of germ cells undergoing apoptosis to wild-type physiological levels.
  • the increase in the number of apoptotic germ cell corpses detected after wild type worms were exposed to 100-Gy IR was no greater than that observed after depletion of Ce-iASSP by RNAi in the presence or absence of exposure to 100-Gy IR ( FIG.
  • iASPP is the most phylogenetically conserved inhibitor of p53, so far identified, and also the most evolutionarily conserved member of the ASPP family.
  • ASPP family members to regulate the apoptotic function of p53 has been conserved between C. elegans and human. This argues that the apoptotic function of p53 is likely to be more conserved than its ability to induce cell cycle arrest; this agrees with recent observations showing that ectopic expression of both C. elegans and Drosophila p53 induces apoptosis but not cell cycle arrest 3,4,7,8 .
  • C. elegans ectopic expression of both C. elegans and Drosophila p53 induces apoptosis but not cell cycle arrest 3,4,7,8 .
  • elegans p53-mediated apoptosis appears to play an important role in maintaining the fidelity of germ cells, which might have incurred DNA damage 3,4 .
  • the most important tumour suppressor function of p53 is also linked to its ability to induce apoptosis. Therefore, ASPP family members, the evolutionarily conserved regulators of p53, must play a critical role in tumourigenesis.
  • iASPP shares more sequence similarity with the N-terminally truncated ASPP2 mutant 53BP2 than the full-length ASPP. Expression of iASPP inhibited the apoptotic function of p53. Like 53BP2, the most profound effect of iASPP on the apoptotic function of p53 is mediated through its ability to act as a competitor of ASPP. However, in C. elegans, iASPP is the only gene that has homology to the human ASPP family. Thus iASPP directly inhibits the apoptotic function of p53 in C. elegans. A similar mechanism might also apply in mammalian cells.
  • iASPP antisense RNA induced a 3 to 5-fold increase in apoptotic cells in U2OS and MCF7 cells.
  • ASPP could stimulate the apoptotic function of p53 by removing the negative effects that iASPP imposed on p53.
  • the failure of iASPP antisense RNA to produce a significant increase of apoptosis in cisplatin treated U2OS and MCF7 cells might be due to the fact that cisplatin stimulates the apoptotic function of p53 by increasing the activities of ASPP. This, in turn, might explain why the expression of ASPP antisense RNA generated a profound inhibitory effect on apoptosis induced by cisplatin.
  • iASPP enhanced the transforming activities of oncogenes such as ras plus E7 or E1A of human papilloma virus and adenovirus but not ras plus mutant p53.
  • oncogenes such as ras plus E7 or E1A of human papilloma virus and adenovirus but not ras plus mutant p53.
  • E7 and E1A are known to induce p53-dependent apoptosis 9 .
  • E7 and E1A can bind and inactivate the tumour suppressor function of Rb, their oncogenic function is largely reduced due to their ability to activate p53-dependent apoptosis. Proteins that can inhibit apoptosis induced by E7 and E1A would enhance the oncogenic function of E7 and E1A.
  • iASPP was able to stimulate the oncogenic function of E7 and E1A by inhibiting the apoptotic function of p53. It is important to point out that iASPP was not as active as mutant p53, p53H175 or p53L173 in co-operating with ras to transform REFs under the experimental conditions described here. Part of the reason was due to the low expression level of iASPP in the assay (data not shown). However, the differences in the transforming activities of iASPP and mutant p53 might also be caused by other known activities of mutant p53, which are independent of its ability to act as a simple dominant negative inhibitor of p53.
  • iASPP is an inhibitor of ASPP in vivo.
  • Our previous study showed that the expression levels of ASPP1 and ASPP2 were down regulated in 60% of human breast tumours express wild type p53. Taken together, the abnormal expression of ASPP family members would account for almost 80% of human breast carcinomas examined.
  • the ASPP family members are encoded by three different genes located on different chromosomes (data not shown). We do not know why the frequency of human breast carcinomas showing down regulation of ASPP is much higher than those showing increased expression of iASPP. However, it is possible that the expression pattern of the ASPP family members varies in different types of human tumours. The percentage of tumours with altered ASPP expression could also differ in various tumour types. Nevertheless, inhibiting the oncogenic function of iASPP could provide an important new strategy to treat tumours expressing wild type p53.
  • C. elegans p53 induces apoptosis very effectively in human cells. Similar to human p53, the apoptotic function of C. elegans p53 is positively and negatively regulated by the human ASPP and iASPP, respectively. These results demonstrate for the first time that the apoptotic function of p53 is conserved despite the limited sequence homology between human and C. elegans p53. The few key residues conserved between human and C. elegans p53 are sufficient for ASPP family members to regulate the apoptotic function of p53 both in vitro and in vivo.
  • the C. elegans p53 showed little ability to transactivate human p53 target genes such as Bax and PIG3 in comparison to human p53 (data not shown). Even though Bax is frequently a p53 target gene during apoptosis, it is not the only target gene that is required for p53 induced apoptosis. p53 is known to transactivate over 20 pro-apoptotic genes none of which has so far proved to be indispensable in p53 induced apoptosis. There are over 4000 putative p53 target genes in the human genome and many of them are pro-apoptotic genes 11 . It is possible that Ce-p53 might transactivate some of these other human p53 target genes as effectively as human p53.
  • Ce-p53 induces apoptosis in human cells
  • the most remarkable and important fact is that the human ASPP family of proteins regulate the apoptotic function of both Ce-p53 and human p53 in a similar way.
  • Ce-iASPP is also able to completely replace human iASPP in all the assays performed in human cells.
  • Chemotherapy Drugs Appear to Modify ASPP Family Protein Function
  • ASPP1 and ASPP2 specifically enhance the apoptotic function of p53 by stimulating the DNA binding and transactivation function of p53 on the promoters of proapoptotic genes (Samuels-Lev et al., 2001), while iASPP is a very conserved inhibitor of p53 (Bergamaschi et al., 2003). Taken together, this suggests that the function of the ASPP family of proteins may be affected by chemotherapy agents, which in turn vary the subsequent p53 apoptotic response, and thus play a role in the effectiveness of drugs used in cancer treatment.
  • Chemotherapy agents were used to investigate whether the apoptosis induced in cell lines correlated directly with p53 expression levels.
  • U-2OS and MCF7 cells were transfected with the Pig3 promoter (Pig3-luc 17 mer) and treated with various chemotherapy drugs (doxorubicin, cisplatin, etoposide, colchicine, deferoxamine mesylate (DFO), daunorubicin, camptothecin and 5′fluorouracil).
  • various chemotherapy drugs doxorubicin, cisplatin, etoposide, colchicine, deferoxamine mesylate (DFO), daunorubicin, camptothecin and 5′fluorouracil.
  • Luciferase levels were assayed 24 hours later, FIG. 13 .
  • the results illustrate that drug-induced apoptosis, as measured by luciferase, did not correlate directly with p53 protein levels (for example, compare doxorubicin and cisplatin responses) indicating that p53 alone is not responsible for the activation of proapoptotic genes. It is possible that the ASPP proteins respond differently to the various chemotherapy drugs to give the discrepancy observed between Pig3 activation and p53 protein expression level.
  • si-RNA short interfering RNA
  • antisense RNA was used to knock down ASPP gene expression. Sequences to generate si-RNA were cloned into pSuper plasmid (Brummelkamp et al., 2002). Since responses of proapoptotic genes differed depending on the drug used, both doxorubicin and cisplatin were tested in U-2OS cells. Pig3-luciferase was transfected together with either antisense or si-iASPP ( FIGS. 14A and 14B ).
  • antisense ASPP1 or antisense ASPP2 both reduce the induction of Pig3-luciferase by doxorubicin ( FIG. 14A ) but antisense or si-RNA for iASPP caused no reduction.
  • a si-RNA for p53 prevented the doxorubicin response ( FIG. 14A ).
  • antisense ASPP1 or antisense ASPP2 had little effect on the cisplatin induction of the Pig3 promoter but the si-RNA for iASPP caused a significant increase. Again siRNA for p53 prevented the cisplatin response.
  • FIG. 10 It is known that p53 binds to the C terminal part of iASPP; specific contact amino acids are indicated in FIG. 10 .
  • Three different peptides containing particular iASPP sequences were designed to inhibit the iASPP/p53 binding and test the effects on proapoptotic genes, FIG. 10 .
  • the three peptides were linked to a Tat sequence and to FITC.
  • the peptides were initially tested for their effects on p53 transactivation of the Bax promoter using UVradiation to activate p53, FIG. 11 .
  • the luciferase activity of the Bax promoter was only slightly affected by the control peptide, which should not affect iASPP & p53 interaction.
  • addition of peptide 3, 6 or 7 caused about a two fold increase in the reporter activity, using 50 ⁇ M peptide.
  • Some peptides were less active at 100 ⁇ M.
  • the Pig3 gene is transactivated by p53 in a different way to Bax.
  • Bax is transactivated by a conventional transcription domain (El-Deiry et al., 1992) (Bourdon et al., 1997)
  • the number of repeats is polymorphic within populations (Contente et al., 2002).
  • Two reporters containing 10 or 17 repeats were used in case there was a difference in response between them.
  • Other variables to consider are time and concentration of peptide and the location of the peptides in the cells since p53 and iASPP are mostly nuclear proteins.
  • a second generation of peptides was used for this experiment, which are identical to the previous ones except for the tag. These peptides were tagged with nine arginine residues, which are thought to give the peptides better penetration into cells (Lindsay, 2002). In this case a peptide 7 from the ASPP2 sequence was included (see FIG. 10 ) and a different control peptide (Yap) was used. Cells were treated with cisplatin and peptides were applied for 12 hours at two different concentrations (25 uM and 50 uM) and the results are shown in FIG. 15 .

Abstract

The invention relates to a polypeptide, or part thereof, which inhibits the apoptotic activity of the tumour suppressor protein p53 and including screening methods to identify agents which interfere with the activity of said polypeptide.

Description

  • The invention relates to a polypeptide, or part thereof, which inhibits the apoptotic activity of the tumour suppressor protein p53; screening methods to identify agents which interfere with the activity of said polypeptide and agents with said activity.
  • Apoptosis, or programmed cell death, is a process by which multi-cellular organisms regulate cell number and differentiation. The process is regulated by factors which either induce or prevent apoptosis. Inducers of apoptosis include Bcl-2 family members, caspase family members and their associated factors Apaf-1 and Fadd. Caspases are synthesised as proenzymes which become activated after proteolytic cleavage. The active caspase then induces many of the morphological and biochemical changes associated with apoptosis. Mitochondria play a pivotal role in the activation process through the release of pro-apoptotic factors such as cytochrome c, AIF and Diablo. The release from mitochondria is controlled by the Bcl-2 family of proteins; (e.g. Bcl-2 and Bcl-xl inhibit release; Bax and Bak induce release).
  • WO9953051 discloses a cytokine dependent protein p21 which has pro-apoptotic activity. p21 is expressed in a cytokine dependent manner in myeloid/erthyroid cells. These cells are dependent on IL-3 for growth and in the absence of IL-3 the translation of p21 is induced resulting in apoptosis and cell death. p21 is a cytoplasmic protein which translocates to the outer mitochondrial membrane to induce pro-apoptotic activities.
  • Tumour suppressor proteins also have pro-apoptotic activities.
  • Tumour suppressor genes encode proteins which function to inhibit cell growth or division and are therefore important with respect to maintaining proliferation, growth and differentiation of normal cells. Mutations in tumour suppressor genes result in abnormal cell-cycle progression whereby the normal cell-cycle check points which arrest the cell-cycle, when, for example, DNA is damaged, are ignored and damaged cells divide uncontrollably. The products of tumour suppressor genes function in all parts of the cell (e.g. cell surface, cytoplasm, nucleus) to prevent the passage of damaged cells through the cell- cycle (i.e. G1, S, G2, M and cytokinesis).
  • Arguably the tumour suppressor gene which has been the subject of the most intense research is p53. p53 encodes a protein which functions as a transcription factor and is a key regulator of the cell division cycle. It was discovered in 1978 as a protein shown to bind with affinity to the SV40 large T antigen. The p53 gene encodes a 393 amino acid polypeptide with a molecular weight of 53 kDa. Genes regulated by the transcriptional activity of p53 contain a p53 recognition sequence in their 5′ regions. These genes are activated when the cellular levels of p53 are elevated due to, for example DNA damage. Examples of genes which respond to p53 include, mdm2, Bax and PIG-3. Bax and PIG-3 are involved in one of the most important functions of p53, the induction of apoptosis.
  • In our co-pending application WO02/12325 we disclose a family of proteins, referred to as ASPP, as specific activators of p53 and revealed a mechanism by which wild type p53 is tolerated in tumours, such as human breast carcinomas. We also disclose an inhibitor of ASPP family members referred to as iASPP. iASPP is an oncogene and is the most conserved member of the ASPP family. iASPP is the only ASPP-like protein found in C. elegans. Similar to human iASPP, the C. elegans homologue functions as a key inhibitor of p53. These findings indicate that regulation of p53 function by members of the ASPP family has been evolutionarily conserved across phyla.
  • The C. elegans iASPP is capable of substituting for human iASPP in all of the assays performed in human cells. Moreover, reciprocal substitution studies reveal that the apoptotic function of C. elegans p53 is enhanced or inhibited by human ASPP and iASPP, respectively. Using RNAi we further demonstrate that iASPP is a key inhibitor of p53 mediated apoptosis in C. elegans. All of these observations show that the regulation of p53 by ASPP family members is evolutionarily conserved. Control of p53 activity plays a pivotal role in development and tumourigenesis. Hence, inhibiting the oncogenic function of iASPP could provide an important new strategy for treating tumours expressing wild type p53. Sequence comparision between C. elegans and human iASPP reveals a conserved domain between the nematode and human sequence which likely explains the functional conservation between the proteins.
  • According to an aspect of the invention there is provided an isolated nucleic acid molecule which encodes a polypeptide, or sequence variant thereof, wherein said polypeptide is a fragment of the polypeptide sequence represented in FIG. 1 a or 1 b, which fragment is selected from the group consisting of:
      • i) a polypeptide fragment consisting of amino acid residues from about residue 128-224 of the amino acid sequence presented in FIG. 1 a or 1 b;
      • ii) a polypeptide fragment consisting of amino acid residues from about 128-244 of the amino acid sequence presented in FIG. 1 a or 1 b wherein said sequence has been modified by addition, deletion or substitution of at least one amino acid residue; and
      • iii) a polypeptide as defined in (i) and (ii) wherein said polypeptide substantially retains the biological activity of the polypeptide represented in FIG. 1 a or 1 b.
  • In a preferred embodiment of the invention said nucleic acid molecule encodes a polypeptide fragment consisting of amino acid residues from about 128-224 of the sequence represented in FIG. 1 a. Preferably said nucleic acid molecule is isolated from a human.
  • In an alternative preferred embodiment of the invention said nucleic acid molecule encodes a polypeptide fragment consisting of amino acid residues from about 128-224 of the sequence represented in FIG. 1 b. Preferably said nucleic acid molecule is isolated from a nematode. Preferably said nematode is of the genus Caenorhabditis spp.
  • In a preferred embodiment of the invention said nucleic acid molecule encodes a polypeptide, or sequence variant thereof, which polypeptide inhibits the activity of a polypeptide represented by the amino acid sequence represented in FIG. 1 a or 1 b.
  • In a preferred embodiment of the invention said nucleic acid molecule is a cDNA.
  • In an alternative preferred embodiment of the invention said nucleic acid molecule is genomic DNA.
  • According to a further aspect of the invention there is provided a polypeptide fragment or sequence variant thereof, encoded by a nucleic acid molecule according to the invention.
  • It will be apparent that fragments which are sequence variants may retain the biological activity of the full length polypeptide or alternatively have antagonistic activity by competing for binding sites in p53. In general, the specificity of polypeptides according to the invention with respect to binding to p53 is shown by binding equilibrium constants. Polypeptides which are capable of selectively binding p53 preferably have binding equilibrium constants of at least about 107 M−1, more preferably at least about 108 M−1, and most preferably at least about 109 M−1.
  • A sequence variant, i.e. a fragment polypeptide and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, truncations which may be present in any combination. Among preferred variants are those that vary from a reference polypeptide by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid by another amino acid of like characteristics. The following non-limiting list of amino acids are considered conservative replacements (similar): a) alanine, serine, and threonine; b) glutamic acid and asparatic acid; c) asparagine and glutamine d) arginine and lysine; e) isoleucine, leucine, methionine and valine and f) phenylalaine, tyrosine and tryptophan.
  • A functionally equivalent polypeptide according to the invention is a variant wherein one or more amino acid residues are substituted with conserved or non-conserved amino acid residues, or one in which one or more amino acid residues includes a substituent group. Conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu and Ile; interchange of the hydroxl residues Ser and Thr; exchange of the acidic residues Asp and Glu; substitution between amide residues Asn and Gln; exchange of the basic residues Lys and Arg; and replacements among aromatic residues Phe and Tyr.
  • In addition, the invention features polypeptide sequences having at least 75% identity with the polypeptide sequences as hereindisclosed, or fragments and functionally equivalent polypeptides thereof. In one embodiment, the polypeptides have at least 85% identity, more preferably at least 90% identity, even more preferably at least 95% identity, still more preferably at least 97% identity, and most preferably at least 99% identity with the amino acid sequences illustrated herein.
  • As mentioned above, the invention also provides, in certain embodiments, “dominant negative” polypeptides derived from the polypeptides hereindisclosed. A dominant negative polypeptide is an inactive variant of a protein, which, by interacting with the cellular machinery, displaces an active protein from its interaction with the cellular machinery or competes with the active protein, thereby reducing the effect of the active protein. For example, a dominant negative receptor which binds a ligand but does not transmit a signal in response to binding of the ligand can reduce the biological effect of expression of the ligand. Likewise, a dominant negative catalytically-inactive kinase which interacts normally with target proteins but does not phosphorylate the target proteins can reduce phosphorylation of the target proteins in response to a cellular signal. Similarly, a dominant negative transcription factor which binds to another transcription factor or to a promoter site in the control region of a gene but does not increase gene transcription can reduce the effect of a normal transcription factor by occupying promoter binding sites without increasing transcription.
  • The end result of the expression of a dominant negative polypeptide in a cell is a reduction in function of active proteins. One of ordinary skill in the art can assess the potential for a dominant negative variant of a protein, and using standard mutagenesis techniques to create one or more dominant negative variant polypeptides. For example, given the teachings contained herein of iASPP polypeptides, one of ordinary skill in the art can modify the sequence of iASPP polypeptides by site-specific mutagenesis, scanning mutagenesis, partial gene deletion or truncation, and the like. See, e.g., U.S. Pat. No. 5,580,723 and Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, 1989. The skilled artisan then can test the population of mutagenized polypeptides for diminution in a selected activity (e.g., p53 binding, modulation of apoptosis) and/or for retention of such an activity. Other similar methods for creating and testing dominant negative variants of a protein will be apparent to one of ordinary skill in the art.
  • According to a further aspect of the invention there is provided a vector comprising a nucleic acid according to the invention.
  • In a further preferred method of the invention said vector is an expression vector conventionally adapted for gene expression.
  • Typically said adaptation includes, by example and not by way of limitation, the provision of transcription control sequences (promoter sequences) which mediate cell/tissue specific expression. These promoter sequences may be cell/tissue specific, inducible or constitutive.
  • Promoter is an art recognised term and, for the sake of clarity, includes the following features which are provided by example only, and not by way of limitation. Enhancer elements are cis acting nucleic acid sequences often found 5′ to the transcription initiation site of a gene (enhancers can also be found 3′ to a gene sequence or even located in intronic sequences and is therefore position independent). Enhancers function to increase the rate of transcription of the gene to which the enhancer is linked. Enhancer activity is responsive to trans acting transcription factors (polypeptides) which have been shown to bind specifically to enhancer elements. The binding/activity of transcription factors (please see Eukaryotic Transcription Factors, by David S Latchman, Academic Press Ltd, San Diego) is responsive to a number of environmental cues which include, by example and not by way of limitation, intermediary metabolites (eg glucose, lipids), environmental effectors (eg light, heat,).
  • Promoter elements also include so called TATA box and RNA polymerase initiation selection (RIS) sequences which function to select a site of transcription initiation. These sequences also bind polypeptides which function, inter alia, to facilitate transcription initiation selection by RNA polymerase.
  • Adaptations also include the provision of selectable markers and autonomous replication sequences which both facilitate the maintenance of said vector in either the eukaryotic cell or prokaryotic host. Vectors which are maintained autonomously are referred to as episomal vectors. Episomal vectors are desirable since these molecules can incorporate large DNA fragments (30-50 kb DNA). Episomal vectors of this type are described in WO98/07876.
  • Adaptations which facilitate the expression of vector encoded genes include the provision of transcription termination/polyadenylation sequences. This also includes the provision of internal ribosome entry sites (IRES) which function to maximise expression of vector encoded genes arranged in bicistronic or multi-cistronic expression cassettes.
  • Expression control sequences also include so-called Locus Control Regions (LCRs). These are regulatory elements which confer position-independent, copy number-dependent expression to linked genes when assayed as transgenic constructs in mice. LCRs include regulatory elements that insulate transgenes from the silencing effects of adjacent heterochromatin, Grosveld et al., Cell (1987), 51: 975-985.
  • These adaptations are well known in the art. There is a significant amount of published literature with respect to expression vector construction and recombinant DNA techniques in general. Please see, Sambrook et al (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbour Laboratory, Cold Spring Harbour, N.Y. and references therein; Marston, F (1987) DNA Cloning Techniques: A Practical Approach Vol III IRL Press, Oxford UK; DNA Cloning: F M Ausubel et al, Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (1994).
  • According to a further aspect of the invention there is provided a cell transformed or transfected with a nucleic acid molecule or vector according to the invention.
  • Preferably, said host cells are eukaryotic cells, for example, insect cells such as cells from a species Spodoptera frugiperda using a baculovirus expression system. This expression system is favoured in the instance where post-translational modification of the polypeptide is required. Host cells and cell lines, can be prokaryotic (e.g., E. coli), or eukaryotic (e.g., CHO cells, COS cells, yeast expression systems and recombinant baculovirus expression in insect cells). Especially useful are mammalian cells such as human, mouse, hamster, pig, goat, primate, etc. They may be of a wide variety of tissue types, and include primary cells and cell lines. Specific examples include keratinocytes, peripheral blood leukocytes, fibroblasts, bone marrow stem cells and embryonic stem cells. The expression vectors require that the pertinent sequence, i.e., those nucleic acids described above, be operably linked to a promoter.
  • According to a further aspect of the invention the invention there is provided a polypeptide according to the invention for use as a pharmaceutical.
  • According to a further aspect of the invention there is provided a nucleic acid according to the invention for use as a pharmaceutical.
  • In a preferred embodiment of the invention said pharmaceutical further comprises a a diluent, carrier or excipient.
  • When administered, the therapeutic 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, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents, such as chemotherapeutic agents.
  • The therapeutics of the invention can be administered by any conventional route, including injection or by gradual infusion over time. The administration may, for example, be oral, intravenous, intraperitoneal, intramuscular, intracavity, subcutaneous, or transdermal. When antibodies are used therapeutically, a preferred route of administration is by pulmonary aerosol. Techniques for preparing aerosol delivery systems containing antibodies are well known to those of skill in the art. Generally, such systems should utilize components which will not significantly impair the biological properties of the antibodies, such as the paratope binding capacity (see, for example, Sciarra and Cutie, “Aerosols,” in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp 1694-1712; incorporated by reference). Those of skill in the art can readily determine the various parameters and conditions for producing antibody aerosols without resort to undue experimentation. When using antisense preparations of the invention, slow intravenous administration is preferred.
  • 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 or can be monitored according to diagnostic methods of the invention discussed herein.
  • 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 dominant negative iASPP or nucleic acid encoding a dominant negative iASPP, for producing the desired response in a unit of weight or volume suitable for administration to a patient. The response can, for example, be measured by determining the signal transduction inhibited by the dominant negative iASPP-1, composition via a reporter system as described herein, by measuring downstream effects such as gene expression, or by measuring the physiological effects of the iASPP composition, such as regression of a tumour, decrease of disease symptoms, modulation of apoptosis, etc.
  • The doses of dominant negative iASPP polypeptide or nucleic acid 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 dominant negative iASPP are formulated and administered in doses between 1 ng and about 500 mg, and between 10 ng and 100 mg, according to any standard procedure in the art. Where nucleic acids encoding dominant negative iASPP are employed, doses of between 1 ng and 0.1 mg generally will be formulated and administered according to standard procedures. Other protocols for the administration of iASPP 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 (e.g., intra-tumoral) and the like vary from the foregoing. Administration of iASPP 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. 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.
  • iASPP 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” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. 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 a syrup, elixir or an emulsion.
  • Compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of iASPP polypeptides or nucleic acids, 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 non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol. Among the acceptable vehicles and 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.
  • According to a further aspect of the invention there is provided a transgenic non-human animal comprising a nucleic acid according to the invention.
  • The invention also includes transgenic non-human animals. As used herein, “transgenic non-human animals” includes non-human animals having one or more exogenous nucleic acid molecules incorporated in germ line cells and/or somatic cells. Thus the transgenic animal include “knockout” animals having a homozygous or heterozygous gene disruption by homologous recombination, animals having episomal or chromosomally incorporated expression vectors, etc. Knockout animals can be prepared by homologous recombination using embryonic stem cells as is well known in the art. The recombination can be facilitated by the cre/lox system or other recombinase systems known to one of ordinary skill in the art. In certain embodiments, the recombinase system itself is expressed conditionally, for example, in certain tissues or cell types, at certain embryonic or post-embryonic developmental stages, inducibly by the addition of a compound which increases or decreases expression, and the like. In general, the conditional expression vectors used in such systems use a variety of promoters which confer the desired gene expression pattern (e.g., temporal or spatial). Conditional promoters also can be operably linked to iASPP family nucleic acid molecules to increase expression of these nucleic acid molecules in a regulated or conditional manner. Trans-acting negative regulators of iASPP activity or expression also can be operably linked to a conditional promoter as described above. Such trans-acting regulators include antisense nucleic acids molecules, nucleic acid molecules which encode dominant negative molecules, ribozyme molecules specific for iASPP nucleic acids, and the like. The transgenic non-human animals are useful in experiments directed toward testing biochemical or physiological effects of diagnostics or therapeutics for conditions characterized by increased or decrease iASPP expression. Other uses will be apparent to one of ordinary skill in the art.
  • According to a further aspect of the invention there is provided the use of the polypeptide, or fragment thereof, in a screening method for the identification of agents which inhibit the binding of said polypeptide to p53.
  • According to a further aspect of the invention there is provided a screening method to identify agents which inhibit the binding of a polypeptide or fragment thereof to p53 comprising:
      • i) forming a preparation comprising
        • a) a polypeptide according to the invention; and
        • b) a p53 polypeptide, or a fragment thereof consisting of the binding site(s) for the polypeptide in (a);
      • ii) providing at least one agent to be tested; and
      • iii) determining the activity of the agent with respect to the binding of the polypeptide in (a) to the polypeptide in (b).
  • In a preferred method of the invention said agent is a polypeptide, preferably a peptide.
  • In preferred method of the invention said peptide comprises an amino acid sequence selected from the group consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • In a preferred method of the invention said peptide is at least 6 amino acid residues in length. Preferaby the length of said peptide is selected from the group consisting of: at least 7 amino acid residues; 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues in length. Alternatively the length of said peptide is at least 20 amino acid residues; 30; 40; 50; 60; 70; 80; 90; or 100 amino acid residues in length.
  • In a further preferred method of the invention said peptide consists of an amino acid sequence consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • It will be apparent to one skilled in the art that modification to the amino acid sequence of peptides agents could enhance the binding and/or stability of the peptide with respect to its target sequence. In addition, modification of the peptide may also increase the in vivo stability of the peptide thereby reducing the effective amount of peptide. necessary to inhibit p53 binding of iASPP. This would advantageously reduce undesirable side effects which may result in vivo. Modifications include, by example and not by way of limitation, acetylation and amidation. Alternatively or preferably, said modification includes the use of modified amino acids in the production of recombinant or synthetic forms of peptides. It will be apparent to one skilled in the art that modified amino acids include, by way of example and not by way of limitation, 4-hydroxyproline, 5-hydroxylysine, N6-acetyllysine, N6-methyllysine, N6,N6-dimethyllysine, N6,N6,N6-trimethyllysine, cyclohexyalanine, D-amino acids, ornithine. Other modifications include amino acids with a C2, C3 or C4 alkyl R group optionally substituted by 1, 2 or 3 substituents selected from halo (eg F, Br, I), hydroxy or C1-C4 alkoxy.
  • It will also be apparent to one skilled in the art that peptides which retain p53 binding activity could be modified by cyclisation. Cyclisation is known in the art, (see Scott et al Chem Biol (2001), 8:801-815; Gellerman et al J. Peptide Res (2001), 57: 277-291; Dutta et al J. Peptide Res (2000), 8: 398-412; Ngoka and Gross J Amer Soc Mass Spec (1999), 10:360-363.
  • In a further preferred method of the invention said antagonist is an antibody or antibody binding part. Preferably said antibody is a monoclonal antibody or binding part thereof.
  • Antibodies, also known as immunoglobulins, are protein molecules which usually 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, α, 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 ‘constant’ regions of the H chains, i.e., IgG1, IgG2, IgG3 and IgG4). 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 Fab fragment.
  • In a further preferred embodiment of the invention said antibody is selected from the group consisting of: F(ab′)2, Fab, Fv and Fd fragments; and antibodies comprising CDR3 regions.
  • Preferably said fragments are single chain antibody variable regions (scFV's) or domain antibodies. If a hybidoma exists for a specific monoclonal antibody it is well within the knowledge of the skilled person to isolate scFv's from mRNA extracted from said hybridoma via RT PCR. Alternatively, phage display screening can be undertaken to identify clones expressing scFv's. Domain antibodies are the smallest binding part of an antibody (approximately 13 kDa). Examples of this technology is disclosed in U.S. Pat. No. 6,248,516, U.S. Pat. No. 6,291,158, U.S. Pat. No. 6,127,197 and EP0368684 which are all incorporated by reference in their entirety.
  • A modified antibody, or variant antibody, and reference antibody, may differ in amino acid sequence by one or more substitutions, additions, deletions, truncations which may be present in any combination. Among preferred variants are those that vary from a reference polypeptide by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid by another amino acid of like characteristics. The following non-limiting list of amino acids are considered conservative replacements (similar): a) alanine, serine, and threonine; b) glutamic acid and asparatic acid; c) asparagine and glutamine d) arginine and lysine; e) isoleucine, leucine, methionine and valine and f) phenylalanine, tyrosine and tryptophan. Most highly preferred are variants which show enhanced biological activity.
  • Preferably said antibody is a humanised or chimeric antibody.
  • A chimeric antibody is produced by recombinant methods to contain the variable region of an antibody with an invariant or constant region of a human antibody.
  • A humanised antibody is produced by recombinant methods to combine the complementarity determining regions (CDRs) of an antibody with both the constant (C) regions and the framework regions from the variable (V) regions of a 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 complimentarity 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 complimentarity 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 elicit 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.
  • According to a further aspect of the invention there is provided an isolated nucleic acid molecule wherein said molecule is isolated from a nematode worm which nucleic acid molecule hybridises a nucleic acid sequence as represented by FIG 1 b.
  • In a preferred embodiment of the invention said nucleic acid molecule hybridises under stringent hybridisation condtions. Preferably said nematode worm is of the genus Caenorhabditis spp.
  • According to a further aspect of the invention there is provided an isolated polypeptide comprising the amino acid as represented in FIG. 1 b which polyeptide is modified by addition, deletion or substitution of at least one amino acid residue.
  • According to a further aspect of the invention there is provided a method of treatment of an animal comprising administering an effective amount of a polypeptide or nucleic acid or vector according to the invention wherein said effective amount induces the apopoptic activity of p53.
  • In a preferred method of the invention said treatment is of cancer.
  • According to an aspect of the invention there is provided a peptide comprising an amino acid sequence selected from the group consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • In a preferred embodiment of the invention said peptide is at least 6 amino acid residues in length. Preferably the length of said peptide is selected from the group consisting of: at least 7 amino acid residues; 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues in length. Alternatively the length of said peptide is at least 20 amino acid residues; 30; 40; 50; 60; 70; 80; 90; or 100 amino acid residues in length.
  • In a further preferred embodiment of the invention said peptide consists of an amino acid sequence consisting of: GPEETD; DGPEETD; TTLSDG; AEFGDE; or PRNYFG.
  • According to a further aspect of the invention there is provided a pharmaceutical composition comprising at least one peptide according to the invention and at least one anti-cancer agent
  • In a preferred embodiment of the invention said said anticancer agent is selected from the group consisting of: cisplatin; carboplatin; cyclosphosphamide; melphalan; carmusline; methotrexate; 5-fluorouracil; cytarabine; mercaptopurine; daunorubicin; doxorubicin; epirubicin; vinblastine; vincristine; dactinomycin; mitomycin C; taxol; L-asparaginase; G-CSF; etoposide; colchicine; derferoxamine mesylate; and camptothecin.
  • According to a yet further preferred aspect of the invention there is provided a complex comprising a peptide according to the invention and at least one antibody, or active binding part thereof.
  • In a preferred embodiment of the invention said antibody or fragment is a cell specific antibody. Preferably said cell is a cancer cell.
  • A complex of a peptide according to the invention and an antibody which specifically binds a cell, preferably a cancer cell, enables the targeting of said peptide to cell. Typically, the complex would be internalised releasing the peptide intracellularly to deliver the peptide to its target. Means to form a complex of antibody and peptide are known in the art and include the use of, for example bi-functional crosslinking agents, these may be hetero-bifunctional or homo-bifunctional. The crosslinkers may be reducible thereby facilitating release of said peptide(s). Cancer specific cell markers are known in the art. For example, tumour rejection antigen precursors. These include by example and not by way of limitation the the MAGE, BAGE, GAGE and DAGE families of tumour rejection antigens, see Schulz et al Proc Natl Acad Sci USA, 1991, 88, pp991-993 which is incorporated by reference.
  • According to an aspect of the invention there is provided a method of treatment of an animal, preferably a human, wherein said animal would benefit from the induction of apoptosis comprising administering an effective amount of a peptide according to the invention.
  • According to a further aspect of the invention there is provided a method of treatment of an animal, preferably a human, wherein said animal would benefit from the induction of apoptosis comprising administering an effective amount of a composition according to the invention
  • In a preferred method of the invention said treatment is cancer treatment.
  • An embodiment of the invention will now be described by example only and with reference to the following figures:
  • FIG. 1 a is the nucleic acid sequence of human iASPP; FIG. 1 b is the C. elegans nucleic acid sequence of iASPP;
  • FIG. 2 a is the amino acid sequence of human iASPP; FIG. 2 b is the C. elegans amino acid sequence of iASPP;
  • FIG. 3 illustrates that FITC labelled peptide (3 a) DGPEETD and (3 b) TTLSDG can penetrate cells;
  • FIG. 4 illustrates the stimulation of the Bax promoter by p53 after incubation with various peptides, in particular DGPEETD;
  • FIG. 5 illustrates the stimulation of p53 transactivation in a human tumour cell line U2SO after UV damage of DNA in the presence of peptide DGPEETD;
  • FIG. 6 illustrates various experiments showing the interaction of C. elegans iASPP and p53;
  • FIG. 7 illustrates a homology comparison between C. elegans iASPP and human iASPP; and
  • FIG. 8 illustrates further experiments showing the interaction of C. elegans iASPP and p53;
  • FIG. 9 illustrates the effect of RNAi on C. elegans iASPP expression;
  • FIG. 10 illustrates p53 contact sites in iASPP;
  • FIG. 11 Influence of peptides on transactivation level of Bax-Luc reporter. Cells were transfected with Bax reporter gene and exposed to UV (20J/m2). Peptides were added for 24 hours in medium at two different concentrations as indicated;
  • FIG. 12 illustrates the influence of peptides on Pig3 reporter activity for 12 hours. Transactivated cells were grown with medium containing Arg-tagged peptides at two concentrations. The left bar graph represents the transactivation activity at a concentration of 25 uM and the second is with a concentration of 50 uM. Yap peptide is used as a control peptide here instead of p9. Except for the control, cells were treated with cisplatin (3 ug/ml);
  • FIG. 13 illustrates the effects of various chemotherapeutic drugs on p53 induced apoptosis;
  • FIG. 14 a illustrates the effects of antisense or si-RNA of ASPP1, ASPP2 or iASPP, in combination with the activity of the anti-cancer agent doxorubicin; FIG. 14 b illustrates the effects of antisense or si-RNA of ASPP1, ASPP2 or iASPP in combination with the activity of the anti-cancer agent cisplatin;
  • FIG. 15 illustrates the activity of inhibitory peptides in combination with cisplatin on ASPP1, ASPP2 and iASPP.
  • MATERIALS AND METHODS
  • Cell Culture, Antibodies and Plasmids
  • Saos-2, MCF-7, and U2OS cells were grown in DMEM supplemented with 10% FCS, 100 IU/ml penicillin-streptomycin and 2 mM glutamine. Anti-p53 antibodies DO-1 and DO-13 are monoclonal antibodies while CM-1 is a rabbit polyclonal antibody specific to p53. The V5 and 9E10 epitopes are recognised by the mouse monoclonal antibodies V5 and 9E10 respectively. The mouse monoclonal PC-10 is specific to the PCNA protein. CD20Leu is an FITC conjugated monoclonal antibody specific for the cell surface marker CD20 (Becton Dickinson). The mouse and rabbit antibodies to ASPP1 and ASPP2 were described previously1. Mouse and rabbit antibody to iASPP (peptide RLQPALPPEAQSVPELEE) was produced as described by Harlow and Lane 13. All expression plasmids used in this study were driven by the CMV immediate early promoter. ASPP1, iASPP and Ce-iASPP are tagged with V5 epitope while Ce-p53 is tagged with 9E10 epitope.
  • DNA Transfection
  • The transfection mix included the DNA of interest in 1×HBS buffer (280 mM NaCl, 10 mM KCl, 1.4 mM Na2HPO4.2H2O, 12 mM Glucose, 39 mM HEPES, adjusted to pH 6.9-7.3) precipitated with 2.5 M CaCl2. The transfection mix was added dropwise to the cells and washed off after 6 hours with DMEM. 16-24 hours following the wash, cells were lysed either in Reporter lysis buffer (Promega) for Luciferase assays and western blots or in NP40 lysis buffer for western or immunoprecipitation procedures.
  • Transactivation Assays
  • For transcriptional assay, 5×105 Saos-2 cells were plated 24 hr prior to transfection in 6 cm dishes. Various combinations of plasmid DNA were transfected using the following amounts. All transfection assays contain 1 μg of reporter plasmid. 50 ng of wild type human p53, 100 ng of plasmid expressing C. elegans p53, 4 μg of ASPP2, or 8 μg of ASPP1, 5 μg of human iASPP or 7.5 μg of Ce-iASPP were used as indicated. After transfection, the cells were lysed in Reporter Lysis Buffer (Promega, WI, USA) 16-24 hr post-wash and assayed using the Luciferase Assay kit (Promega, WI, USA). The fold activation of a particular reporter was determined by the activity of the transfected plasmid above the activity of vector alone. The fold increase of p53 transactivation activity by ASPP was obtained by the activity p53 in combination with ASPP divided by the activity of p53 alone on the promoters used in each assay.
  • Cell Transformation Assay
  • Rat Embryo Fibroblasts (REFs) obtained from Biowhittaker were grown in DMEM to 50% confluence in a 90 mm dish. Cells were then transformed as described previously 14. Briefly, 2 μg of EJ ras 6.6, 2 μg of pCE (E1A), 5 μg of pCB6-16E, 5 μg of wild type human p53, 1 μg or 5 μg of human or C. elegans iASPP were transfected into the REFS as indicated. All the cells were transfected with the same amount of plasmid DNA expressing neo gene. The transfected cells were then selected with 400 μg/ml of G418 and the morphologically transformed colonies were scored after 3-4 weeks after transfection.
  • Flow Cytometry
  • For FACS analysis, 106 Saos-2 cells were plated 24-48 h prior to transfection in 10 cm plates. The cells were then transfected with 2 μg of a plasmid expressing CD20. CD20 expression was used as a transfection marker. The transfections consisted of 1 μg of human p53 or 4 μg of Ce-p53, 10 μg of ASPP1 and ASPP2, 2 μg of Bax, 15 μg of anti-sense iASPP, 7.5 μg-10 μg of human iASPP or 7.5 μg of Ce-iASPP plasmid as indicated. 36 hours after the transfection, both attached and floating cells were harvested using 4 mM EDTA/PBS and stained with FITC conjugated anti-CD20 antibody CD20Leu. For each experiment, one dish of cells was transfected with the control vector only without CD20. These cells were later stained with antibody CD20Leu under the same conditions as those co-transfected with CD20 plasmid to serve as a negative control. The cells lacking expression of CD20 plasmid were used to set the base line to allow the gating of the CD20 positive (and hence transfected) cells. After staining with the antibody CD20Leu, the cells were fixed and stained with propidium iodide. The DNA content of all the cells expressing CD20 was analysed using the flow cytometer (Becton Dickson) as described15.
  • Protein Biochemistry
  • For western blotting, cells grown in monolayers were washed with 1×PBS and lysed in either NP40 lysis buffer (1% Nonidet P40, 50 mM Tris pH8.0, 150 mM NaCl, 1 mM EDTA pH 8.0) or luciferase reporter lysis buffer. The protein concentration of the cell extracts was determined against a standard curve using the BioRad protein assay system (BioRad). Between 15-100 μg of extract was mixed with 5×sample buffer and loaded on SDS-PAGE gels. The gels were wet transferred on to Protran nitrocellulose membrane and the resulting blots blocked in 10% reconstituted milk powder in 1×PBS. Subsequently the blots were incubated with primary antibody prepared either in tissue culture medium or as undiluted hybridoma supernatant and incubated with the appropriate secondary HRP conjugated antibody (Dako). In between each stage the blot was washed with repeat changes of TBST (10 mM Tris pH 8.0, 150 mM NaCl, 0.5% Tween 20). The blot was exposed to hyperfiln following the use of ECL substrate solution (Amersham Life Science).
  • For immunoprecipitation cells were lysed in NP40 lysis buffer on ice for 30 minutes and pre-cleared with protein G beads for 1 hour at 4 C. The protein concentration was determined and 1000 μg of the extract was incubated with antibody pre-bound to protein G beads for 4 hours at 4 C. The beads were washed twice in NP40 lysis buffer and twice in NET buffer (50 mM Tris pH 8.0, 150 mM NaCl, 1 mM EDTA pH 8.0). The IP beads were mixed with 5×sample buffer and loaded onto a SDS-PAGE gel.
  • In Vitro Translation and In Vitro Immunoprecipitation
  • ASPP family members and p53 were in vitro translated and labelled with 35S—Methionine using the TNT T7 Quick coupled Transcription/Translation System (Promega). For the experiments shown in FIG. 1E, 15, 30 and 45 μl of in vitro translated lysates of iASPP were added in addition to p53 and ASPP2. Rabbit anti-p53 antibody CM1 was used to detect the presence of unlabelled p53.
  • For FIG. 6A, 5-10 μl of human iASPP lysate (unlabelled) was incubated with 15 μl of the lysate containing the in vitro translated Ce-p53. The mixture of proteins was allowed to co-translate at 30 C for 1 hr. 200 μl of phosphate-buffered saline (PBS) was then added to the mixture of proteins and incubated at 4 C for a further hour on a rotating wheel. The anti-V5 antibody irmnobilised on protein G agarose beads was added to the binding reactions and incubated on a rotating wheel at 4 C for 16 hours. The beads were then washed with PBS. The bound proteins were released in SDS gel sample buffer and analysed by 10% SDS-polyacrylamide gel electrophoresis (PAGE). Ce-p53 was detected by autoradiography and Human iASPP was detected by anti-V5 antibody following a western blot. For the rest of the figures, the proteins were labelled with 35S -Methionine and immunoprecipitated as above using the indicated antibodies. Results were visualised using autoradiography.
  • In vivo Labelling of Cells with 35S -Methionine and 35S -Cysteine
  • U20S cells in the absence or presence of transfected plasmids (as indicated in FIG. 1A and 2C) were washed with PBS and then incubated with 250 μci/ml of 35S—methionine and 250 μci of 35-cysteine in DMEM lacking both methionine and cysteine for 2 hours at 37C. Cells were then washed with PBS before harvesting. For FIG. 2C, twenty four hours after transfection the cells were in vivo labelled with 35S-methionine and 35S-cysteine for 2hrs. Cells expressing CD20 (transfected cells) were stained with FITC-conjugated anti-CD20 antibody. A biotin conjugated anti-FITC antibody was then added to the cell pellet, and after the incubation, the cells were mixed with streptavidin conjugated magnetic beads to isolate the CD20 expressing cells. The cells were then lysed with NP40 lysis buffer and the proteins immunoprecipitated with mouse anti iASPP antibody. The immunoprecipitates were washed with NET buffer and resolved by SDS-PAGE and autoradiography.
  • Cell Fixation
  • Monolayers of cells were grown in 30 mm dishes and washed with 1×PBS. Cells were fixed with 1 ml of 4% para-formaldehyde for 15 minutes and then washed in 1×PBS. 1 ml of 0.2% Triton-X100 in 1×PBS was used to permeabilise the cells for 2 minutes and this was washed off with three washes of 1×PBS. Primary antibody was prepared in tissue culture medium at the appropriate concentration and added to the dishes for 3 hours. The dishes were washed with 1×PBS and the secondary antibody of either anti-rabbit TRITC (Tetramethyl rhodamine isothiocyanate) or anti-mouse FITC (Fluorescein isothiocyanate) prepared in tissue culture medium at the manufacturers recommended dilution (Sigma, UK) and added to the dishes for one hour. The cells were washed in 1×PBS and left to air dry. Citifluor shielding agent (Citifluor, UK) was applied as a drop to the surface of the cells and a cover slip placed on top. A drop of immersion oil on top of the cover slip allowed the immunocomplexes to be visualised using a Zeiss Axiophot fluorescence microscope. Antibodies 9E10 and V5 were used to detect the expression of epitope tagged Ce-p53 (9E10), human iASPP (V5) and Ce-iASPP (V5) respectively. Human p53 was detected by DO.1 antibody.
  • Cloning Ce-p53 and Ce-iASPP cDNA
  • cDNAs carrying the complete coding regions of Ce-ape-1 (iASPP) and Ce-cep-1 (p53) were generated by RT-PCR using the Promega Access kit, cloned into the vector pCR4-TOPO (Invitrogen) and sequenced. C. elegans p53 and iASPP were then subcloned into a mammalian expression vector pcDNA3 in frame with the epitope of 9E10 and V5 respectively. The full-length Ce-ape-1 is predicted to be trans-spliced to SL1 (Y. Kohara, unpublished).
  • RNA Interference (RNAi) and Cell Corpse Assays
  • RNAi was performed by feeding or microinjection using established procedures (Fire et al., 1998; Timmons and Fire, 1998) In order to eliminate Ce-iASPP and Ce-p53 activity by RNAi, N2 animals were first subjected to Ce-iASPP RNAi feeding. Subsequently, 20 F1 animals were removed from the feeding plate, injected with Ce-p53 dsRNA and returned to independent Ce- iASPP dsRNA feeding plates. The F2 progeny of animals fed Ce-iASPP (+/− injection of Ce-p53 dsRNA) were stained with SYTO12 and apoptotic corpses scored as described12. Hence, because all animals have been subjected to Ce-iASSP feeding RNAi, any differences in the mean number of apoptotic cell corpses detected in the two populations are likely to result from Ce-p53 dsRNA injection.
  • EXAMPLES
  • iASPP is the Most Conserved Member of the ASPP Family
  • Sequence analysis indicates that the C. elegans p53 gene, cep-1, is a distant member of the p53 family, however, the residues critical for ASPP and DNA binding activity appear to be conserved3,4 Hence, we searched the C. elegans genome for an ASPP homologue and found that F46F3.4 is the only C. elegans gene encoding a protein with significant sequence homology to all three members of the ASPP family. The gene corresponding to F46F3.4 has been named ape-1 (for apoptotic enhancer) based on the mutant phenotype produced by ape-1(RNAi) (see below); however, the protein product will henceforth be referred to as Ce-iASPP. Ce-iASPP consists of 769 amino acids; sequence comparisons reveal that the C-terminus of Ce-iASPP is the region most conserved with other ASPP members (FIG. 7). It was previously shown that the C-terminus of ASPP2 interacts with p53. Moreover, all but one (6 out of 7) residue involved in this interaction are conserved in both iASPP and Ce-iASPP. Taken together these results suggested that Ce-iASPP might interact with both human and C. elegans p53. This was tested in vitro by co-immunoprecipitation. As shown in FIG. 8B, Ce-iASPP interacts with both human and Ce-p53. The interaction between Ce-p53 and Ce-iASPP was further confirmed by reciprocal immunoprecipitation (FIG. 8B, right panel).
  • Ce-iASPP contains the hallmark ankyrin repeats and SH3 domain found in other ASPP family members and is expressed in the cytoplasm and nucleus of human cells (FIG. 8C). The expression of human iASPP is predominantly nuclear but cytoplasmic staining is also detectable. Both human and C. elegans p53 are primarily expressed in the nucleus of transfected human cells (FIG. 8C). Since ASPP and iASPP can positively and negatively regulate the apoptotic function of p53, respectively, we tested the effect of Ce-iASPP on the activities of p53. When Ce-iASPP was co-expressed with human p53 in mammalian cells, it produced a small reduction in the transactivation and apoptotic function of p53, presumably by inhibiting endogenous ASPP function. However, in the presence of ASPP1 or ASPP2, co-expression of Ce-iASPP prevented ASPP1 or ASPP2 from stimulating the transactivation and apoptotic function of p53 to the same extent as human iASPP (FIG. 8D). Furthermore, the ability of human and C. elegans iASPP to inhibit apoptosis is p53-dependent since they both failed to inhibit Bax induced apoptosis under the same conditions (FIG. 8E). The ability of iASPP to inhibit the activity of p53 is not due to the reduced expression of p53 (FIG. 8F). Like human iASPP, the Ce-iASPP also has oncogenic activity. The expression of Ce-iASPP enhanced the transforming activity of ras and E1A. Moreover, expression of Ce-iASPP inhibited the suppressor function of wild type human p53 (FIG. 8G). These results demonstrate that Ce-iASPP is more likely to be an orthologue of human iASPP than of ASPP. Also, the ability of Ce-iASPP to inhibit p53 suggests that the regulation of p53 apoptotic function by the ASPP family of proteins is evolutionarily conserved.
  • The Regulation of p53 by ASPP Family of Proteins is Evolutionarily Conserved
  • It was unclear whether Ce-p53 could induce apoptosis in mammalian cells because of the limited sequence similarity between human and C. elegans p53, although most of the Ce-p53 residues that contact ASPP are conserved. If Ce-iASPP inhibits the activities of human p53 in a manner similar to that of human iASPP, this argues that the regulation of p53 by the ASPP family has been evolutionarily conserved. This further suggests that the activities of Ce-p53 could be subject to regulation by the ASPP family of proteins. To address these issues, Ce-p53 was tested for its ability to interact in vitro with members of the human ASPP family by co-immunoprecipitation. As shown in FIG. 6A, Ce-p53 interacts with ASPP2 and iASPP. The expression of Ce-p53 induced apoptosis in human cells with an efficiency similar to human p53. Remarkably, expression of human ASPP, ASPP2 in particular, significantly enhanced the ability of Ce-p53 to induce apoptosis to an extent similar to human p53 and the expression of human iASPP also inhibited the apoptotic function of Ce-p53 (FIG. 6B). In addition, both human and C. elegans iASPP inhibited Ce-p53 induced apoptosis to the same extent (FIG. 6C). The ability of Ce-p53 to transactivate p53 target gene promoters, such as Bax-luc, was also tested and was found to be much lower than that of human p53. Interestingly, co-expression of ASPP2 and Ce-p53 resulted in a very small but detectable increase in the transactivation function of Ce-p53, indicating that the human ASPP family could regulate Ce-p53 in a manner similar to that of human p53 (FIG. 6D). The small increase in the transactivation function of Ce-p53 by ASPP2 is not observed on the mdm2 promoter. All of these results suggest that the residues conserved between human and C. elegans p53 are crucial and sufficient for the apoptotic function of p53 and for p53 to be regulated by the ASPP family.
  • iASPP is an Evolutionarily Conserved Inhibitor of p53 In Vivo
  • Like human p53, one of the most important functions of C. elegans p53 is its ability to induce apoptosis in germ cells in response to DNA damage 3,4. Knowing that co-expression of human or C. elegans iASPP can inhibit the apoptotic function of p53 in mammalian cell lines, we hypothesised that expression of Ce-iASPP might similarly protect C. elegans germ cells from death by apoptosis. This question was addressed in vivo using RNA mediated interference (RNAi) 5. Depletion of endogenous Ce-iASPP increased the number of germ cells undergoing apoptosis, indicating that the normal function of Ce-iASPP is to inhibit apoptosis (FIG. 9A, lanes 1 and 5, 6). The enhancement of germ cell apoptosis caused by depletion of Ce-iASPP was not detected when RNAi was performed in a mutant lacking the C. elegans CED-3 caspase, indicating that the core apoptotic machinery is involved in this process (FIG. 9A, lanes 3 and 4) 6. We also obtained additional support for the hypothesis that the primary role of Ce-iASPP is to inhibit the pro-apoptotic activity of Ce-p53, which is normally stimulated in response to genotoxic stress 3,4. First, it was found that the increase in the number of C. elegans germ cells undergoing apoptosis after depletion of Ce-iASPP was abrogated by simultaneously depleting Ce-p53 by RNAi (FIG. 9A, compare lanes 5, 6 and 8). Moreover, the depletion of both Ce-iASPP and Ce-p53 by RNAi did not completely eliminate apoptosis, but instead returned the number of germ cells undergoing apoptosis to wild-type physiological levels. Second, the increase in the number of apoptotic germ cell corpses detected after wild type worms were exposed to 100-Gy IR was no greater than that observed after depletion of Ce-iASSP by RNAi in the presence or absence of exposure to 100-Gy IR (FIG. 9A, lanes 2 and 7). These results clearly demonstrate that iASPP is an important inhibitor of p53 function in C. elegans, although we can not exclude the possibility that a genetic knockout might reveal that Ce-iASPP has additional activities. Since the regulation of p53 by the ASPP family is highly conserved, it is likely that iASPP is also a key inhibitor of p53 in other organisms including humans.
  • We show here that iASPP is the most phylogenetically conserved inhibitor of p53, so far identified, and also the most evolutionarily conserved member of the ASPP family. Remarkably, the ability of ASPP family members to regulate the apoptotic function of p53 has been conserved between C. elegans and human. This argues that the apoptotic function of p53 is likely to be more conserved than its ability to induce cell cycle arrest; this agrees with recent observations showing that ectopic expression of both C. elegans and Drosophila p53 induces apoptosis but not cell cycle arrest 3,4,7,8. In C. elegans p53-mediated apoptosis appears to play an important role in maintaining the fidelity of germ cells, which might have incurred DNA damage 3,4. Interestingly, the most important tumour suppressor function of p53 is also linked to its ability to induce apoptosis. Therefore, ASPP family members, the evolutionarily conserved regulators of p53, must play a critical role in tumourigenesis.
  • The Oncoprotein iASPP
  • iASPP shares more sequence similarity with the N-terminally truncated ASPP2 mutant 53BP2 than the full-length ASPP. Expression of iASPP inhibited the apoptotic function of p53. Like 53BP2, the most profound effect of iASPP on the apoptotic function of p53 is mediated through its ability to act as a competitor of ASPP. However, in C. elegans, iASPP is the only gene that has homology to the human ASPP family. Thus iASPP directly inhibits the apoptotic function of p53 in C. elegans. A similar mechanism might also apply in mammalian cells. In agreement with this, iASPP antisense RNA induced a 3 to 5-fold increase in apoptotic cells in U2OS and MCF7 cells. In this latter model, ASPP could stimulate the apoptotic function of p53 by removing the negative effects that iASPP imposed on p53. The failure of iASPP antisense RNA to produce a significant increase of apoptosis in cisplatin treated U2OS and MCF7 cells might be due to the fact that cisplatin stimulates the apoptotic function of p53 by increasing the activities of ASPP. This, in turn, might explain why the expression of ASPP antisense RNA generated a profound inhibitory effect on apoptosis induced by cisplatin. It is also under this condition that the anti-apoptotic function of iASPP is most pronounced. Thus the apoptotic function of p53 is negatively regulated by iASPP and positively regulated by ASPP. The competition between these two opposing signals could determine the apoptotic state of p53 and ultimately cell fate. Regardless of whether iASPP acts as a dominant negative regulator of ASPP or a direct inhibitor of p53, a competition between iASPP and ASPP for binding to p53 is critical for the apoptotic function of p53. Consistent with this model, a change in the percentage of p53 complexed with ASPP2 was seen in response to DNA damage. It is likely that the percentage of p53 complexed with iASPP and ASPP would be regulated by signals that induce death or survival.
  • Being an inhibitor of p53, iASPP enhanced the transforming activities of oncogenes such as ras plus E7 or E1A of human papilloma virus and adenovirus but not ras plus mutant p53. This is particularly interesting since E7 and E1A are known to induce p53-dependent apoptosis 9. While E7 and E1A can bind and inactivate the tumour suppressor function of Rb, their oncogenic function is largely reduced due to their ability to activate p53-dependent apoptosis. Proteins that can inhibit apoptosis induced by E7 and E1A would enhance the oncogenic function of E7 and E1A. Therefore, like the dominant negative p53 mutants, iASPP was able to stimulate the oncogenic function of E7 and E1A by inhibiting the apoptotic function of p53. It is important to point out that iASPP was not as active as mutant p53, p53H175 or p53L173 in co-operating with ras to transform REFs under the experimental conditions described here. Part of the reason was due to the low expression level of iASPP in the assay (data not shown). However, the differences in the transforming activities of iASPP and mutant p53 might also be caused by other known activities of mutant p53, which are independent of its ability to act as a simple dominant negative inhibitor of p53. Nevertheless the ability to confer cellular resistance to the cytotoxic effects of UV and cisplatin suggested that the overexpression of iASPP would be selected for in human tumours expressing wild type p53. Consistent with this, iASPP expression is increased in human breast carcinomas expressing wild type p53. The majority of tumours (7 out of 8) expressing high levels of iASPP also express wild type p53 and normal levels of ASPP, indicating that iASPP is an inhibitor of ASPP in vivo. Our previous study showed that the expression levels of ASPP1 and ASPP2 were down regulated in 60% of human breast tumours express wild type p53. Taken together, the abnormal expression of ASPP family members would account for almost 80% of human breast carcinomas examined. The ASPP family members are encoded by three different genes located on different chromosomes (data not shown). We do not know why the frequency of human breast carcinomas showing down regulation of ASPP is much higher than those showing increased expression of iASPP. However, it is possible that the expression pattern of the ASPP family members varies in different types of human tumours. The percentage of tumours with altered ASPP expression could also differ in various tumour types. Nevertheless, inhibiting the oncogenic function of iASPP could provide an important new strategy to treat tumours expressing wild type p53.
  • Evolutionarily Conserved Regulation of p53 by the ASPP Family
  • Sequence comparison reveals that there is 38% identity between the human and C. elegans iASPP amino acid sequences; within the ankyrin repeats and SH3 domain, the homology is as high as 78% (residues 154-227 of human iASPP and 557-630 of Ce-iASPP, 55/74 residues are similar). Most of the iASPP residues contacting p53 are conserved. The structural conservation between human and C. elegans iASPP is reflected by their ability to regulate p53 function in human cells. Like human iASPP, C. elegans iASPP interacts with and inhibits the transactivation and apoptotic function of human p53 in cell lines. The conservation of ASPP/p53 regulation is further demonstrated in a study of C. elegans p53 in human cells. It is interesting and important to point out that the sequence homology between human and C. elegans p53 is very limited (13.7% identity at protein level). The highest level of p53 homology between the two species is around 50% in a very limited region (residues 9/18 residues are similar). However, many of the ASPP2 contact residues identified from a crystal structure10 are conserved between human and C. elegans p53 (5 out of 8 residues are conserved). The ability of C. elegans p53 to interact with human ASPP family members in vitro highlights the importance of these conserved residues.
  • Remarkably, C. elegans p53 induces apoptosis very effectively in human cells. Similar to human p53, the apoptotic function of C. elegans p53 is positively and negatively regulated by the human ASPP and iASPP, respectively. These results demonstrate for the first time that the apoptotic function of p53 is conserved despite the limited sequence homology between human and C. elegans p53. The few key residues conserved between human and C. elegans p53 are sufficient for ASPP family members to regulate the apoptotic function of p53 both in vitro and in vivo.
  • The C. elegans p53 showed little ability to transactivate human p53 target genes such as Bax and PIG3 in comparison to human p53 (data not shown). Even though Bax is frequently a p53 target gene during apoptosis, it is not the only target gene that is required for p53 induced apoptosis. p53 is known to transactivate over 20 pro-apoptotic genes none of which has so far proved to be indispensable in p53 induced apoptosis. There are over 4000 putative p53 target genes in the human genome and many of them are pro-apoptotic genes 11. It is possible that Ce-p53 might transactivate some of these other human p53 target genes as effectively as human p53. Unfortunately none of the ones we tested so far belong to this category. Although transactivation of the human Bax-luc reporter in human cells by Ce-p53 is not very strong, it is important to point out that the co-expression of ASPP stimulated the transactivation function of Ce-p53 only on the promoters of Bax but not on mdm2. This pattern of ASPP action is similar to that seen with human p53. Alternatively, Ce-p53 might induce apoptosis independent of its transcriptional activity; human p53 is known to induce apoptosis through both transcriptional dependent and independent pathways 12. Regardless of how Ce-p53 induces apoptosis in human cells, the most remarkable and important fact is that the human ASPP family of proteins regulate the apoptotic function of both Ce-p53 and human p53 in a similar way. Ce-iASPP is also able to completely replace human iASPP in all the assays performed in human cells. These results argue strongly that the regulation of p53 function by ASPP family is conserved from worm to human. The link between p53 and the ASPP family suggests that the regulation of p53 by the ASPP family should be the future target in developing new strategies for cancer therapy. Tumours expressing wild type p53 can be sensitised to treatments by enhancing the activity of ASPP and removing the activity of iASPP.
  • Chemotherapy Drugs Appear to Modify ASPP Family Protein Function
  • As 50% of human tumours maintain wild-type p53, one approach to treating cancer might be to reactivate p53 in tumour cells to induce apoptosis. Indeed, chemotherapy and radiotherapy agents generate DNA damage, which could activate functional p53 in tumour cells. However, the pathways affected by these drugs differ and tumour cells can develop protective strategies to enable survival. As mentioned previously, ASPP1 and ASPP2 specifically enhance the apoptotic function of p53 by stimulating the DNA binding and transactivation function of p53 on the promoters of proapoptotic genes (Samuels-Lev et al., 2001), while iASPP is a very conserved inhibitor of p53 (Bergamaschi et al., 2003). Taken together, this suggests that the function of the ASPP family of proteins may be affected by chemotherapy agents, which in turn vary the subsequent p53 apoptotic response, and thus play a role in the effectiveness of drugs used in cancer treatment.
  • Specifically, experiments were carried out to investigate the hypothesis that chemotherapy drugs could be made more effective in cell lines undergoing DNA damage by modulating the activity of the ASPPs.
  • Chemotherapy agents were used to investigate whether the apoptosis induced in cell lines correlated directly with p53 expression levels. U-2OS and MCF7 cells were transfected with the Pig3 promoter (Pig3-luc 17 mer) and treated with various chemotherapy drugs (doxorubicin, cisplatin, etoposide, colchicine, deferoxamine mesylate (DFO), daunorubicin, camptothecin and 5′fluorouracil).
  • Luciferase levels were assayed 24 hours later, FIG. 13. The results illustrate that drug-induced apoptosis, as measured by luciferase, did not correlate directly with p53 protein levels (for example, compare doxorubicin and cisplatin responses) indicating that p53 alone is not responsible for the activation of proapoptotic genes. It is possible that the ASPP proteins respond differently to the various chemotherapy drugs to give the discrepancy observed between Pig3 activation and p53 protein expression level.
  • To test whether the ASPPs play a role in the process, short interfering RNA (si-RNA) (Elbashir et al., 2001) and antisense RNA were used to knock down ASPP gene expression. Sequences to generate si-RNA were cloned into pSuper plasmid (Brummelkamp et al., 2002). Since responses of proapoptotic genes differed depending on the drug used, both doxorubicin and cisplatin were tested in U-2OS cells. Pig3-luciferase was transfected together with either antisense or si-iASPP (FIGS. 14A and 14B). The results show that antisense ASPP1 or antisense ASPP2 both reduce the induction of Pig3-luciferase by doxorubicin (FIG. 14A) but antisense or si-RNA for iASPP caused no reduction. A si-RNA for p53 prevented the doxorubicin response (FIG. 14A). In contrast, antisense ASPP1 or antisense ASPP2 had little effect on the cisplatin induction of the Pig3 promoter but the si-RNA for iASPP caused a significant increase. Again siRNA for p53 prevented the cisplatin response.
  • These data could be interpreted to indicate that endogenous p53 activity was induced by doxorubicin cooperating with ASPP1 and ASPP2 in U-2OS cells but in response to cisplatin endogenous p53 activity was being restrained by iASPP. These differential effects might be due to the modulatory effects of the drugs on ASPP or iASPP activity.
  • The Disruption of iASPP/p53 Binding can Enhance Apoptosis and Could Potentially be Used in Cancer Therapy
  • Inhibiting the p53/iASPP interaction with synthetic molecules should lead to p53-mediated apoptosis in p53-positive stressed cells.
  • It is known that p53 binds to the C terminal part of iASPP; specific contact amino acids are indicated in FIG. 10. Three different peptides containing particular iASPP sequences were designed to inhibit the iASPP/p53 binding and test the effects on proapoptotic genes, FIG. 10. The three peptides were linked to a Tat sequence and to FITC.
  • The peptides were initially tested for their effects on p53 transactivation of the Bax promoter using UVradiation to activate p53, FIG. 11. The luciferase activity of the Bax promoter was only slightly affected by the control peptide, which should not affect iASPP & p53 interaction. However, addition of peptide 3, 6 or 7 caused about a two fold increase in the reporter activity, using 50 μM peptide. Some peptides were less active at 100 μM.
  • The study was continued with the Pig3-luciferase reporter. The Pig3 gene is transactivated by p53 in a different way to Bax. Whereas Bax is transactivated by a conventional transcription domain (El-Deiry et al., 1992) (Bourdon et al., 1997), p53 binds to the Pig3 promoter via repeat sequences (TGYCC)n, where Y=C or T (Polyak et al., 1997). The number of repeats is polymorphic within populations (Contente et al., 2002). Two reporters containing 10 or 17 repeats were used in case there was a difference in response between them. Other variables to consider are time and concentration of peptide and the location of the peptides in the cells since p53 and iASPP are mostly nuclear proteins.
  • A second generation of peptides was used for this experiment, which are identical to the previous ones except for the tag. These peptides were tagged with nine arginine residues, which are thought to give the peptides better penetration into cells (Lindsay, 2002). In this case a peptide 7 from the ASPP2 sequence was included (see FIG. 10) and a different control peptide (Yap) was used. Cells were treated with cisplatin and peptides were applied for 12 hours at two different concentrations (25 uM and 50 uM) and the results are shown in FIG. 15. Despite some variability in the results the P3-iASPP or P7-iASPP peptides showed a substantial increase in the reporter activity over the response seen with control Yap peptide and cisplatin treatment, suggesting that apoptosis induced during chemotherapy could be enhanced through the use of iASPP peptide inhibitors.
    TABLE 1
    Human Mouse C.El. Drosophila Fugu I Fugu II Fugu III Fugu IV
    Human (352) X X X X X X X X
    Mouse (260) 93.7 X X X X X X X
    C.El. (769) 20.4 38.8 X X X X X X
    Drosophila (1071) 43.2 40.0 32.4 X X X X X
    Fugu I (260) 51.9 45.0 48.1 55.4 X X X X
    Fugu II (252) 54.8 54.8 51.6 58.7 X X X X
    Fugu III (144) 54.2 53.5 54.2 64.6 X X X X
    Fugu IV (132) 51.5 50.8 55.3 62.9 X X X X

Claims (54)

1. An isolated nucleic acid molecule which encodes a polypeptide, or sequence variant thereof, wherein said polypeptide is a fragment of the polypeptide sequence represented in SEQ ID NO: 8 or 9, wherein the fragment is:
a polypeptide fragment consisting of amino acid residues from about residue 128-224 of the amino acid sequence presented in SEQ ID NO: 8 or 9;
or a polypeptide fragment consisting of amino acid residues from about residue 128-224 of the amino acid sequence presented in SEQ ID NO: 8 or 9 wherein said sequence has been modified by addition, deletion or substitution of at least one amino acid residue, wherein the polypeptide inhibits the apoptoic activity of p53.
2. The nucleic acid molecule according to claim 1, wherein said molecule encodes a fragment consisting of amino acid residues from about residue 128-224 of the sequence represented in SEQ ID NO: 8.
3. The nucleic acid molecule according to claim 2, wherein said molecule is isolated from a human.
4. The nucleic acid molecule according to claim 1, wherein said molecule encodes a fragment consisting of amino acid residues from about residue 128-224 of the sequence represented in SEQ ID NO: 9.
5. The nucleic acid molecule according to claim 4, wherein said molecule is isolated from a nematode.
6. The nucleic acid molecule according to claim 5, wherein said nematode is of the genus Caenorhabditis spp.
7. (canceled)
8. The nucleic acid molecule according to claim 1, wherein said nucleic acid molecule is a cDNA or genomic DNA.
9. (canceled)
10. A polypeptide fragment or sequence variant thereof, encoded by the nucleic acid molecule according to claim 1.
11. A vector comprising the nucleic acid according to claim 1.
12. The vector according to claim 11, wherein said vector is an expression vector.
13. A cell tranformed or transfected with the nucleic acid molecule according to claim 1.
14. A pharmaceutical composition comprising the nucleic acid according to claim 1.
15. A pharmaceutical composition comprising the polypeptide according to claim 10.
16. (canceled)
17. A transgenic non-human animal comprising the nucleic acid molecule according to claim 1.
18. (canceled)
19. A screening method to identify agents which inhibit the binding of a polypeptide, or fragment thereof, to p53 comprising:
forming a preparation comprising
the polypeptide of claim 10; and
a p53 polypeptide, or a fragment thereof consisting of the binding site(s) for the polypeptide of claim 10;
providing at least one agent to be tested; and
determining the activity of the agent with respect to the binding of the polypeptide of claim 10 to the p53 polypeptide, or a fragment thereof consisting of the binding site(s) for the polypeptide of claim 10.
20. The method according to claim 19, wherein said agent is a polypeptide or a peptide.
21. (canceled)
22. The method according to claim 20, wherein said polypeptide is an antibody or binding part thereof.
23. The method according to claim 22, wherein said antibody is a monoclonal antibody.
24. The method according to claim 22, wherein said fragment is an Fab fragment.
25. The method according to claim 24, wherein said Fab fragment is an F(ab′)2 fragment, an Fab fragment, an Fv fragment, or CDR3 regions.
26. The method according to claim 23, wherein said antibody is humanised.
27. The method according to claim 23, wherein said antibody is a chimeric antibody.
28. An isolated nucleic acid molecule, wherein said molecule is isolated from a nematode worm and hybridises to the nucleic acid sequence shown in SEQ ID NO: 9, wherein said nucleic acid molecule encodes an inhibitor of p53 and inhibits the apoptotic activity of p53.
29. The nucleic acid molecule according to claim 28, wherein said molecule hybridises under stringent hybridisation conditions.
30. (canceled)
31. An isolated polypeptide comprising the amino acid sequence as represented in SEQ ID NO: 9 or a variant polypeptide which polypeptide is modified by addition, deletion or substitution of at least one amino acid residue and is an inhibitor of p53.
32. A method of treatment of an animal, comprising administering an effective amount of the polypeptide according to claim 10, wherein said effective amount induces the apoptotic activity of p53.
33. A method of treatment of an animal comprising administering an effective amount of a nucleic acid molecule according to claim 1, wherein said effective amount induces the apoptotic activity of p53.
34. The method according to claim 32, wherein said treatment is of cancer.
35. The polypeptide of claim 10, wherein the polypeptide is a peptide comprising the amino acid sequence DGPEETD (SEQ ID NO: 2); GPEETD (SEQ ID NO: 2); TTLSDG (SEQ ID NO: 3); AEFGDE (amino acids 294-299 of SEQ ID NO: 8); or PRNYFG (SEQ ID NO: 4).
36. The peptide according to claim 35, wherein said peptide is at least 6 amino acid residues.
37. The peptide according to claim 35, wherein said peptide is at least 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues.
38. The peptide according to claim 35, wherein said peptide is at least 20; 30; 40; 50; 60; 70; 80; 90; or 100 amino acid residues.
39. The peptide according to claim 35, wherein the peptide consists of the amino acid sequence DGPEETD (SEQ ID NO: 2); GPEETD (SEQ ID NO: 1); TTLSDG (SEQ ID NO: 3); AEFGDE (amino acids 294-299 of SEQ ID NO: 8); or PRNYFG (SEQ ID NO: 4).
40. (canceled)
41. The peptide according to claim 35, wherein the peptide further comprises at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 arginine residues.
42. (canceled)
43. A pharmaceutical composition comprising the peptide of claim 35.
44. The pharmaceutical composition according to claim 43, wherein said composition further includes a carrier, diluent or excipient.
45. The pharmaceutical composition of claim 35 further comprising at least one anti-cancer agent.
46. The pharmaceutical composition according to claim 45 wherein said anticancer agent is cisplatin; carboplatin; cyclosphosphamide; melphalan; carmusline; methotrexate; 5-fluorouracil; cytarabine; mercaptopurine; daunorubicin; doxorubicin; epirubicin; vinblastine; vincristine; dactinomycin; mitomycin C; taxol; L-asparaginase; G-CSF; etoposide; colchicine; derferoxamine mesylate; or camptothecin.
47. (canceled)
48. (canceled)
49. A complex comprising the peptide according to claim 35 and an antibody, or binding part thereof.
50. The complex according to claim 49, wherein said antibody or binding part is a cell specific antibody.
51. The complex according to claim 49 wherein said antibody is a cancer cell specific antibody.
52. A method of treatment of an animal that would benefit from the induction of apoptosis, comprising administering an effective amount of the peptide of claim 35 to the animal.
53. (canceled)
54. The method according to claim 52, wherein said treatment is cancer treatment.
US10/522,043 2002-10-07 2003-10-03 Polypeptide Abandoned US20060100143A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/043,058 US7745571B2 (en) 2002-10-07 2008-03-05 Peptide inhibitors of IASPP

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GB0223193A GB0223193D0 (en) 2002-10-07 2002-10-07 Polypeptide
GB0223193.4 2002-10-07
GB0306261.9 2003-03-19
GB0306261A GB0306261D0 (en) 2003-03-19 2003-03-19 Polypeptide
PCT/GB2003/004296 WO2004031229A2 (en) 2002-10-07 2003-10-03 P53 binding polypeptide

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/043,058 Division US7745571B2 (en) 2002-10-07 2008-03-05 Peptide inhibitors of IASPP

Publications (1)

Publication Number Publication Date
US20060100143A1 true US20060100143A1 (en) 2006-05-11

Family

ID=32071253

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/522,043 Abandoned US20060100143A1 (en) 2002-10-07 2003-10-03 Polypeptide
US12/043,058 Expired - Fee Related US7745571B2 (en) 2002-10-07 2008-03-05 Peptide inhibitors of IASPP

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/043,058 Expired - Fee Related US7745571B2 (en) 2002-10-07 2008-03-05 Peptide inhibitors of IASPP

Country Status (9)

Country Link
US (2) US20060100143A1 (en)
EP (2) EP1558638B1 (en)
JP (1) JP2006517787A (en)
AT (2) ATE425183T1 (en)
AU (1) AU2003299132A1 (en)
CA (1) CA2487206A1 (en)
DE (1) DE60326623D1 (en)
HK (1) HK1083220A1 (en)
WO (1) WO2004031229A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100168388A1 (en) * 2007-01-31 2010-07-01 Federico Bernal Stabilized p53 peptides and uses thereof
US8859723B2 (en) 2010-08-13 2014-10-14 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8927500B2 (en) 2012-02-15 2015-01-06 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8987414B2 (en) 2012-02-15 2015-03-24 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9604919B2 (en) 2012-11-01 2017-03-28 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9880171B2 (en) 2012-03-02 2018-01-30 Ludwig Institute For Cancer Research Ltd. iASPP phosphorylation and metastatic potential
US10023613B2 (en) 2015-09-10 2018-07-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of MCL-1
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US11091522B2 (en) 2018-07-23 2021-08-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0514538D0 (en) * 2005-07-14 2005-08-24 Ludwig Inst Cancer Res Variant polypeptide and screening assay
GB0716322D0 (en) * 2007-08-21 2007-10-03 Cancer Rec Tech Ltd Crystal structure
US9873718B2 (en) 2013-05-08 2018-01-23 The Regents Of The University Of California Structure-based peptide inhibitors of P53 aggregation as a new approach to cancer therapeutics
WO2017059239A1 (en) * 2015-10-02 2017-04-06 Arizona Board Of Regents On Behalf Of Arizona State University Methods for processing biopolymeric arrays

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2001774C (en) 1988-10-28 2001-10-16 James A. Wells Method for identifying active domains and amino acid residues in polypeptides and hormone variants
JP2919890B2 (en) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル Single domain ligand, receptor consisting of the ligand, method for producing the same, and use of the ligand and the receptor
US6291158B1 (en) 1989-05-16 2001-09-18 Scripps Research Institute Method for tapping the immunological repertoire
PT918874E (en) 1996-08-16 2001-08-30 Medical Res Council EPISOMIC AUTHORSPRODUCTION EXPRESSION VECTORS CONFERING A GENERA SPECIFIC EXPRESSION OF THE FABRICS
KR100272659B1 (en) 1997-06-28 2000-12-01 김영환 Pattern for measuring line width of a metal line of a semiconductor device and method for measuring the same
JP2002511259A (en) 1998-04-09 2002-04-16 ジェンセット 5 'ESTs and encoded human proteins
US6582933B1 (en) * 1998-12-03 2003-06-24 Ono Pharmaceutical Co., Ltd. Rel1A binding inhibitor, process for producing the same and utilization thereof
CA2364650A1 (en) 1999-03-12 2000-09-21 Craig A. Rosen 50 human secreted proteins
DE69934259T2 (en) * 1999-07-23 2007-05-31 Glaxo Group Ltd., Greenford Combination of an anti-Ep-CAM antibody with a chemotherapeutic agent
CA2417368A1 (en) 2000-08-04 2002-02-14 Xin Lu Suppressor gene

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9527896B2 (en) 2007-01-31 2016-12-27 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US8889632B2 (en) 2007-01-31 2014-11-18 Dana-Farber Cancer Institute, Inc. Stabilized p53 peptides and uses thereof
US10202431B2 (en) 2007-01-31 2019-02-12 Aileron Therapeutics, Inc. Stabilized P53 peptides and uses thereof
US20100168388A1 (en) * 2007-01-31 2010-07-01 Federico Bernal Stabilized p53 peptides and uses thereof
US10301351B2 (en) 2007-03-28 2019-05-28 President And Fellows Of Harvard College Stitched polypeptides
US9957299B2 (en) 2010-08-13 2018-05-01 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8859723B2 (en) 2010-08-13 2014-10-14 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9522947B2 (en) 2011-10-18 2016-12-20 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US9096684B2 (en) 2011-10-18 2015-08-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10308699B2 (en) 2011-10-18 2019-06-04 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8927500B2 (en) 2012-02-15 2015-01-06 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US8987414B2 (en) 2012-02-15 2015-03-24 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US9505804B2 (en) 2012-02-15 2016-11-29 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10213477B2 (en) 2012-02-15 2019-02-26 Aileron Therapeutics, Inc. Peptidomimetic macrocycles
US10227380B2 (en) 2012-02-15 2019-03-12 Aileron Therapeutics, Inc. Triazole-crosslinked and thioether-crosslinked peptidomimetic macrocycles
US9880171B2 (en) 2012-03-02 2018-01-30 Ludwig Institute For Cancer Research Ltd. iASPP phosphorylation and metastatic potential
US9604919B2 (en) 2012-11-01 2017-03-28 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10669230B2 (en) 2012-11-01 2020-06-02 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US9845287B2 (en) 2012-11-01 2017-12-19 Aileron Therapeutics, Inc. Disubstituted amino acids and methods of preparation and use thereof
US10471120B2 (en) 2014-09-24 2019-11-12 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10905739B2 (en) 2014-09-24 2021-02-02 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and formulations thereof
US10253067B2 (en) 2015-03-20 2019-04-09 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof
US10023613B2 (en) 2015-09-10 2018-07-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles as modulators of MCL-1
US11091522B2 (en) 2018-07-23 2021-08-17 Aileron Therapeutics, Inc. Peptidomimetic macrocycles and uses thereof

Also Published As

Publication number Publication date
EP1558638A2 (en) 2005-08-03
WO2004031229A2 (en) 2004-04-15
HK1083220A1 (en) 2006-06-30
AU2003299132A1 (en) 2004-04-23
CA2487206A1 (en) 2004-04-15
EP1947113A1 (en) 2008-07-23
ATE537186T1 (en) 2011-12-15
WO2004031229A3 (en) 2004-09-23
US7745571B2 (en) 2010-06-29
ATE425183T1 (en) 2009-03-15
DE60326623D1 (en) 2009-04-23
EP1558638B1 (en) 2009-03-11
JP2006517787A (en) 2006-08-03
EP1947113B1 (en) 2011-12-14
US20080260756A1 (en) 2008-10-23

Similar Documents

Publication Publication Date Title
US7745571B2 (en) Peptide inhibitors of IASPP
US7470721B2 (en) Compositions and methods for identifying agents which modulate PTEN function and PI-3 kinase pathways
Hong et al. USP7, a ubiquitin-specific protease, interacts with ataxin-1, the SCA1 gene product
US20120251543A1 (en) Methods and compositions for modulating apoptosis
EP2039367B1 (en) Prophylactic/therapeutic agent for neurodegenerative disease
EP2328917B1 (en) Neuronal viability factor and use thereof
US20060127397A1 (en) RAG polypeptides, nucleic acids, and their use
US20040228866A1 (en) Suppressor genes
JP4314386B2 (en) Bcl-2 regulator (BMF) sequences and their use in the regulation of apoptosis
US6586203B1 (en) ARF-P19, a novel regulator of the mammalian cell cycle
US8492106B2 (en) Identification methods for ergothioneine transporter modulators and therapeutic uses thereof
US20030181380A1 (en) Modulating lymphoid commitment and survival
US8404806B2 (en) Isolated BRCA1 peptides and method of use
JP4375960B2 (en) Modification of biological metabolism
TWI250209B (en) A novel G protein-coupled receptor, GAVE8
JPWO2003030936A1 (en) Life-style related diseases or anorexia remedies and screening methods thereof
US6770458B1 (en) Purified and isolated serine-threonine kinase receptors associated protein and use of same in the modulation of the biological activity of TGF-β
KR20060129263A (en) Secreted neural apoptosis inhibiting proteins
US20060057153A1 (en) Method and composition for the inhibition of mitosis
US20040229797A1 (en) Proteins having effects of controlling cell migration and cell death
JP2005521424A (en) Novel pancortin-pabroprotein interactions and methods of their use
WO2000034310A9 (en) Purified and isolated serine-threonine kinase receptors associated protein
Hopp Regulation of apoptosis in photoreceptors by thebcl-2 gene family andp53 in mouse models of retinal degeneration

Legal Events

Date Code Title Description
AS Assignment

Owner name: LUDWIG INSTITUTE FOR CANCER RESEARCH, NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LU, XIN;REEL/FRAME:017033/0676

Effective date: 20051110

AS Assignment

Owner name: UCL CRUCIFORM LIMITED, ENGLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SELWOOD, DAVID;REEL/FRAME:017116/0344

Effective date: 20051109

AS Assignment

Owner name: GENOME RESEARCH LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KUWABARA, PATRICIA;REEL/FRAME:017309/0113

Effective date: 20060303

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION