[go: up one dir, main page]

WO2012112869A2 - Targeted cancer therapy using ets transcriptional inhibitors - Google Patents

Targeted cancer therapy using ets transcriptional inhibitors Download PDF

Info

Publication number
WO2012112869A2
WO2012112869A2 PCT/US2012/025608 US2012025608W WO2012112869A2 WO 2012112869 A2 WO2012112869 A2 WO 2012112869A2 US 2012025608 W US2012025608 W US 2012025608W WO 2012112869 A2 WO2012112869 A2 WO 2012112869A2
Authority
WO
WIPO (PCT)
Prior art keywords
tel
polypeptide
domain
seq
amino acid
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.)
Ceased
Application number
PCT/US2012/025608
Other languages
French (fr)
Other versions
WO2012112869A3 (en
Inventor
Xiaobo CAO
William Roy SMYTHE
Dawit GIZACHEW
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.)
Baylor Scott and White Health
Original Assignee
Scott and White Healthcare
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Scott and White Healthcare filed Critical Scott and White Healthcare
Publication of WO2012112869A2 publication Critical patent/WO2012112869A2/en
Publication of WO2012112869A3 publication Critical patent/WO2012112869A3/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention generally concerns the fields of cell biology, molecular biology, and medicine, including cancer medicine.
  • the present invention concerns mutants of an E-twenty six (ETS) transcriptional repressor protein for cancer therapy.
  • ETS E-twenty six
  • MM malignant mesothelioma
  • non-small cell lung cancer Dazzi et al, 1990; Herbst and Shin, 2002; Sridhar et al, 2003; Rogers et al, 2009; Mukohara et al, 2005.
  • EGFR, c-Met (HGFR) and IGF1R may cooperate to regulate tumor growth and survival.
  • the inhibition of one receptor can be compensated by activation of the others, because there is a degree of functional redundancy between these receptors (Bachleitner-Hofmann et al, 2008; Guo et al., 2008; Whittaker et al., 2010).
  • the complex crosstalk between these pathways could therefore significantly reduce the efficacy of single receptor targeted therapies. Therefore, elucidating and targeting common downstream effectors is of some importance.
  • the ETS family consists of more than 30 members, which include both transcription factors and repressors (Silverman et al., 2002; Wei et al., 2010).
  • Most ETS proteins are nuclear targets of diverse signaling pathways, such as the mitogen- activated protein (MAP) kinase signaling pathway, and undergo post-translational modifications including phosphorylation, glycosylation, acetylation, ubiquitination, and sumoylation (Cao et al., 2009). These modifications have a profound impact on the activity and subcellular localization of all ETS proteins.
  • ETS-1, 2 and PU are nuclear targets of diverse signaling pathways, such as the mitogen- activated protein (MAP) kinase signaling pathway.
  • l which are known positive transcriptional regulators of Bcl-xl, are imported to the nucleus following MAPK phosphorylation, and can be exported by an active transport pathway (Sevilla et al., 2001; Sevilla et al., 1999; Nelson et al., 2010).
  • Tel is a transcriptional repressor of Bcl-xl, and is also negatively regulated by phosphorylation and sumoylation, by nuclear export, or as a result of localization to Tel-bodies (Roukens et al., 2008; Maki et al., 2004). Tel interactions with relevant corepressors, mSin3A, N-CoR and HDAC3, are believed to mediate the transcriptional repression of multiple target genes, including Bcl-xl (Wang et al., 2001).
  • the present invention concerns methods and/or compositions related to E-twenty six (ETS) protein inhibitors for cancer therapy and/or prevention in a mammal.
  • Tel a transcription repressor member in the ETS family
  • Tel has been identified as a tumor suppressor. Delivery of Tel into tumor cells leads to tumor cell death, growth arrest and inhibition of invasion. Tel can serve as a pan ETS inhibitor, because it competes with other members of ETS family proteins on similar ETS binding sites.
  • there is a mutated ETS transcriptional repressor protein that is capable of binding a particular DNA promoter sequence and that abolishes other ETS transcription factors' activities.
  • ETS proteins are nuclear targets of diverse signaling pathways, such as the mitogen-activated protein (MAP) kinase signaling pathway, and undergo post-translational modifications, including phosphorylation, glycosylation, acetylation, ubiquitination, and sumoylation, for example.
  • Tel is a transcriptional repressor that is negatively regulated by phosphorylation and sumoylation, by nuclear export, and/or as a result of localization to Tel- bodies, for example.
  • Tel interactions with relevant co-repressors including mSin3A, N-CoR and HDAC3, mediate the transcriptional repression of multiple target genes, including Bcl-xl, in certain aspects of the invention.
  • the present invention there are Tel repressors that have altered post-transcriptional modifications compared to wild-type Tel.
  • the modifications can occur at one or more of the sites for ubiquitination, sumoylation, phosphorylation, and/or auto- inhibition, in certain embodiments.
  • the transcriptional repressor activity of the mutant Tel is greater than that for wild-type Tel.
  • the mutants have enhanced Tel repressive function compared to wild-type Tel.
  • Tel is negatively regulated at the post-transcriptional level, such as through ubiquitination, sumoylation, phosphorylation, and/or auto-inhibition.
  • Tel can be modified by sumoylation at Kl l and K99 and can be modified by phosphorylation at S213.
  • the exemplary results provided herein indicate that the phosphorylation of Tel at Ser213 blocks the Tel's repression on Bcl-xl transcription. This negative regulation of Tel function following receptor activation contributes to the overexpression of the Bcl-xl protein in cancer cells.
  • the one or more mutations provide enhanced repressor activity, such as compared to wild type.
  • a Tel mutant having two or more mutations has greater repressor activity compared to the single mutant activity.
  • the exemplary double mutant Tel K11R/S213A is a stronger repressor of Bcl-xl transcription (for example) than either single mutation alone. Whereas both phosphorylation and sumoylation are useful for Tel export, for example, in certain embodiments these two types of modification function cooperatively or independently.
  • the phosphorylation and sumoylation work synergistically, for example to mediate the downregulatory nuclear export of the ETS transcriptional repressors.
  • the mutant Tel polypeptide is hypophosphorylated compared to wild-type Tel.
  • Exemplary phosphorylation sites on Tel reside within the linker inhibitory damper (LID) domain outside of its ETS DNA-binding domain.
  • LID linker inhibitory damper
  • the Tel polypeptide may have mutations of any suitable kind, but in specific embodiments the polypeptide retains transcriptional repressor activity, including activity that is greater than wild-type when measured by identical or similar assays.
  • the mutation may be a point mutation, inversion, or deletion, for example.
  • the mutation may alter the ability to be subject to one or more post-translational modifications.
  • the amino acid that may be mutated such that phosphorylation cannot occur includes serine, tyrosine, or threonine, and one or more of these amino acids may be changed to another amino acid, particularly one that is not capable of being phosphorylated.
  • the mutation may change the respective encoded amino acid, but in some cases there may be mutations that do not change the respective encoded amino acid.
  • Tel mutants encompassed by the invention also include those that lack part of the protein.
  • the mutant may lack the N-terminal and/or C-terminal.
  • the protein lacks part or all of the inhibitory CID domain.
  • the protein is modified such that it has greater protein solubility than wild type.
  • the Tel protein has single, double, or triple or more modifications that render greater solubility for the protein.
  • the Tel V112E mutant has increased protein solubility.
  • a protein transduction domain (such as the TAT peptide sequence or Antennapedia homeodomain, for example), for example, is present on Tel to enable the polypeptide to penetrate into human cells.
  • the protein transduction domain is present on the N-terminus of the Tel mutant, whereas in other cases the protein transduction domain is present on the C-terminus of the Tel mutant.
  • the methods and compositions of the invention are employed for cancer treatment and/or prevention, wherein the cancer is of any kind of cancer.
  • the cancer is mesothelioma, thoracic cancer (including lung cancer, such as non- small cell lung carcinoma, for example), breast, colon, pancreas, prostate, skin, liver, kidney, bone, spleen, cervix, ovary, testes, bladder, gall bladder, or brain cancer.
  • the individual to be treated with the invention is a mammal, including a human, dog, cat, horse, pig, sheep, or goat, for example.
  • TEL polypeptide having one or both of the following: a) an amino acid substitution at one or more sites corresponding to SEQ ID NO:3, said sites selected from the group consisting of a serine corresponding to position 213, a serine corresponding to position 257, a lysine corresponding to position 11, a lysine corresponding to position 99, and a valine corresponding to position 112; and/or b) a deletion in the polypeptide corresponding to SEQ ID NO:3, said deletion occurring in one or more of a protein transduction domain, a TAT domain, a SAM domain, a LID domain, an ETS domain, or a CID domain, wherein the polypeptide has transcriptional repressor activity.
  • the polypeptide has increased transcriptional repressor activity over wild- type TEL polypeptide.
  • the polypeptide is further defined as having an amino acid substitution at the serine corresponding to position 213, at the serine corresponding to position 257, at the lysine corresponding to position 11, at the lysine corresponding to position 99, and/or at the lysine corresponding to position 99.
  • the substitution at the serine corresponding to position 213 is alanine
  • the substitution at the serine corresponding to position 257 is alanine
  • the substitution at the lysine corresponding to position 11 is arginine
  • the substitution at the lysine corresponding to position 99 is arginine.
  • the polypeptide further comprises a protein transduction domain.
  • TEL polypeptide having a mutation at a phosphorylation site, a ubiquitination site, a sumoylation site, a glycosylation site, an acetylation site, or a combination thereof, wherein the polypeptide has transcriptional repressor activity.
  • the polypeptide has increased transcriptional repressor activity over wild-type TEL polypeptide.
  • the mutation occurs within a C-terminal inhibitory domain (CID), a linker inhibitory damper domain (LID), ETS DNA-binding domain (ETS), or a sterile alpha motif (SAM) domain.
  • CID C-terminal inhibitory domain
  • LID linker inhibitory damper domain
  • ETS ETS DNA-binding domain
  • SAM sterile alpha motif
  • the polypeptide may be further defined as comprising (corresponding to positions in SEQ ID NO:3) a mutation at serine 213, comprising a mutation at serine 257, comprising a mutation at valine 112, and/or having a mutation at lysine 11.
  • the polypeptide may further comprise deletion of part or all of the CID domain and/or the polypeptide may further comprise a protein transduction domain, such as from HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat- (35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1, or Pep-2, for example.
  • a protein transduction domain such as from HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat- (35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1,
  • R0 is H or an amino acid sequence comprising 1 to 500 consecutive amino acids
  • Rl is an amino acid sequence comprising 1 to 11 consecutive amino acid residues of a protein transduction domain
  • R2 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a SAM domain
  • R3 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a LID domain
  • R4 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of an ETS domain
  • R5 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a CID domain
  • R6 is H or an amino acid sequence comprising 1 to 500 consecutive amino acids, provided that R2, R3, R4, and R5 are not all bonds, provided that the isolated recombinant amino acid sequence
  • Rl is selected from the group consisting of SEQ ID NO:5-23; R2 comprises SEQ ID NO:24; R3 comprises SEQ ID NO:25; R4 comprises SEQ ID NO:26; and/or R5 comprises SEQ ID NO:27.
  • the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:28.
  • R2, R3, and R4 are not bonds but R5 is a bond.
  • Rl comprises a sequence selected from the group consisting of SEQ ID NO:5-23;
  • R2 comprises SEQ ID NO:24;
  • R3 comprises SEQ ID NO:25;
  • R4 comprises SEQ ID NO:26.
  • the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:29.
  • R2 is a bond and R3, R4, and R5 are not bonds.
  • the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:30.
  • R2 and R5 are bonds and R3 and R4 are not bonds.
  • R2 is a bond and R3, R4 and R5 are not bonds.
  • the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:32. In some cases,
  • Rl comprises a TAT sequence (SEQ ID NO:6); R2 comprises SEQ ID NO:35; R2 comprises SEQ ID NO:36; R2 comprises SEQ ID NO:37; R3 comprises SEQ ID NO:38; R3 comprises SEQ ID NO:39; and/or R3 comprises SEQ ID NO:40.
  • wild-type Tel without at least part of the CID domain is employed.
  • there is a Tel lacking the CID domain but the remainder of the Tel protein variant is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the corresponding region in the wild-type Tel protein.
  • there is a Tel lacking the CID domain but the remainder of the Tel protein variant has one, two, three, four, five, or more amino acid substitutions or deletions.
  • there is a Tel lacking the CID domain but the remainder of the Tel protein variant has no more than one, two, three, four, five, or more amino acid substitutions or deletions.
  • there is a Tel lacking the CID domain but the remainder of the Tel protein variant has at least one, two, three, four, five, or more amino acid substitutions or deletions.
  • all of the CID domain is absent from the Tel protein, whereas in certain cases part of the CID domain is absent, such as 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid(s) from the CID domain are absent.
  • a Tel protein comprising TAT, the V112E mutation, and/or lacking part or all of the CID domain is employed.
  • An exemplary TAT-Tel wild-type with V112E but without CID is provided in SEQ ID NO:43 and may be employed in the invention, in certain embodiments.
  • a recombinant TEL polypeptide comprising an ETS DNA-binding domain (ETS) and a) a sterile alpha motif (SAM) domain having a deletion or an amino acid substitution at a lysine or a valine; and/or b) a linker inhibitory damper domain (LID) domain having a deletion or an amino acid substitution at one or more serines; and/or c) an amino acid substitution at a lysine corresponding to position 11 in SEQ ID NO:3.
  • the polypeptide further comprises a protein transduction domain, a nuclear localization signal, and/or a C-terminal inhibitory domain (CID) domain.
  • the polypeptide has increased transcriptional repressor activity over wild-type TEL polypeptide.
  • the polypeptide may be further defined as having an amino acid substitution at the serine corresponding to position 213 in SEQ ID NO:3, as having an amino acid substitution at the serine corresponding to position 257 in SEQ ID NO:3, as having an amino acid substitution at the lysine corresponding to position 99 in SEQ ID NO:3, and/or as having an amino acid substitution at the valine corresponding to position 112 in SEQ ID NO:3.
  • Exemplary protein transduction domains utilized in the invention include, for example, HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat- (35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1, or Pep-2.
  • an isolated recombinant TEL polypeptide comprising: a) an amino acid substitution at one or more sites in the polypeptide corresponding to SEQ ID NO:3, said sites selected from the group consisting of a serine corresponding to position 213, a serine corresponding to position 257, a lysine corresponding to position 11, a lysine corresponding to position 99, and a valine corresponding to position 112; and/or b) a deletion in the polypeptide corresponding to SEQ ID NO:3, said deletion occurring in one or more of a protein transduction domain, a TAT domain, a SAM domain, a LID domain, an ETS domain, or a CID domain, wherein the polypeptide has transcriptional repressor activity.
  • the method comprises the step of administering to the individual a therapeutically effective amount of a polypeptide of the invention.
  • the individual has a thoracic malignancy.
  • the individual has mesothelioma.
  • the method comprises an additional therapy for cancer.
  • Kits that comprise the polypeptide of the invention are encompassed, said polypeptide housed in a suitable container.
  • FIG. 1A provides that stimulation of cells by HGF and EGF causes identical phosphorylation events by multiple kinases as measured using the proteome profile human phosphor-MAPK array.
  • FIG IB shows that activated Map kinase phosphorylates ETS proteins including Tel.
  • the mesothelioma cell line, 145 was grown under conditions of serum starvation or serum starvation plus 20 minutes of HGF (100 ng/ml) stimulation.
  • the endogenous ETS-2, PU.l and Tel proteins were immunoprecipitated from the cell lysates using the respective antibodies. The immunoprecipitates were then analyzed by western blot using phosphor-Ser/Thr antibodies.
  • Tel is phosphorylated on Serine 213 after HGF stimulation.
  • the Tel serine mutant S213A, S257A and wild- type Tel were transfected into 145 cells which were then serum starved or HGF stimulated. Tel was immunoprecipitated from cell lysates and its phosphorylation status was determined using anti phosphor-Ser/Thr antibodies.
  • FIG. 2 shows that CHIP (IP) assay evaluation of the impact of Tel phosphorylation on its binding to the Bcl-xl promoter.
  • 145 and A549 cells transfected with wild- type or Tel S213A mutants were serum-starved for 24 hours and then subjected to treatment with or without HGF or EGF for 20 minutes.
  • Cross-linked Tel-DNA complexes were immunoprecipitated using a Tel antibody and Bcl-xl promoter fragments were PCR amplified.
  • FIG. 3 A demonstrates that the Tel phosphorylation site mutant S213A is not exported from the nucleus after EGF stimulation.
  • 145 cells were transfected with Tel or Tel S213A expression vectors and 24 hours after serum starvation, these transfected cells were exposed to HGF stimulation for 20 minutes. Acetone-fixed cells were then incubated with Tel antibodies followed by a fluorescein isothiocyanate (FITC)-labeled secondary antibody. Fluorescein signals were visualized under a Nikon 80i fluorescent microscope. 4,6-Diamidino-2- phenylindole (DAPI) was used as a counterstain for nuclear DNA.
  • FIG. 3B Tel S213A achieves a stronger repression of the Bcl-xl promoter in the presence of growth factor activation.
  • A549 and 145 cells seeded in 24-well plates were cotransfected with Bcl-xl promoter pXL-luc and either Tel S213A or Tel vectors along with p-CMV-P-galactosidase. Twenty-four hours after transfection, the cells were lysed, and luciferase and ⁇ -galactosidase activities were measured with a luminometer. The luciferase activities were normalized to those of ⁇ -galactosidase, and the data shown are the average of triplicate determinations. This experiment was repeated twice.
  • FIG. 4A shows that growth factor stimulation of cells fails to decrease the repression of Bcl-xl by Tel S213A.
  • 145 mesothelioma cells were transfected with GFP, Tel wild type or Tel S213A and grown under conditions of serum starvation or serum starvation plus HGF stimulation (100 ng/ml) for 48 hours. The Bcl-xl expression levels in these cells were then determined by western blotting.
  • FIG. 4B To determine the gene targets of Tel repression, both A549 and 145 cells were transfected with GFP, Tel wild type or Tel S213A. Forty-eight hours after transfection the Blc-2 target protein levels were detected by western blotting.
  • FIG. 5A demonstrates measurement of the short-term tumor growth suppression of Tel S213A by cell proliferation assay.
  • A549 and 145 cells were seeded in 96 well plates and transfected with 0.2 ⁇ g of Tel or Tel S213A cDNA, and cultured for 48 hours. Cell growth inhibition was measured by the XTT assay. The data shown are the average of four independent determinations.
  • FIG. 5B Demonstration of long-term tumor growth suppression by Tel S213A in a colony formation assay.
  • A549 and 145 cells were transfected with various doses of Tel or Tel S213A cDNA and plated in 10 cm cell culture plates at 10 4 cells per plate.
  • Tel S213A induces a stronger apoptotic response in cancer cells.
  • A549 cells were transfected with pGL-2, Tel wild type and Tel S213A, collected and stained with FITC-labeled Annexin V and PL Annexin V-positive populations were quantified by flow cytometry. The data shown are the results of triplicate determinations.
  • FIG. 6A illustrates the impact of multiple-site mutations of Tel on Bcl-xl promoter repression.
  • A549 cells were seeded in 96-well plates and transfected with a Bcl-xl promoter reporter, pXL-luc, and co-transfected with Tel wild-type, Tel K11R, Tel S213A, Tel Kl lR, Tel K11R/S213A and a p-CMV-P-galactosidase transfection control vector.
  • the cells were lysed and both the luciferase and ⁇ -galactosidase activities were measured using a luminometer.
  • FIG. 6B Comparison of Bcl-xl protein levels following transfection with the indicated Tel mutants. A549 cells were seeded into 6 well plates (10 6 cells per well) and were transfected with Tel, Tel mutants and GFP expression vectors. Forty-eight hours after transfection, the Bcl-xl protein levels were determined by western blot.
  • FIG. 7 shows analysis of EGFR phosphorylation and Bcl-xl using human non-small cell lung carcinoma and mesothelioma tissue arrays. Correlations between the Bcl-xl levels and phosphorylated EGFR levels were demonstrated by Chi-square analysis (P ⁇ 0.01).
  • FIG. 8 provides an exemplary schematic representation of the negative regulation of Tel by phosphorylation.
  • FIG. 9 illustrates exemplary modifications of wild-type TEL.
  • FIG. 10 provides the sequence of Tel, SEQ ID NO:3, which illustrates different exemplary domains in the Tel protein.
  • the ETS domain comprises amino acids 338-422, the figure marks it as 331-422 merely to illustrate a nuclear localization signal.
  • FIG. 11 illustrates one embodiment by which phosphorylations at S213/S257 decrease Tel's repression on Bcl-xl transcription and increase Bcl-xl transcription.
  • FIG. 12 shows an embodiment of another mechanism by which LID can repress CID.
  • FIG. 13 illustrates an embodiment wherein there is synergism of sumoylation and phosphorylation on negative regulation of Tel function.
  • an effective amount or "therapeutically effective amount” as used herein is defined as an amount of the agent that will decrease, reduce, inhibit or otherwise abrogate the growth of a neoplasm, induce apoptosis, inhibit angiogenesis of a neoplasm, inhibit metastasis, or induce cytotoxicity in a neoplasm.
  • an effective amount is an amount sufficient to detectably and repeatedly ameliorate, reduce, minimize or limit the extent of the disease or its symptoms.
  • ETS transcriptional factors are molecular targets for selective cancer therapy, because they play roles in maintaining malignancy of cancer cells. It is difficult to develop small molecules that directly inhibit the function of ETS transcription factors, because these proteins bind to the same DNA sequence in humans, and the loss of function of one transcription factor due to a small molecular inhibitor, for example, can be compensated by other ETS transcription factors. This difficulty can be supported by the ineffectiveness of dominant negative mutant and siRNA/antisense approaches, for example.
  • the present invention provides novel ETS transcriptional inhibitors and encompasses Tel polypeptides that are modified compared to wild type but retain transcriptional repressor activity, including enhanced transcriptional repressor activity compared to wild type, for example.
  • the Tel gene can be negatively regulated at the post-transcriptional level through ubiquitination, glycosylation, acetylation, sumoylation and phosphorylation, for example.
  • the mutant may have a single mutation or more than one mutation.
  • the exemplary double-mutants TEL 11-213 enhance Tel's repressive function in tumor cells by abolishing one or more negative modifications, in certain embodiments.
  • the present invention includes mutants with increased protein solubility, including the Tel V112E mutant having increased protein solubility.
  • the invention also includes mutants with improved binding to promoter DNA compared to wild type Tel including, for example, a mutant with a deleted part or all CID domain that improves its binding to promoter DNA.
  • the Tel ETS domain is a therapeutic protein, in certain embodiments.
  • TAT peptide sequence is attached at its N terminus to enable protein to penetrate into human cells, in particular aspects of the invention.
  • the phosphorylation of Tel increases Bcl-xl transcription through tyrosine kinase receptors' phosphorylation and activation of ETS2, a transcriptional activator.
  • stimulation by HGF or EGF activates an identical downstream signal transduction pathway and causes the activation of ERK, leading to the nuclear export of Tel.
  • the exemplary phosphorylation site mutant TelS213A enhances Tel repression of Bcl-xl transcriptional activity.
  • the nuclear localization of the Tel S213A mutant was detected under HGF/EGF exposure.
  • a luciferase reporter assay revealed that the transient expression of Tel S213A results in further repression of Bcl-xl promoter activity.
  • the present invention encompasses the impact of the activation of tyrosine kinase receptors upon transcriptional regulation, including Bcl-xl transcriptional regulation.
  • Tel Ser213 phosphorylation is responsible for receptor kinase activation via the MEK-ERK pathway in cancer cells.
  • At least the phosphorylation of Tel at Ser213 blocks the repression of Bcl-xl transcription by Tel. This negative regulation of Tel function following receptor activiation contributes to the overexpression of the prosurvival Bcl-xl protein in cancer cells.
  • Tel Protein and Exemplary Modifications Thereof there are mutants of Tel protein utilized in methods and/or compositions for cancer therapy and/or prevention.
  • the Tel protein mutants may be of any kind, but in particular aspects there are one, two, three, four, five, or more mutations compared to the wild-type Tel protein.
  • the mutation(s) result in a functionally detectable difference compared to the wild-type protein, although in some cases the mutations result in a Tel protein having no detectable functional difference compared to the wild-type protein.
  • the function that is characterized is transcriptional repressor activity.
  • the function that is characterized is as an inducer of apoptosis.
  • both the transcriptional repressor activity and the apoptotic inducer functions are characterized.
  • Measurement of transcriptional repressor activity may occur by any suitable means in the art, but in specific embodiments the measurement comprises cell transfection with the candidate mutant in a cell having detectable transcription of a reporter polynucleotide or other polynucleotide, followed by Western or northern. Transcription repression can be measured by luciferase assay using several genes' promoter constructs.
  • electrophoresis mobility shift assay (EMSA) and Chromatin immunoprecipitation (ChIP) can be used to measure the interaction between transcription factor or repressor and DNA promoters.
  • Real-time RT-PCR also can be adopted to measure the mRNAs levels of multiple ETV6/TEL regulated genes.
  • Measurement of apoptosis inducer activity may occur by any suitable means in the art, but in specific embodiments the measurement comprises XTT assay (conversion of the water-soluble XTT assay (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5- carboxanilide) reagent to an orange formazan product by actively respiring cells); colony formation assay; Annexin V binding; and/or flow cytometry. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay can also be applied.
  • TUNEL Terminal deoxynucleotidyl transferase dUTP nick end labeling
  • One or more amino acids of Tel may be mutated, in certain aspects of the invention, and in some cases, the amino acid is within a functional domain, for example related to a particular activity for the protein.
  • the mutation may be in the C-terminal inhibitory domain, CID, (amino acids 423-452 of the human wild-type protein), which interferes with the DNA binding domain.
  • CID C-terminal inhibitory domain
  • An additional example of a domain that might be inhibited includes the linker inhibitory damper domain, LID, (amino acids 124-338 of the human wild- type protein, and S213 and S257 are present therein).
  • SAM domain amino acids 41-123, and K99 is present therein
  • ETS domain amino acids 338-422
  • CTBP1 amino acids 423-427
  • Exemplary sequences for wild- type Tel domains are as follows: SEQ ID NO:24 (SAM); SEQ ID NO:25 (LID); SEQ ID NO:26 (ETS); SEQ ID NO:27 (CID). Sequence for an exemplary construct having TAT, SAM, LID, ETS, and CID is in SEQ ID NO:28. Sequence for an exemplary construct having TAT, SAM, LID, and ETS is in SEQ ID NO:29. Sequence for an exemplary construct having TAT, LID, ETS, and CID is in SEQ ID NO:30. Sequence for an exemplary construct having TAT, LID, and ETS is in SEQ ID NO:31.
  • Sequence for an exemplary construct having TAT, ETS, and CID is in SEQ ID NO:32. Sequence for an exemplary construct having TAT and ETS is in SEQ ID NO:33.
  • Exemplary sequences for mutated Tel domains are as follows: SEQ ID NO:35 (SAM having K99R mutation); SEQ ID NO:36 (SAM having V112E mutation); SEQ ID NO:37 (SAM having K99R and V112E mutations); SEQ ID NO:38 (LID having S213A mutation); SEQ ID NO:39 (LID having S257A mutation); and SEQ ID NO:40 (LID having S213A and S257A mutations).
  • Exemplary nuclear localization signals are provided in SEQ ID NO:34 (from Tel, located in the LID domain); SEQ ID NO:41 (ETS1 NLS signal); and SEQ ID NO:42 (SV40 large antigen NLS signal).
  • an "amino acid” can include any naturally or non-naturally occurring amino acid that is phosphorylated, ubiquinated, sumoylated, glycosylated, acetylated and that any amino acid sequence of the present invention may include one or a combination of such amino acids.
  • Specific naturally- occurring amino acids (or moieties of naturally occurring amino acids) that are capable of particular modifications are as follows: serine, threonine, and tyrosine can be phosphorylated; lysine can be ubiquinated, and lysine can be sumoylated.
  • N-linked glycans attach to a nitrogen of asparagine or arginine side-chains; O- linked glycans attach to the hydroxy oxygen of serine, threonine, tyrosine, hydroxylysine, or hydroxyproline side-chains, or to oxygens on lipids such as ceramide; phospho-glycans linked through the phosphate of a phospho-serine; and C-linked glycans are a rare form of glycosylation where a sugar is added to a carbon on a tryptophan side-chain.
  • a lysine may be acetylated.
  • FIG. 9 illustrates exemplary modifications of wild-type TEL.
  • the polypeptide comprises a protein transduction domain, for example on the N-terminus or C-terminus.
  • FIG. 10 provides SEQ ID NO:3, which illustrates different exemplary domains in the Tel protein.
  • the wild-type protein amino acid sequence is also at GenBank® Accession No. P41212 (SEQ ID NO:3), which is incorporated by reference herein in its entirety.
  • Other names for Tel include at least transcription factor ETV6, ETS translocation variant 6, and ETV6.
  • Tel2 which is a naturally occurring variant of Tell having a truncation on the N-terminus, may be employed in the invention, such as the modification occurring in a Tel2 polypeptide sequence (NP_057219 in GenBank® is the protein sequence, incorporated by reference herein; SEQ ID NO:4).
  • FIG. 11 illustrates one embodiment by which phosphorylations at S213/S257 decrease Tel's repression on Bcl-xl transcription and increase Bcl-xl transcription.
  • Tel is the bcl-xl transcriptional repressor.
  • CID within the Tel protein, has an inhibitory interaction with Tel's ETS domain. The existence of CID can weaken Tel-Bcl-xl promoter binding and promote the Tel moving away from promoter.
  • LID domain of Tel can negatively regulate CID's negative impact on Tel ETS domain, (as in, a negative of a negative is positive). Therefore Tel can repress Bcl-xl transcription at the promoter level. This mechanism is illustrated in FIG. 11 in the upper left picture.
  • CID can negatively regulate Tel ETS domain and promote Bcl-xl transcription (FIG. 11, bottom-right picture).
  • mutations at S213/S257 are useful to regulate Tel function. Blockage of phosphorylation of S213/S257 enhances Tel function, in particular aspects. Deletion of CID is also beneficial, in specific embodiments. K11/K99 are sumoylation sites, and in specific embodiments Kl l/99 sumoylation can promote Tel nuclear exportation, which abolishes Tel's repression. Therefore, in certain embodiments of the invevntion, sumoylation and phosphorylation of Tel are blocked to achieve enhanced Bcl-xl repression.
  • FIG. 13 illustrates an embodiment wherein there is synergism of sumoylation and phosphorylation on negative regulation of Tel function. It also illustrates that Tel as well as ETS 1/2 have several co-factors to work together at DNA promoter level.
  • the present invention concerns novel mutant Tel compositions comprising at least one proteinaceous molecule.
  • a "proteinaceous molecule,” “proteinaceous composition,” “proteinaceous compound,” “proteinaceous chain” or “proteinaceous material” generally refers, but is not limited to, a protein of greater than about 400 amino acids or the full length endogenous sequence translated from a gene; a polypeptide of greater than about 350 amino acids; a polypeptide of greater than about 300 amino acids; a polypeptide of greater than about 250 amino acids; a polypeptide of greater than about 200 amino acids; a polypeptide of greater than about 150 amino acids; and/or a peptide of from about 3 to about 100 amino acids. All the "proteinaceous” terms described above may be used interchangeably herein.
  • the size of the at least one proteinaceous molecule may comprise, but is not limited to, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59, about 60, about 61, about 62, about 63, about 64, about 65, about 66, about 67, about 68, about 69, about 70, about 71, about 72, about 73, about 74, about 75, about 76, about 77, about 78, about 79, about 80, about
  • an "amino molecule” refers to any amino acid, amino acid derivitive or amino acid mimic as would be known to one of ordinary skill in the art.
  • the residues of the proteinaceous molecule are sequential, without any non-amino molecule interrupting the sequence of amino molecule residues.
  • the sequence may comprise one or more non-amino molecule moieties.
  • the sequence of residues of the proteinaceous molecule may be interrupted by one or more non- amino molecule moieties.
  • the Tel molecule is modified such that it comprises a protein transduction domain (PTD), which may also be referred to as a cell- penetrating peptide, for example.
  • PTD protein transduction domain
  • exemplary cell penetrating peptides that are hydrophilic include the following:
  • R9-Tat GRRRRRRRRRPPQ (SEQ ID NO: 15)
  • Exemplary amphiphilic cell-penetrating peptides include the following:
  • FBP GALFLGWLGAAGSTMGAWSQPKKKRKV (SEQ ID NO: 19)
  • MPG ac-GALFLGFLGAAGSTMGAWSQPKKKRKV-cya (SEQ ID NO: 19)
  • Pep-2 ac-KETWFETWFTEWSQPKKKRKV-cya (SEQ ID NO:23)
  • Cya brings more positive charge to the peptide, which in certain embodiments helps the peptide penetrate through cell membrane.
  • proteinaceous composition encompasses amino molecule sequences comprising at least one of the 20 common amino acids in naturally synthesized proteins, or at least one modified or unusual amino acid, including but not limited to those shown on Table 1 below.
  • the proteinaceous composition comprises at least one protein, polypeptide or peptide.
  • the proteinaceous composition comprises a biocompatible protein, polypeptide or peptide.
  • biocompatible refers to a substance that produces no significant untoward effects when applied to, or administered to, a given organism according to the methods and amounts described herein. Organisms include, but are not limited to mammals, such as humans, dogs, cats, horses, pigs, goats, and sheep. Such untoward or undesirable effects are those such as significant toxicity or adverse immunological reactions.
  • biocompatible protein, polypeptide or peptide containing compositions will generally be mammalian proteins or peptides or synthetic proteins or peptides each essentially free from toxins, pathogens and harmful immunogens.
  • Proteinaceous compositions may be made by any technique known to those of skill in the art, including the expression of proteins, polypeptides or peptides through standard molecular biological techniques, the isolation of proteinaceous compounds from natural sources, or the chemical synthesis of proteinaceous materials.
  • the nucleotide and protein, polypeptide and peptide sequences for various genes have been previously disclosed, and may be found at computerized databases known to those of ordinary skill in the art.
  • One such database is the National Center for Biotechnology Information's GenBank® and GenPept databases.
  • the coding regions for these known genes may be amplified and/or expressed using the techniques disclosed herein or as would be known to those of ordinary skill in the art.
  • various commercial preparations of proteins, polypeptides and peptides are known to those of skill in the art.
  • a proteinaceous compound may be purified.
  • purified will refer to a specific or protein, polypeptide, or peptide composition that has been subjected to fractionation to remove various other proteins, polypeptides, or peptides, and which composition substantially retains its activity, as may be assessed, for example, by the protein assays, as would be known to one of ordinary skill in the art for the specific or desired protein, polypeptide or peptide.
  • any protein, polypeptide or peptide containing component may be used in the compositions and methods disclosed herein.
  • the proteinaceous material is biocompatible.
  • the formation of a more viscous composition will be advantageous in that will allow the composition to be more precisely or easily applied to the tissue and to be maintained in contact with the tissue throughout the procedure.
  • the use of a peptide composition, or more preferably, a polypeptide or protein composition is contemplated.
  • Ranges of viscosity include, but are not limited to, about 40 to about 100 poise. In certain aspects, a viscosity of about 80 to about 100 poise is preferred.
  • Proteins and peptides suitable for use in this invention may be autologous proteins or peptides, although the invention is clearly not limited to the use of such autologous proteins.
  • autologous protein, polypeptide or peptide refers to a protein, polypeptide or peptide which is derived or obtained from an organism.
  • Organisms that may be used include, but are not limited to, a bovine, a reptilian, an amphibian, a piscine, a rodent, an avian, a canine, a feline, a fungal, a plant, or a prokaryotic organism, with a selected animal or human subject being preferred.
  • the "autologous protein, polypeptide or peptide” may then be used as a component of a composition intended for application to the selected animal or human subject.
  • the autologous proteins or peptides are prepared, for example from whole plasma of the selected donor.
  • the plasma is placed in tubes and placed in a freezer at about 80°C for at least about 12 hours and then centrifuged at about 12,000 times g for about 15 minutes to obtain the precipitate.
  • the precipitate, such as fibrinogen may be stored for up to about one year (Oz, 1990).
  • compositions of the present invention are particularly suitable for use in tissue adhesion and wound healing
  • preferred proteins are contemplated.
  • Preferred protein include albumin, fibrinogen or gelatin, with albumin being most preferred.
  • a proteinaceous material that possesses one or more of the following characteristics: it forms a solution with a high percentage of protenaceous material solubilized; it possesses a high viscosity (i.e. about 40 to about 100 poise); it has the correct molecular charge to bind the dye if it is a non-covalent mixture (i.e. anionic protein and cationic dye, or cationic protein and anionic dye); it has the correct amino-acids present to form covalent cross-links (i.e.
  • tyrosines one or more tyrosines, histidines, tryptophans and/or methionines
  • biocompatible i.e. from mammalian origin for mammals, preferably from human origin for humans, from canine origin for canines, etc. ; it is autologous; it is non-allergenic, and/or it is non-immunogenic).
  • the biological functional equivalent may comprise a Tel polynucleotide that has been engineered to contain distinct sequences while at the same time retaining the capacity to encode the respective mutant protein. This can be accomplished to the degeneracy of the genetic code, i.e., the presence of multiple codons, which encode for the same amino acids.
  • one of skill in the art may wish to introduce a restriction enzyme recognition sequence into a polynucleotide while not disturbing the ability of that polynucleotide to encode a mutant Tel protein.
  • a polynucleotide may encode a biologically functionally equivalent polypeptide wherein certain amino acids may be substituted for other amino acids without appreciable loss of repressor activity and, in certain cases, with enhanced repressor activity.
  • these are "conservative" changes that do not disrupt the biological activity of the protein, as the structural change is not one that impinges of the protein's ability to carry out its designed function.
  • the change(s) is not conservative but still allows the polypeptide to retain functional activity. It is thus contemplated by the inventors that various changes may be made in the sequence of genes and proteins disclosed herein, while still fulfilling the goals of the present invention.
  • biologically functional equivalents it is well understood by the skilled artisan that, inherent in the definition of a "biologically functional equivalent" protein and/or polynucleotide, is the concept that there is a limit to the number of changes that may be made within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity.
  • Biologically functional equivalents are thus defined herein as those proteins (and polynucleotides) wherein selected amino acids (or codons) may be substituted.
  • Functional activity includes at least the ability to repress transcription of a polynucleotide, such as Bcl-xl, for example.
  • Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and/or the like.
  • An analysis of the size, shape and/or type of the amino acid side-chain substituents reveals that arginine, lysine and/or histidine are all positively charged residues; that alanine, glycine and/or serine are all a similar size; and/or that phenylalanine, tryptophan and/or tyrosine all have a generally similar shape.
  • arginine, lysine and/or histidine; alanine, glycine and/or serine; and/or phenylalanine, tryptophan and/or tyrosine; are defined herein as biologically functional equivalents.
  • hydropathic index of amino acids may be considered.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and/or charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine ( 0.4); threonine ( 0.7); serine ( 0.8); tryptophan ( 0.9); tyrosine ( 1.3); proline ( 1.6); histidine ( 3.2); glutamate ( 3.5); glutamine ( 3.5); aspartate ( 3.5); asparagine ( 3.5); lysine ( 3.9); and/or arginine ( 4.5).
  • the present invention in many aspects, relies on the synthesis of peptides and polypeptides in cyto, via transcription and translation of appropriate polynucleotides. These peptides and polypeptides will include the twenty "natural" amino acids, and post-translational modifications thereof. However, in vitro peptide synthesis permits the use of modified and/or unusual amino acids. A table of exemplary, but not limiting, modified and/or unusual amino acids is provided herein.
  • peptide mimetics that mimic elements of protein secondary and tertiary structure are described in Johnson et al. (1993).
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and/or antigen.
  • a peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule.
  • ⁇ -turns within proteins which are known to be highly antigenic. Likely ⁇ turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains.
  • Other approaches have focused on the use of small, multidisulfide- containing proteins as attractive structural templates for producing biologically active conformations that mimic the binding sites of large proteins. Vita et al. (1998). A structural motif that appears to be evolutionarily conserved in certain toxins is small (30-40 amino acids), stable, and high permissive for mutation. This motif is composed of a beta sheet and an alpha helix bridged in the interior core by three disulfides.
  • Beta II turns have been mimicked successfully using cyclic L-pentapeptides and those with D-amino acids.
  • Johannesson et al. (1999) report on bicyclic tripeptides with reverse turn inducing properties.
  • alpha-helix mimetics are disclosed in U.S. Patents 5,446,128; 5,710,245; 5,840,833; and 5,859,184. These structures render the peptide or protein more thermally stable, also increase resistance to proteolytic degradation. Six, seven, eleven, twelve, thirteen and fourteen membered ring structures are disclosed.
  • Beta- turns permit changed side substituents without having changes in corresponding backbone conformation, and have appropriate termini for incorporation into peptides by standard synthesis procedures.
  • Other types of mimetic turns include reverse and gamma turns. Reverse turn mimetics are disclosed in U.S. Patents 5,475,085 and 5,929,237, and gamma turn mimetics are described in U.S. Patents 5,672,681 and 5,674,976.
  • compositions of the Tel mutant protein of the invention it may be desirable to combine these compositions with other agents effective in the treatment of hyperproliferative disease, such as anti-cancer agents.
  • An "anticancer” agent is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, and/or increasing the lifespan of a subject with cancer.
  • these other compositions would be provided in a combined amount effective to kill or inhibit proliferation of the cell.
  • This process may involve contacting the cells with the expression construct and the agent(s) or multiple factor(s) at the same time. This may be achieved by contacting the cell with a single composition or pharmacological formulation that includes both agents, or by contacting the cell with two distinct compositions or formulations, at the same time, wherein one composition includes the expression construct and the other includes the second agent(s).
  • Tumor cell resistance to chemotherapy and radiotherapy agents represents a major problem in clinical oncology.
  • One goal of current cancer research is to find ways to improve the efficacy of cancer therapy by combining it with other cancer therapy.
  • Tel mutant therapy of the present invention could be used similarly in conjunction with chemotherapeutic, radiotherapeutic, immunotherapeutic, hormonal, or gene therapy intervention, for example.
  • the other cancer therapy may include surgery, radiation, immunotherapy, hormonal therapy, or chemotherapy, including carboplatin, cisplatin, docetaxel, erlotinib, etoposide, gemcitabine, irinotecan, paclitaxel, pemetrexed, topotecan, vinorelbine, and/or combinations thereof.
  • the other cancer therapy may include surgery, radiation, immunotherapy, hormonal therapy, or chemotherapy, such as cisplatin, raltitrexed, pemetrexed, gemcitabine, vinorelbine, carboplatin, and/or combinations thereof.
  • the present invention may be used simultaneously or may precede or follow the other agent treatment by intervals ranging from minutes to weeks.
  • the other agent and Tel mutant are applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the other agent and Tel mutant would still be able to exert an advantageously combined effect on the cell.
  • Tel mutant therapy is "A” and the secondary agent, such as radiation therapy or chemotherapy, for example, is "B":
  • Cancer therapies also include a variety of combination therapies with both chemical-and radiation-based treatments.
  • Combination chemotherapies include, for example, cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrexate, or any analog or derivative variant of the foregoing.
  • One or more chemotherapy agents may be included in the chemotherapy.
  • ⁇ -rays X-rays
  • X-rays X-rays
  • UV-irradiation UV-irradiation
  • Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens.
  • Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
  • contacted and “exposed,” when applied to a cell are used herein to describe the process by which a therapeutic construct and a chemotherapeutic or radiotherapeutic agent are delivered to a target cell or are placed in direct juxtaposition with the target cell.
  • both agents are delivered to a cell in a combined amount effective to kill the cell or prevent it from dividing.
  • Immunotherapeutics generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells.
  • the immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell.
  • the antibody alone may serve as an effector of therapy or it may recruit other cells to actually effect cell killing.
  • the antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent.
  • the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target.
  • Various effector cells include cytotoxic T cells and NK cells.
  • Immunotherapy thus, could be used as part of a combined therapy, in conjunction with the present therapy.
  • the general approach for combined therapy is discussed below.
  • the tumor cell must bear some marker that is amenable to targeting, i.e., is not present on the majority of other cells.
  • Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and pi 55.
  • the secondary treatment is a gene therapy in which a therapeutic polynucleotide is administered before, after, or at the same time as the Tel mutant.
  • a variety of proteins are encompassed within the invention, including antisense directed against inducers of cellular proliferation; inhibitors of cellular proliferation, such as tumor suppressors, including p53, pl6, C-CAM, Rb, APC, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, zacl, p73, VHL, MMAC1 / PTEN, DBCCR- 1, FCC, rsk-3, p27, p27/pl6 fusions, p21/p27 fusions, anti-thrombotic genes (e.g., COX-1, TFPI), PGS, Dp, E2F, ras, myc, neu, raf, erb, fms, trk, ret, gsp,
  • VEGF vascular endothelial growth factor
  • FGF FGF
  • thrombospondin BAI- 1, GDAIF, or their receptors
  • MCC MCC
  • regulators of programmed cell death such as members of the Bcl-2 family, ICE-like proteases, and so forth.
  • Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene therapy, immunotherapy and/or alternative therapies.
  • Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed.
  • Tumor resection refers to physical removal of at least part of a tumor.
  • treatment by surgery includes laser surgery, cryosurgery, electro surgery, and miscopically controlled surgery (Mohs' surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue.
  • a cavity may be formed in the body.
  • Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy.
  • Such treatment may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months.
  • These treatments may be of varying dosages as well.
  • agents may be used in combination with the present invention to improve the therapeutic efficacy of treatment.
  • additional agents include immunomodulatory agents, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adehesion, or agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers.
  • Immunomodulatory agents include tumor necrosis factor; interferon alpha, beta, and gamma; IL- 2 and other cytokines; F42K and other cytokine analogs; or MIP-1 , MIP-lbeta, MCP-1, RANTES, and other chemokines.
  • cell surface receptors or their ligands such as Fas / Fas ligand, DR4 or DR5 / TRAIL would potentiate the apoptotic inducing abililties of the present invention by establishment of an autocrine or paracrine effect on hyperproliferative cells. Increases intercellular signaling by elevating the number of GAP junctions would increase the anti-hyperproliferative effects on the neighboring hyperproliferative cell population.
  • cytostatic or differentiation agents can be used in combination with the present invention to improve the anti-hyerproliferative efficacy of the treatments. Inhibitors of cell adehesion are contemplated to improve the efficacy of the present invention.
  • cell adhesion inhibitors are focal adhesion kinase (FAKs) inhibitors and Lovastatin. It is further contemplated that other agents that increase the sensitivity of a hyperproliferative cell to apoptosis, such as the antibody c225, could be used in combination with the present invention to improve the treatment efficacy.
  • FAKs focal adhesion kinase
  • Lovastatin Lovastatin
  • Hormonal therapy may also be used in conjunction with the present invention or in combination with any other cancer therapy previously described.
  • the use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or estrogen. This treatment is often used in combination with at least one other cancer therapy as a treatment option or to reduce the risk of metastases.
  • compositions of the present invention comprise an effective amount of one or more Tel mutant compositions of the invention dissolved or dispersed in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate.
  • the preparation of an pharmaceutical composition that contains at least one composition of the present invention or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference.
  • for animal e.g.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the pharmaceutical compositions is contemplated.
  • composition of the present invention may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection.
  • the present invention can be administered intravenously, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, topically, intramuscularly, subcutaneously, mucosally, orally, topically, locally, inhalation (e.g. , aerosol inhalation), injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g. , liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference).
  • composition of the present invention may be formulated into a composition in a free base, neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts, e.g. , those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms such as formulated for parenteral administrations such as injectable solutions, or aerosols for delivery to the lungs, or formulated for alimentary administrations such as drug release capsules and the like.
  • the composition of the present invention suitable for administration is provided in a pharmaceutically acceptable carrier with or without an inert diluent.
  • the carrier should be assimilable and includes liquid, semisolid, i.e., pastes, or solid carriers. Except insofar as any conventional media, agent, diluent or carrier is detrimental to the recipient or to the therapeutic effectiveness of a the composition contained therein, its use in administrable composition for use in practicing the methods of the present invention is appropriate.
  • carriers or diluents include fats, oils, water, saline solutions, lipids, liposomes, resins, binders, fillers and the like, or combinations thereof.
  • composition may also comprise various antioxidants to retard oxidation of one or more component. Additionally, the prevention of the action of microorganisms can be brought about by preservatives such as various antibacterial and antifungal agents, including but not limited to parabens (e.g., methylparabens, propylparabens), chlorobutanol, phenol, sorbic acid, thimerosal or combinations thereof.
  • parabens e.g., methylparabens, propylparabens
  • chlorobutanol phenol
  • sorbic acid thimerosal or combinations thereof.
  • the composition is combined with the carrier in any convenient and practical manner, i.e., by solution, suspension, emulsification, admixture, encapsulation, absorption and the like. Such procedures are routine for those skilled in the art.
  • the composition is combined or mixed thoroughly with a semi- solid or solid carrier.
  • the mixing can be carried out in any convenient manner such as grinding.
  • Stabilizing agents can be also added in the mixing process in order to protect the composition from loss of therapeutic activity, i.e., denaturation in the stomach.
  • stabilizers for use in an the composition include buffers, amino acids such as glycine and lysine, carbohydrates such as dextrose, mannose, galactose, fructose, lactose, sucrose, maltose, sorbitol, mannitol, etc.
  • the present invention may concern the use of a pharmaceutical lipid vehicle compositions that include one or more compositions of the present invention, one or more lipids, and an aqueous solvent.
  • lipid will be defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds are well known to those of skill in the art, and as the term "lipid” is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man). However, a lipid is usually a biological substance.
  • Biological lipids are well known in the art, and include for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • neutral fats phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof.
  • lipids are also encompassed by the compositions and methods of the present invention.
  • composition of the present invention may be dispersed in a solution containing a lipid, dissolved with a lipid, emulsified with a lipid, mixed with a lipid, combined with a lipid, covalently bonded to a lipid, contained as a suspension in a lipid, contained or complexed with a micelle or liposome, or otherwise associated with a lipid or lipid structure by any means known to those of ordinary skill in the art.
  • the dispersion may or may not result in the formation of liposomes.
  • the actual dosage amount of a composition of the present invention administered to an animal patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
  • compositions may comprise, for example, at least about 0.1% of an active compound.
  • the an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.
  • the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein.
  • a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc. can be administered, based on the numbers described above.
  • the Tel mutant compositions of the present invention are formulated to be administered via an alimentary route.
  • Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually.
  • these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft- shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • the active compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like (Mathiowitz et ah, 1997; Hwang et ah, 1998; U.S. Pat. Nos. 5,641,515; 5,580,579 and 5,792, 451, each specifically incorporated herein by reference in its entirety).
  • the tablets, troches, pills, capsules and the like may also contain the following: a binder, such as, for example, gum tragacanth, acacia, cornstarch, gelatin or combinations thereof; an excipient, such as, for example, dicalcium phosphate, mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate or combinations thereof; a disintegrating agent, such as, for example, corn starch, potato starch, alginic acid or combinations thereof; a lubricant, such as, for example, magnesium stearate; a sweetening agent, such as, for example, sucrose, lactose, saccharin or combinations thereof; a flavoring agent, such as, for example peppermint, oil of wintergreen, cherry flavoring, orange flavoring, etc.
  • a binder such as, for example, gum tragacanth, acacia, cornstarch, gelatin or combinations thereof
  • an excipient such as, for
  • the dosage unit form When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar, or both. When the dosage form is a capsule, it may contain, in addition to materials of the above type, carriers such as a liquid carrier. Gelatin capsules, tablets, or pills may be enterically coated. Enteric coatings prevent denaturation of the composition in the stomach or upper bowel where the pH is acidic. See, e.g., U.S. Pat. No. 5,629,001.
  • the basic pH therein dissolves the coating and permits the composition to be released and absorbed by specialized cells, e.g., epithelial enterocytes and Peyer's patch M cells.
  • a syrup of elixir may contain the active compound sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor.
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compounds may be incorporated into sustained-release preparation and formulations.
  • compositions of the present invention may alternatively be incorporated with one or more excipients in the form of a mouthwash, dentifrice, buccal tablet, oral spray, or sublingual orally- administered formulation.
  • a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution).
  • the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically- effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
  • the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
  • suppositories are solid dosage forms of various weights and shapes, usually medicated, for insertion into the rectum. After insertion, suppositories soften, melt or dissolve in the cavity fluids.
  • traditional carriers may include, for example, polyalkylene glycols, triglycerides or combinations thereof.
  • suppositories may be formed from mixtures containing, for example, the active ingredient in the range of about 0.5% to about 10%, and preferably about 1% to about 2%.
  • the Tel mutant composition of the invention may be administered via a parenteral route.
  • parenteral includes routes that bypass the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered for example, but not limited to intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally (see U.S. Pat. Nos. 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363, each specifically incorporated herein by reference in its entirety).
  • Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Patent 5,466,468, specifically incorporated herein by reference in its entirety).
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (i.e., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • a coating such as lecithin
  • surfactants for example
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • aqueous solutions for parenteral administration in an aqueous solution
  • the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in isotonic NaCl solution and either added hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580).
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • a powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
  • the Tel mutant compound may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or inhalation.
  • Pharmaceutical compositions for topical administration may include the active compound formulated for a medicated application such as an ointment, paste, cream or powder. Ointments include all oleaginous, adsorption, emulsion and water-solubly based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only.
  • Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin.
  • Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram.
  • Possible bases for compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base.
  • Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the active ingredient and provide for a homogenous mixture.
  • Transdermal administration of the present invention may also comprise the use of a "patch".
  • the patch may supply one or more active substances at a predetermined rate and in a continuous manner over a fixed period of time.
  • the pharmaceutical compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described e.g., in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in its entirety).
  • the delivery of drugs using intranasal microparticle resins Takenaga et ah, 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725, 871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts.
  • transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety).
  • aerosol refers to a colloidal system of finely divided solid of liquid particles dispersed in a liquefied or pressurized gas propellant.
  • the typical aerosol of the present invention for inhalation will consist of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent.
  • Suitable propellants include hydrocarbons and hydrocarbon ethers.
  • Suitable containers will vary according to the pressure requirements of the propellant.
  • Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms. VII. Kits of the Invention
  • compositions described herein may be comprised in a kit.
  • one or more Tel mutants are comprised in a kit and are housed in suitable container means.
  • kits may comprise a suitably aliquoted Tel mutant of the present invention.
  • the components of the kits may be packaged either in aqueous media or in lyophilized form, for example.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there are more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
  • the kits of the present invention also will typically include a means for containing the Tel mutant and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.
  • the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.
  • the Tel mutants may also be formulated into a syringeable composition, in which case, the container means may itself be a syringe, pipette, and/or other such like apparatus, from which the formulation may be applied to an infected area of the body, injected into an individual, and/or even applied to and/or mixed with the other components of the kit.
  • the components of the kit may be provided as dried powder(s).
  • the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
  • kits of the present invention will also typically include a means for containing the vials in close confinement for commercial sale, such as, e.g., injection and/or blow-molded plastic containers into which the desired vials are retained.
  • a means for containing the vials in close confinement for commercial sale such as, e.g., injection and/or blow-molded plastic containers into which the desired vials are retained.
  • kits of the invention may also comprise, and/or be packaged with, an instrument for assisting with the injection/administration and/or placement of the ultimate therapeutic composition within the body of an animal.
  • an instrument may be a syringe, pipette, forceps, and/or any such medically approved delivery vehicle.
  • the human mesothelioma cell line 145 and the human lung cancer cell line HI 299 were maintained in RPMI 1640 medium (Thermo Scientific Waltham, MA) supplemented with 10% Fetal Bovine Serum (Invitrogen, Carlsbad, CA).
  • A549 human lung adenocarcinoma cells were maintained in F-12K medium (ATCC, Manassas, VA).
  • Tel cDNA was purchased from Origene Corp (Rockville, MD). All Tel mutants were generated through standard DNA mutagenesis.
  • Anti-Bcl-xl antibodies and all anti-MAP kinase antibodies were purchased from Cell Signaling Technology (Beverly, MA). All antibodies used to detect ETS proteins were obtained from Santa Cruz Biotechnology (Santa Cruz, CA).
  • Anti-Actin monoclonal antibody was purchased from Sigma- Aldrich (St. Louis, MO).
  • Chromatin immunoprecipitation assay 145 and A549 cells were plated at a density of 5 X 10 5 per well in a 6 well plate and transfected with Tel and Tel S213A expressing plasmids using Lipofectamine 2000. The cells were serum- starved with or without a lOOng/ml HGF or EGF simulation for 20 minutes. The chromatin was crosslinked by treating the transfected cells with 1% formaldehyde for 15 minutes, sonicated to an average length of 1000 bp and immunoprecipitated with Tel antibody (Santa Cruz) at 4oC overnight.
  • the immunoprecipitated chromatin was then reverse crosslinked and PCR amplified with primers specific for BCL-XL promoter (forward 5 '-GCCTAAGGCGGATTTGA ATGTAG-3 ' (SEQ ID NO: l); reverse 5 '-GAAGGGAGAGAAAGAGATTC AGGAA-3 '(SEQ ID NO:2)).
  • Luciferase assay A549, H1299 and 145 cells were transfected in triplicate in a 24 well plate using Lipofectamine 2000 with 200 ng of Tel or Tel S213A cDNA and pXL plasmid generated by cloning the human Bcl-xl promoter into the pGL2 vector (Promega, Madison, WI). A plasmid expressing ⁇ -Gal (25 ng) was used as an internal control for transfection efficiency. The transfected cells were cultured in serum containing media for 24 hours and then subjected to serum starvation and stimulation by HGF/EGF for 20 minutes. Luciferase and ⁇ - Galactosidase activities were measured and the luciferase values were normalized to the ⁇ - Galactosidase control.
  • XTT assays A549 and I 45 cells were seeded in 96 well plates at 5xl0 3 cells per well and then transfected with various amounts of Tel and Tel S213A using Lipofectamine 2000. The transfected cells were cultured for a further 48 hours and assayed following the standard protocol provided with an XTT kit (Roche, Indianapolis, IN).
  • Clonogenic assays were performed to determine the cytotoxic effects of Tel and the Tel S213A. I 45 and A549 cells were transfected with Tel and Tel S213A at various concentrations by electroporation using nucleofector solution L (Lonza Group, Basel, Switzerland). lxlO 4 cells were then plated per 10 mm dish in triplicate for each concentration and cultured for 2 weeks. The cells were then fixed using 2.5% formaldehyde and stained with 1% crystal violet.
  • Western blotting was performed using a standard protocol. Briefly, I 45, H1299 and A549 cells were transfected with Tel, Tel S213A and Tel S213A/K11R cDNA and cultured for 48 hours. The cells were then lysed and equal amounts of total protein were resolved on 4-20 % Tris-glycine gels. The resolved proteins were then transferred onto a nitrocellulose membrane and the membranes were incubated with antibodies raised against Bcl2 family proteins. The ECL system was used to visualize the signals.
  • Tissue arrays (US Biomax, Rockville, MD) were deparaffinized in xylene substitute and rehydrated in PBS. Antigen retrieval was performed with citrate buffer (pH 6.5) for 20 minutes at 99°C, followed by the block of endogenous peroxidase activity. Sections were incubated with blocking serum in PBS containing 5% bovine serum albumin, followed by incubation with rabbit anti-human Bcl-xl polyclonal antibody or with rabbit anti-phosphorylated human EGFR polyclonal antibody for 24 hours, followed by incubation with a biotinylated goat secondary anti-rabbit antibody (1:200 dilution, Vector Laboratories, Burlingame, CA).
  • Immunoreactive signals were detected using a streptavidin- biotin-peroxidase complex (Vector Laboratories), according to the manufacturer's recommended procedures. All of the slides were counterstained with hematoxylin (Sigma- Aldrich).
  • MAPKs mitogen- activated protein kinases
  • Akt Akt
  • GSK-3 Akt
  • p70 S6 kinase Proteome ProfilerTM Human Phospho-MAPK Arrays.
  • FIG. 1A The array signals, shown in FIG. 1A, indicated that kinase activities from growth factor stimulated cells were much stronger than those from serum-starved 145 cells. There were no significant differences found between the c-Met and EGFR groups. The activation of MAPKs was further confirmed by western blotting.
  • ETS-2, PU. l and Tel proteins were analyzed in 145 cells under conditions of serum starvation or HGF stimulation by immunoprecipitation and western blot analysis. Whereas the levels of total ETS proteins were observed to be equivalent in the cells, the levels of phosphorylated ETS-2, PU.l and Tel were clearly elevated (FIG. IB). Similar results were observed in lung cancer cells upon EGF stimulation. The inventors then mapped the serine sites that were phosphorylated after growth factor exposure. FIG. 1C demonstrated that only S213A mutation prevented Tel being phosphorylated.
  • TEL PHOSPHORYLATION NEGATIVELY REGULATES THE TEL-BCL-XL PROMOTER INTERACTION UPON THE GROWTH FACTOR STIMULATION OF
  • Bcl-xl promoter construct XLp-Luc, Tel and Tel S213A cDNA expression vectors were co-transfected into A549 and I 45 cells.
  • Bcl-xl promoter activity is significantly decreased in serum-starved Tel transfected 145 cells in comparison with the I 45 cells under growth factor stimulation.
  • Tel S213A expression leads to decreased Bcl-xl expression under both growth factor stimulated and growth factor deprived conditions.
  • TEL S213A IS A MORE POTENT REPRESSOR OF BCL-XL EXPRESSION THAN
  • Bcl-2 family proteins were evaluated by western blotting following the transfection of cancer cells with Tel, Tel S213A and control expression vector. As shown in FIG. 4B, the expression of Bcl-xl was decreased below basal levels after Tel cDNA transfection. Bcl-xl expression was further reduced when Tel S213A was expressed in both A549 and 145 cells, whereas the Bcl-2 levels remained unchanged. Mcl-1 was found to be upregulated in these cells on a western blot suggesting that an apoptosis "defense mechanism" operates in these tumor cells (Cao et ah, 2007; Xu et ah, 2010).
  • TEL S213A IS MORE POTENT INDUCER OF APOPTOSIS THAN WILD TYPE TEL
  • the current data also indicate that Tel Ser213 phosphorylation is the key response to Bcl-xl transcriptional repression. Tel has multiple serine and threonine sites that can potentially be phosphorylated by ERK kinase (Irvin et ah, 2003). Although in certain embodiments Ser213 (not Ser257) is the only Tel site phosphorylated by ERK in the tested cell lines, Ser 257 has been reported to be the target of p38, which is an activator of different stress signals (Hanson et ah, 2008). Hence, the phosphorylation of Tel by both ERK and p38 is a unique property of this ETS protein. Both ERK and stress signaling pathways converge via Tel in the nucleus under certain circumstances and provide further stimulation of Bcl-xl transcription in cancer cells.
  • the identified phosphorylation sites on Tel reside within the linker inhibitory damper (LID) domain, outside its ETS DNA-binding domain.
  • LID linker inhibitory damper
  • CID amino acids 423-452
  • LID domain amino acids 124-3378
  • CID amino acids 423-452
  • LID domain serves as negative regulator of CID (Green et ah, 2010). Decreased Tel binding to DNA due to the phosphorylation of S213 indicates that LID, once phosphorylated, in certain embodiments of the invention loses its inhibitory interaction with the CID, which would negatively regulate Tel- promoter binding.
  • Tel is also modified by sumoylation at Kl l and K99 (Wood et ah, 2003; Chakrabarti et ah, 2000).
  • the present studies indicate that the double mutant Tel K11R/S213A is a stronger repressor of Bcl-xl transcription than either single mutation alone.
  • both phosphorylation and sumoylation are required for Tel export, the order in which Tel is phosphorylated and sumoylated is unclear, as is whether these two types of modification function cooperatively or independently.
  • ETS members such as NET and YAN
  • phosphorylation and sumoylation generally work synergistically to mediate the downregulatory nuclear export of the ETS transcriptional repressors (Tootle and Rebay, 2005).
  • Dazzi H Hasleton PS, Thatcher N, et al. Malignant pleural mesothelioma and epidermal growth factor receptor (EGF-R). Relationship of EGF-R with histology and survival using fixed paraffin embedded tissue and the F4, monoclonal antibody. Br J Cancer 1990; 61: 924-926.
  • Foulds CE Nelson ML, Blaszczak AG, et al. Ras/mitogen- activated protein kinase signaling activates Ets-1 and Ets-2 by CBP/p300 recruitment. Mol Cell Biol 2004; 24: 10954-10964.
  • Wood LD Irvin BJ, Nucifora G, et al. Small ubiquitin-like modifier conjugation regulates nuclear export of TEL, a putative tumor suppressor. Proc Natl Acad Sci U S A 2003; 100: 3257-3262.
  • Xu H Krystal GW. Actinomycin D decreases Mcl-1 expression and acts synergistically with ABT-737 against small cell lung cancer cell lines. Clin Cancer Res; 16: 4392-4400.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Toxicology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention generally concerns methods and compositions for treating an individual with cancer utilizing polypeptide mutants of TEL. In particular embodiments, TEL polypeptides or peptides are employed having mutations at one or more of phosphorylation, acetylation, glycosylation, ubiquitination, or sumoylation sites, for example.

Description

TARGETED CANCER THERAPY USING ETS TRANSCRIPTIONAL INHIBITORS
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0001] This application claims priority to U.S. Provisional Patent Application Serial No. 61/443,918, filed February 17, 2011, which is incorporated by reference herein in its entirety.
[0002] This invention was made with government support under Grant No. R01- CA098545-01A1 awarded by the National Institutes of Health. The government has certain rights in the invention.
TECHNICAL FIELD
[0003] The present invention generally concerns the fields of cell biology, molecular biology, and medicine, including cancer medicine. In particular aspects, the present invention concerns mutants of an E-twenty six (ETS) transcriptional repressor protein for cancer therapy.
BACKGROUND OF THE INVENTION
[0004] Multiple receptor tyrosine kinases are overexpressed in a wide variety of solid tumors including malignant mesothelioma (MM) and non-small cell lung cancer (Dazzi et al, 1990; Herbst and Shin, 2002; Sridhar et al, 2003; Rogers et al, 2009; Mukohara et al, 2005). EGFR, c-Met (HGFR) and IGF1R may cooperate to regulate tumor growth and survival. The inhibition of one receptor can be compensated by activation of the others, because there is a degree of functional redundancy between these receptors (Bachleitner-Hofmann et al, 2008; Guo et al., 2008; Whittaker et al., 2010). The complex crosstalk between these pathways could therefore significantly reduce the efficacy of single receptor targeted therapies. Therefore, elucidating and targeting common downstream effectors is of some importance.
[0005] The association between Bcl-xl and these receptors has been well established (Dai et al., 2003; Jost et al., 2001; Sekharam et al., 2003). As a downstream factor, the elevation of Bcl-xl indicates a role for these activated receptor tyrosine kinases in tumor progression and resistance to chemotherapies. Multiple lines of evidence suggest that receptor tyrosine kinases upregulate Bcl-xl expression through ETS family transcription factors (Lee et al, 2009; Sevilla et al., 2001).
[0006] The ETS family consists of more than 30 members, which include both transcription factors and repressors (Silverman et al., 2002; Wei et al., 2010). Most ETS proteins are nuclear targets of diverse signaling pathways, such as the mitogen- activated protein (MAP) kinase signaling pathway, and undergo post-translational modifications including phosphorylation, glycosylation, acetylation, ubiquitination, and sumoylation (Cao et al., 2009). These modifications have a profound impact on the activity and subcellular localization of all ETS proteins. Among these proteins, ETS-1, 2 and PU. l, which are known positive transcriptional regulators of Bcl-xl, are imported to the nucleus following MAPK phosphorylation, and can be exported by an active transport pathway (Sevilla et al., 2001; Sevilla et al., 1999; Nelson et al., 2010). Tel is a transcriptional repressor of Bcl-xl, and is also negatively regulated by phosphorylation and sumoylation, by nuclear export, or as a result of localization to Tel-bodies (Roukens et al., 2008; Maki et al., 2004). Tel interactions with relevant corepressors, mSin3A, N-CoR and HDAC3, are believed to mediate the transcriptional repression of multiple target genes, including Bcl-xl (Wang et al., 2001).
BRIEF SUMMARY OF THE INVENTION
[0007] In certain aspects, the present invention concerns methods and/or compositions related to E-twenty six (ETS) protein inhibitors for cancer therapy and/or prevention in a mammal. Tel, a transcription repressor member in the ETS family, has been identified as a tumor suppressor. Delivery of Tel into tumor cells leads to tumor cell death, growth arrest and inhibition of invasion. Tel can serve as a pan ETS inhibitor, because it competes with other members of ETS family proteins on similar ETS binding sites. In particular embodiments of the invention, there is a mutated ETS transcriptional repressor protein that is capable of binding a particular DNA promoter sequence and that abolishes other ETS transcription factors' activities.
[0008] Most ETS proteins are nuclear targets of diverse signaling pathways, such as the mitogen-activated protein (MAP) kinase signaling pathway, and undergo post-translational modifications, including phosphorylation, glycosylation, acetylation, ubiquitination, and sumoylation, for example. Tel is a transcriptional repressor that is negatively regulated by phosphorylation and sumoylation, by nuclear export, and/or as a result of localization to Tel- bodies, for example. Tel interactions with relevant co-repressors, including mSin3A, N-CoR and HDAC3, mediate the transcriptional repression of multiple target genes, including Bcl-xl, in certain aspects of the invention.
[0009] In certain cases of the present invention, there are Tel repressors that have altered post-transcriptional modifications compared to wild-type Tel. The modifications can occur at one or more of the sites for ubiquitination, sumoylation, phosphorylation, and/or auto- inhibition, in certain embodiments. In particular aspects of the invention, the transcriptional repressor activity of the mutant Tel is greater than that for wild-type Tel. Thus, in certain embodiments the mutants have enhanced Tel repressive function compared to wild-type Tel.
[0010] In particular embodiments of the invention, Tel is negatively regulated at the post-transcriptional level, such as through ubiquitination, sumoylation, phosphorylation, and/or auto-inhibition. For example, Tel can be modified by sumoylation at Kl l and K99 and can be modified by phosphorylation at S213. The exemplary results provided herein indicate that the phosphorylation of Tel at Ser213 blocks the Tel's repression on Bcl-xl transcription. This negative regulation of Tel function following receptor activation contributes to the overexpression of the Bcl-xl protein in cancer cells.
[0011] In some aspects of the invention, there are Tel proteins with one or more mutations. In certain cases, the one or more mutations provide enhanced repressor activity, such as compared to wild type. In particular aspects, a Tel mutant having two or more mutations has greater repressor activity compared to the single mutant activity. In the present invention, the exemplary double mutant Tel K11R/S213A is a stronger repressor of Bcl-xl transcription (for example) than either single mutation alone. Whereas both phosphorylation and sumoylation are useful for Tel export, for example, in certain embodiments these two types of modification function cooperatively or independently. As with certain other ETS members, such as NET and YAN, in certain aspects of the invention the phosphorylation and sumoylation work synergistically, for example to mediate the downregulatory nuclear export of the ETS transcriptional repressors.
[0012] In certain embodiments of the invention, the mutant Tel polypeptide is hypophosphorylated compared to wild-type Tel. Exemplary phosphorylation sites on Tel (S213 and S257, for example) reside within the linker inhibitory damper (LID) domain outside of its ETS DNA-binding domain. Recent study has indicated that the autoregulation of Tel-promoter interaction involves two domains, the C-terminal inhibitory domain (CID) and LID domains (Green et al., 2010). CID (amino acids 423-452) negatively interferes with the Tel DNA binding domain. LID domain (amino acids 124-338) serves as negative regulator of the CID. Decreased Tel binding to DNA because of the phosphorylation of S213 indicates that in certain embodiments of the invention LID, once phosphorylated, loses its inhibitory interaction with the CID, which would then negatively regulate Tel-promoter binding.
[0013] The Tel polypeptide may have mutations of any suitable kind, but in specific embodiments the polypeptide retains transcriptional repressor activity, including activity that is greater than wild-type when measured by identical or similar assays. The mutation may be a point mutation, inversion, or deletion, for example. The mutation may alter the ability to be subject to one or more post-translational modifications. In an exemplary case, the amino acid that may be mutated such that phosphorylation cannot occur includes serine, tyrosine, or threonine, and one or more of these amino acids may be changed to another amino acid, particularly one that is not capable of being phosphorylated. In the context of a polynucleotide that encodes a Tel polypeptide, the mutation may change the respective encoded amino acid, but in some cases there may be mutations that do not change the respective encoded amino acid.
[0014] Tel mutants encompassed by the invention also include those that lack part of the protein. For example, the mutant may lack the N-terminal and/or C-terminal. In some embodiments, the protein lacks part or all of the inhibitory CID domain. In some cases, the protein is modified such that it has greater protein solubility than wild type. In specific embodiments, the Tel protein has single, double, or triple or more modifications that render greater solubility for the protein. In one certain case, the Tel V112E mutant has increased protein solubility. In some embodiments, a protein transduction domain (such as the TAT peptide sequence or Antennapedia homeodomain, for example), for example, is present on Tel to enable the polypeptide to penetrate into human cells. In some cases the protein transduction domain is present on the N-terminus of the Tel mutant, whereas in other cases the protein transduction domain is present on the C-terminus of the Tel mutant.
[0015] In particular embodiments of the invention, the methods and compositions of the invention are employed for cancer treatment and/or prevention, wherein the cancer is of any kind of cancer. In some aspects, however, the cancer is mesothelioma, thoracic cancer (including lung cancer, such as non- small cell lung carcinoma, for example), breast, colon, pancreas, prostate, skin, liver, kidney, bone, spleen, cervix, ovary, testes, bladder, gall bladder, or brain cancer. The individual to be treated with the invention is a mammal, including a human, dog, cat, horse, pig, sheep, or goat, for example.
[0016] In one embodiment of the invention, there is an isolated TEL polypeptide having one or both of the following: a) an amino acid substitution at one or more sites corresponding to SEQ ID NO:3, said sites selected from the group consisting of a serine corresponding to position 213, a serine corresponding to position 257, a lysine corresponding to position 11, a lysine corresponding to position 99, and a valine corresponding to position 112; and/or b) a deletion in the polypeptide corresponding to SEQ ID NO:3, said deletion occurring in one or more of a protein transduction domain, a TAT domain, a SAM domain, a LID domain, an ETS domain, or a CID domain, wherein the polypeptide has transcriptional repressor activity. In a specific embodiment, the polypeptide has increased transcriptional repressor activity over wild- type TEL polypeptide. In some specific embodiments, the polypeptide is further defined as having an amino acid substitution at the serine corresponding to position 213, at the serine corresponding to position 257, at the lysine corresponding to position 11, at the lysine corresponding to position 99, and/or at the lysine corresponding to position 99. In specific embodiments, the substitution at the serine corresponding to position 213 is alanine, the substitution at the serine corresponding to position 257 is alanine, the substitution at the lysine corresponding to position 11 is arginine, and/or the substitution at the lysine corresponding to position 99 is arginine. In certain aspects, the polypeptide further comprises a protein transduction domain.
[0017] In some embodiments of the invention, there is an isolated TEL polypeptide having a mutation at a phosphorylation site, a ubiquitination site, a sumoylation site, a glycosylation site, an acetylation site, or a combination thereof, wherein the polypeptide has transcriptional repressor activity. In specific cases, the polypeptide has increased transcriptional repressor activity over wild-type TEL polypeptide. In aspects of the invention, the mutation occurs within a C-terminal inhibitory domain (CID), a linker inhibitory damper domain (LID), ETS DNA-binding domain (ETS), or a sterile alpha motif (SAM) domain. The polypeptide may be further defined as comprising (corresponding to positions in SEQ ID NO:3) a mutation at serine 213, comprising a mutation at serine 257, comprising a mutation at valine 112, and/or having a mutation at lysine 11. The polypeptide may further comprise deletion of part or all of the CID domain and/or the polypeptide may further comprise a protein transduction domain, such as from HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat- (35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1, or Pep-2, for example.
[0018] In embodiments of the invention, there is an isolated recombinant amino acid sequence of formula: R0 - Rl - R2 - R3 - R4 - R5 -R6, wherein R0 is H or an amino acid sequence comprising 1 to 500 consecutive amino acids; Rl is an amino acid sequence comprising 1 to 11 consecutive amino acid residues of a protein transduction domain; R2 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a SAM domain; R3 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a LID domain; R4 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of an ETS domain; R5 is a bond or an amino acid sequence comprising 10 or more consecutive amino acid residues of a CID domain; and R6 is H or an amino acid sequence comprising 1 to 500 consecutive amino acids, provided that R2, R3, R4, and R5 are not all bonds, provided that the isolated recombinant amino acid sequence is not SEQ ID NO: 3, and provided that the amino acid sequence has transcriptional repressor activity.
[0019] In embodiments of the invention wherein there is an isolated recombinant amino acid sequence of formula: R0 - Rl - R2 - R3 - R4 - R5 -R6, in some aspects R2, R3, R4, and R5 are not bonds. In specific cases, Rl is selected from the group consisting of SEQ ID NO:5-23; R2 comprises SEQ ID NO:24; R3 comprises SEQ ID NO:25; R4 comprises SEQ ID NO:26; and/or R5 comprises SEQ ID NO:27.
[0020] In embodiments of the invention wherein there is an isolated recombinant amino acid sequence of formula: R0 - Rl - R2 - R3 - R4 - R5 -R6, the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:28. In particular aspects, R2, R3, and R4 are not bonds but R5 is a bond. In specific cases, Rl comprises a sequence selected from the group consisting of SEQ ID NO:5-23; R2 comprises SEQ ID NO:24; R3 comprises SEQ ID NO:25; R4 comprises SEQ ID NO:26. In specific embodiments, the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:29. In certain aspects, R2 is a bond and R3, R4, and R5 are not bonds. In some aspects, the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:30. In particular cases, R2 and R5 are bonds and R3 and R4 are not bonds. In some embodiments, R2 is a bond and R3, R4 and R5 are not bonds. In certain embodiments, the amino acid sequence of the polypeptide of the invention comprises SEQ ID NO:32. In some cases,
[0021] In embodiments of the invention wherein there is an isolated recombinant amino acid sequence of formula: R0 - Rl - R2 - R3 - R4 - R5 -R6, Rl comprises a TAT sequence (SEQ ID NO:6); R2 comprises SEQ ID NO:35; R2 comprises SEQ ID NO:36; R2 comprises SEQ ID NO:37; R3 comprises SEQ ID NO:38; R3 comprises SEQ ID NO:39; and/or R3 comprises SEQ ID NO:40.
[0022] In some embodiments of the invention, wild-type Tel without at least part of the CID domain is employed. In certain aspects, there is a Tel lacking the CID domain but the remainder of the Tel protein variant is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the corresponding region in the wild-type Tel protein. In certain aspects, there is a Tel lacking the CID domain but the remainder of the Tel protein variant has one, two, three, four, five, or more amino acid substitutions or deletions. In certain aspects, there is a Tel lacking the CID domain but the remainder of the Tel protein variant has no more than one, two, three, four, five, or more amino acid substitutions or deletions. In certain aspects, there is a Tel lacking the CID domain but the remainder of the Tel protein variant has at least one, two, three, four, five, or more amino acid substitutions or deletions.
[0023] In some embodiments of the invention, all of the CID domain is absent from the Tel protein, whereas in certain cases part of the CID domain is absent, such as 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid(s) from the CID domain are absent.
[0024] In specific embodiments, a Tel protein comprising TAT, the V112E mutation, and/or lacking part or all of the CID domain is employed. An exemplary TAT-Tel wild-type with V112E but without CID is provided in SEQ ID NO:43 and may be employed in the invention, in certain embodiments.
[0025] In one embodiment of the invention, there is a recombinant TEL polypeptide comprising an ETS DNA-binding domain (ETS) and a) a sterile alpha motif (SAM) domain having a deletion or an amino acid substitution at a lysine or a valine; and/or b) a linker inhibitory damper domain (LID) domain having a deletion or an amino acid substitution at one or more serines; and/or c) an amino acid substitution at a lysine corresponding to position 11 in SEQ ID NO:3. In a specific embodiment, the polypeptide further comprises a protein transduction domain, a nuclear localization signal, and/or a C-terminal inhibitory domain (CID) domain. In particular aspects, the polypeptide has increased transcriptional repressor activity over wild-type TEL polypeptide. The polypeptide may be further defined as having an amino acid substitution at the serine corresponding to position 213 in SEQ ID NO:3, as having an amino acid substitution at the serine corresponding to position 257 in SEQ ID NO:3, as having an amino acid substitution at the lysine corresponding to position 99 in SEQ ID NO:3, and/or as having an amino acid substitution at the valine corresponding to position 112 in SEQ ID NO:3.
[0026] Exemplary protein transduction domains utilized in the invention include, for example, HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat- (35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1, or Pep-2.
[0027] In an embodiment of the invention, there is an isolated recombinant TEL polypeptide comprising: a) an amino acid substitution at one or more sites in the polypeptide corresponding to SEQ ID NO:3, said sites selected from the group consisting of a serine corresponding to position 213, a serine corresponding to position 257, a lysine corresponding to position 11, a lysine corresponding to position 99, and a valine corresponding to position 112; and/or b) a deletion in the polypeptide corresponding to SEQ ID NO:3, said deletion occurring in one or more of a protein transduction domain, a TAT domain, a SAM domain, a LID domain, an ETS domain, or a CID domain, wherein the polypeptide has transcriptional repressor activity.
[0028] In some embodiments of the invention, there is a method of treating an individual for cancer, comprising the step of administering to the individual a therapeutically effective amount of a polypeptide of the invention. In certain aspects, the individual has a thoracic malignancy. In some cases, the individual has mesothelioma. In some embodiments, the method comprises an additional therapy for cancer.
[0029] Kits that comprise the polypeptide of the invention are encompassed, said polypeptide housed in a suitable container.
[0030] The foregoing has outlined rather broadly the features and technical advantages of the present invention in order that the detailed description of the invention that follows may be better understood. Additional features and advantages of the invention will be described hereinafter which form the subject of the claims of the invention. It should be appreciated by those skilled in the art that the conception and specific embodiment disclosed may be readily utilized as a basis for modifying or designing other structures for carrying out the same purposes of the present invention. It should also be realized by those skilled in the art that such equivalent constructions do not depart from the spirit and scope of the invention as set forth in the appended claims. The novel features which are believed to be characteristic of the invention, both as to its organization and method of operation, together with further objects and advantages will be better understood from the following description when considered in connection with the accompanying figures. It is to be expressly understood, however, that each of the figures is provided for the purpose of illustration and description only and is not intended as a definition of the limits of the present invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0031] For a more complete understanding of the present invention, reference is now made to the following descriptions taken in conjunction with the accompanying drawings.
[0032] FIG. 1A provides that stimulation of cells by HGF and EGF causes identical phosphorylation events by multiple kinases as measured using the proteome profile human phosphor-MAPK array. FIG IB shows that activated Map kinase phosphorylates ETS proteins including Tel. The mesothelioma cell line, 145, was grown under conditions of serum starvation or serum starvation plus 20 minutes of HGF (100 ng/ml) stimulation. The endogenous ETS-2, PU.l and Tel proteins were immunoprecipitated from the cell lysates using the respective antibodies. The immunoprecipitates were then analyzed by western blot using phosphor-Ser/Thr antibodies. C: Tel is phosphorylated on Serine 213 after HGF stimulation. The Tel serine mutant S213A, S257A and wild- type Tel were transfected into 145 cells which were then serum starved or HGF stimulated. Tel was immunoprecipitated from cell lysates and its phosphorylation status was determined using anti phosphor-Ser/Thr antibodies.
[0033] FIG. 2 shows that CHIP (IP) assay evaluation of the impact of Tel phosphorylation on its binding to the Bcl-xl promoter. 145 and A549 cells transfected with wild- type or Tel S213A mutants were serum-starved for 24 hours and then subjected to treatment with or without HGF or EGF for 20 minutes. Cross-linked Tel-DNA complexes were immunoprecipitated using a Tel antibody and Bcl-xl promoter fragments were PCR amplified. [0034] FIG. 3 A demonstrates that the Tel phosphorylation site mutant S213A is not exported from the nucleus after EGF stimulation. 145 cells were transfected with Tel or Tel S213A expression vectors and 24 hours after serum starvation, these transfected cells were exposed to HGF stimulation for 20 minutes. Acetone-fixed cells were then incubated with Tel antibodies followed by a fluorescein isothiocyanate (FITC)-labeled secondary antibody. Fluorescein signals were visualized under a Nikon 80i fluorescent microscope. 4,6-Diamidino-2- phenylindole (DAPI) was used as a counterstain for nuclear DNA. FIG. 3B: Tel S213A achieves a stronger repression of the Bcl-xl promoter in the presence of growth factor activation. A549 and 145 cells seeded in 24-well plates were cotransfected with Bcl-xl promoter pXL-luc and either Tel S213A or Tel vectors along with p-CMV-P-galactosidase. Twenty-four hours after transfection, the cells were lysed, and luciferase and β-galactosidase activities were measured with a luminometer. The luciferase activities were normalized to those of β-galactosidase, and the data shown are the average of triplicate determinations. This experiment was repeated twice.
[0035] FIG. 4A shows that growth factor stimulation of cells fails to decrease the repression of Bcl-xl by Tel S213A. 145 mesothelioma cells were transfected with GFP, Tel wild type or Tel S213A and grown under conditions of serum starvation or serum starvation plus HGF stimulation (100 ng/ml) for 48 hours. The Bcl-xl expression levels in these cells were then determined by western blotting. FIG. 4B. To determine the gene targets of Tel repression, both A549 and 145 cells were transfected with GFP, Tel wild type or Tel S213A. Forty-eight hours after transfection the Blc-2 target protein levels were detected by western blotting.
[0036] FIG. 5A demonstrates measurement of the short-term tumor growth suppression of Tel S213A by cell proliferation assay. A549 and 145 cells were seeded in 96 well plates and transfected with 0.2 μg of Tel or Tel S213A cDNA, and cultured for 48 hours. Cell growth inhibition was measured by the XTT assay. The data shown are the average of four independent determinations. FIG. 5B. Demonstration of long-term tumor growth suppression by Tel S213A in a colony formation assay. A549 and 145 cells were transfected with various doses of Tel or Tel S213A cDNA and plated in 10 cm cell culture plates at 104 cells per plate. After 20 days, the cells were fixed and stained and the colonies were counted, the results demonstrate that TelS213A is a stronger repressor of Bcl-xl than wild type Tel. The data are the average of triplicate determinations. FIG 5C. Tel S213A induces a stronger apoptotic response in cancer cells. A549 cells were transfected with pGL-2, Tel wild type and Tel S213A, collected and stained with FITC-labeled Annexin V and PL Annexin V-positive populations were quantified by flow cytometry. The data shown are the results of triplicate determinations.
[0037] FIG. 6A illustrates the impact of multiple-site mutations of Tel on Bcl-xl promoter repression. A549 cells were seeded in 96-well plates and transfected with a Bcl-xl promoter reporter, pXL-luc, and co-transfected with Tel wild-type, Tel K11R, Tel S213A, Tel Kl lR, Tel K11R/S213A and a p-CMV-P-galactosidase transfection control vector. At 24 hours after transfection, the cells were lysed and both the luciferase and β-galactosidase activities were measured using a luminometer. The luciferase activity was normalized using the β-galactosidase activity levels, and the data shown are the averages of triplicate determinations. This promoter analysis was repeated three times. FIG. 6B. Comparison of Bcl-xl protein levels following transfection with the indicated Tel mutants. A549 cells were seeded into 6 well plates (106 cells per well) and were transfected with Tel, Tel mutants and GFP expression vectors. Forty-eight hours after transfection, the Bcl-xl protein levels were determined by western blot.
[0038] FIG. 7 shows analysis of EGFR phosphorylation and Bcl-xl using human non-small cell lung carcinoma and mesothelioma tissue arrays. Correlations between the Bcl-xl levels and phosphorylated EGFR levels were demonstrated by Chi-square analysis (P < 0.01).
[0039] FIG. 8 provides an exemplary schematic representation of the negative regulation of Tel by phosphorylation.
[0040] FIG. 9 illustrates exemplary modifications of wild-type TEL.
[0041] FIG. 10 provides the sequence of Tel, SEQ ID NO:3, which illustrates different exemplary domains in the Tel protein. Although the ETS domain comprises amino acids 338-422, the figure marks it as 331-422 merely to illustrate a nuclear localization signal.
[0042] FIG. 11 illustrates one embodiment by which phosphorylations at S213/S257 decrease Tel's repression on Bcl-xl transcription and increase Bcl-xl transcription.
[0043] FIG. 12 shows an embodiment of another mechanism by which LID can repress CID.
[0044] FIG. 13 illustrates an embodiment wherein there is synergism of sumoylation and phosphorylation on negative regulation of Tel function. DETAILED DESCRIPTION OF THE INVENTION
[0045] As used herein, the use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." Some embodiments of the invention may consist of or consist essentially of one or more elements, method steps, and/or methods of the invention. It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein.
[0046] The term "effective amount" or "therapeutically effective amount" as used herein is defined as an amount of the agent that will decrease, reduce, inhibit or otherwise abrogate the growth of a neoplasm, induce apoptosis, inhibit angiogenesis of a neoplasm, inhibit metastasis, or induce cytotoxicity in a neoplasm. Thus, an effective amount is an amount sufficient to detectably and repeatedly ameliorate, reduce, minimize or limit the extent of the disease or its symptoms.
I. General Embodiments of the Invention
[0047] ETS transcriptional factors are molecular targets for selective cancer therapy, because they play roles in maintaining malignancy of cancer cells. It is difficult to develop small molecules that directly inhibit the function of ETS transcription factors, because these proteins bind to the same DNA sequence in humans, and the loss of function of one transcription factor due to a small molecular inhibitor, for example, can be compensated by other ETS transcription factors. This difficulty can be supported by the ineffectiveness of dominant negative mutant and siRNA/antisense approaches, for example.
[0048] The present invention provides novel ETS transcriptional inhibitors and encompasses Tel polypeptides that are modified compared to wild type but retain transcriptional repressor activity, including enhanced transcriptional repressor activity compared to wild type, for example. The Tel gene can be negatively regulated at the post-transcriptional level through ubiquitination, glycosylation, acetylation, sumoylation and phosphorylation, for example. The mutant may have a single mutation or more than one mutation. For example, the exemplary double-mutants TEL 11-213 enhance Tel's repressive function in tumor cells by abolishing one or more negative modifications, in certain embodiments. Furthermore, the present invention includes mutants with increased protein solubility, including the Tel V112E mutant having increased protein solubility. The invention also includes mutants with improved binding to promoter DNA compared to wild type Tel including, for example, a mutant with a deleted part or all CID domain that improves its binding to promoter DNA. The Tel ETS domain is a therapeutic protein, in certain embodiments. TAT peptide sequence is attached at its N terminus to enable protein to penetrate into human cells, in particular aspects of the invention.
[0049] As shown herein, the phosphorylation of Tel increases Bcl-xl transcription through tyrosine kinase receptors' phosphorylation and activation of ETS2, a transcriptional activator. In the present invention, stimulation by HGF or EGF activates an identical downstream signal transduction pathway and causes the activation of ERK, leading to the nuclear export of Tel. The exemplary phosphorylation site mutant TelS213A enhances Tel repression of Bcl-xl transcriptional activity. As provided herein, the nuclear localization of the Tel S213A mutant was detected under HGF/EGF exposure. A luciferase reporter assay revealed that the transient expression of Tel S213A results in further repression of Bcl-xl promoter activity. This repressive function is independent of growth factor stimulation, as determined by comparisons with wild type Tel cDNA-transfected cells. Furthermore, the results of a CHIP assay demonstrated that growth factor stimulation reduced the binding of wild-type Tel to the Bcl-xl promoter, whereas the binding of Tel S213A to this promoter was unaffected. Consequently, Tel S213A transfection results in a much stronger reduction of Bcl-xl protein expression in comparison with wild-type Tel cDNAs. Transfection of Tel S213A also leads to an increased inhibition of cell proliferation and promotes apoptosis. The sumoylation and phosphorylation of double mutant Tel K11R/S213A causes an even stronger repression of Bcl-xl. Finally, tissue array analyses indicate that the activation of EGFR correlates with Bcl-xl expression. Thus, in certain embodiments phosphorylation is a negative regulator of Tel function.
[0050] Thus, the present invention encompasses the impact of the activation of tyrosine kinase receptors upon transcriptional regulation, including Bcl-xl transcriptional regulation. Tel Ser213 phosphorylation is responsible for receptor kinase activation via the MEK-ERK pathway in cancer cells. At least the phosphorylation of Tel at Ser213 blocks the repression of Bcl-xl transcription by Tel. This negative regulation of Tel function following receptor activiation contributes to the overexpression of the prosurvival Bcl-xl protein in cancer cells.
[0051] Tel Protein and Exemplary Modifications Thereof [0052] In particular aspects of the invention, there are mutants of Tel protein utilized in methods and/or compositions for cancer therapy and/or prevention. The Tel protein mutants may be of any kind, but in particular aspects there are one, two, three, four, five, or more mutations compared to the wild-type Tel protein. In certain embodiments, the mutation(s) result in a functionally detectable difference compared to the wild-type protein, although in some cases the mutations result in a Tel protein having no detectable functional difference compared to the wild-type protein. In specific cases, the function that is characterized is transcriptional repressor activity. In particular aspects, the function that is characterized is as an inducer of apoptosis. In certain cases, both the transcriptional repressor activity and the apoptotic inducer functions are characterized. Measurement of transcriptional repressor activity may occur by any suitable means in the art, but in specific embodiments the measurement comprises cell transfection with the candidate mutant in a cell having detectable transcription of a reporter polynucleotide or other polynucleotide, followed by Western or northern. Transcription repression can be measured by luciferase assay using several genes' promoter constructs. In addition, electrophoresis mobility shift assay (EMSA) and Chromatin immunoprecipitation (ChIP) can be used to measure the interaction between transcription factor or repressor and DNA promoters. Real-time RT-PCR also can be adopted to measure the mRNAs levels of multiple ETV6/TEL regulated genes. Measurement of apoptosis inducer activity may occur by any suitable means in the art, but in specific embodiments the measurement comprises XTT assay (conversion of the water-soluble XTT assay (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5- carboxanilide) reagent to an orange formazan product by actively respiring cells); colony formation assay; Annexin V binding; and/or flow cytometry. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay can also be applied.
[0053] One or more amino acids of Tel may be mutated, in certain aspects of the invention, and in some cases, the amino acid is within a functional domain, for example related to a particular activity for the protein. For example, the mutation may be in the C-terminal inhibitory domain, CID, (amino acids 423-452 of the human wild-type protein), which interferes with the DNA binding domain. An additional example of a domain that might be inhibited includes the linker inhibitory damper domain, LID, (amino acids 124-338 of the human wild- type protein, and S213 and S257 are present therein). Other examples are the SAM domain (amino acids 41-123, and K99 is present therein), the ETS domain (amino acids 338-422), and the region for binding to CTBP1 (amino acids 423-427), which stabilizes Tel by blocking CID (Roukens et ah, 2010).
[0054] Exemplary sequences for wild- type Tel domains are as follows: SEQ ID NO:24 (SAM); SEQ ID NO:25 (LID); SEQ ID NO:26 (ETS); SEQ ID NO:27 (CID). Sequence for an exemplary construct having TAT, SAM, LID, ETS, and CID is in SEQ ID NO:28. Sequence for an exemplary construct having TAT, SAM, LID, and ETS is in SEQ ID NO:29. Sequence for an exemplary construct having TAT, LID, ETS, and CID is in SEQ ID NO:30. Sequence for an exemplary construct having TAT, LID, and ETS is in SEQ ID NO:31. Sequence for an exemplary construct having TAT, ETS, and CID is in SEQ ID NO:32. Sequence for an exemplary construct having TAT and ETS is in SEQ ID NO:33. Exemplary sequences for mutated Tel domains are as follows: SEQ ID NO:35 (SAM having K99R mutation); SEQ ID NO:36 (SAM having V112E mutation); SEQ ID NO:37 (SAM having K99R and V112E mutations); SEQ ID NO:38 (LID having S213A mutation); SEQ ID NO:39 (LID having S257A mutation); and SEQ ID NO:40 (LID having S213A and S257A mutations). Exemplary nuclear localization signals are provided in SEQ ID NO:34 (from Tel, located in the LID domain); SEQ ID NO:41 (ETS1 NLS signal); and SEQ ID NO:42 (SV40 large antigen NLS signal).
[0055] As used herein, an "amino acid" can include any naturally or non-naturally occurring amino acid that is phosphorylated, ubiquinated, sumoylated, glycosylated, acetylated and that any amino acid sequence of the present invention may include one or a combination of such amino acids. Specific naturally- occurring amino acids (or moieties of naturally occurring amino acids) that are capable of particular modifications are as follows: serine, threonine, and tyrosine can be phosphorylated; lysine can be ubiquinated, and lysine can be sumoylated. For glycosylation, N-linked glycans attach to a nitrogen of asparagine or arginine side-chains; O- linked glycans attach to the hydroxy oxygen of serine, threonine, tyrosine, hydroxylysine, or hydroxyproline side-chains, or to oxygens on lipids such as ceramide; phospho-glycans linked through the phosphate of a phospho-serine; and C-linked glycans are a rare form of glycosylation where a sugar is added to a carbon on a tryptophan side-chain. For acetylation, a lysine may be acetylated. [0056] FIG. 9 illustrates exemplary modifications of wild-type TEL. In specific embodiments, as shown in the figure, the polypeptide comprises a protein transduction domain, for example on the N-terminus or C-terminus.
[0057] FIG. 10 provides SEQ ID NO:3, which illustrates different exemplary domains in the Tel protein. The wild-type protein amino acid sequence is also at GenBank® Accession No. P41212 (SEQ ID NO:3), which is incorporated by reference herein in its entirety. A skilled artisan recognizes that other names for Tel include at least transcription factor ETV6, ETS translocation variant 6, and ETV6. A skilled artisan also recognizes that Tel2, which is a naturally occurring variant of Tell having a truncation on the N-terminus, may be employed in the invention, such as the modification occurring in a Tel2 polypeptide sequence (NP_057219 in GenBank® is the protein sequence, incorporated by reference herein; SEQ ID NO:4).
[0058] FIG. 11 illustrates one embodiment by which phosphorylations at S213/S257 decrease Tel's repression on Bcl-xl transcription and increase Bcl-xl transcription. Tel is the bcl-xl transcriptional repressor. CID, within the Tel protein, has an inhibitory interaction with Tel's ETS domain. The existence of CID can weaken Tel-Bcl-xl promoter binding and promote the Tel moving away from promoter. In one certain aspect of the invention, LID domain of Tel can negatively regulate CID's negative impact on Tel ETS domain, (as in, a negative of a negative is positive). Therefore Tel can repress Bcl-xl transcription at the promoter level. This mechanism is illustrated in FIG. 11 in the upper left picture. Once phosphorylated at S213/S257, LID will have a structure change and lose its control on CID, in certain embodiments of the invention. Therefore, CID can negatively regulate Tel ETS domain and promote Bcl-xl transcription (FIG. 11, bottom-right picture).
[0059] Thus, in certain embodiments of the invention, mutations at S213/S257 are useful to regulate Tel function. Blockage of phosphorylation of S213/S257 enhances Tel function, in particular aspects. Deletion of CID is also beneficial, in specific embodiments. K11/K99 are sumoylation sites, and in specific embodiments Kl l/99 sumoylation can promote Tel nuclear exportation, which abolishes Tel's repression. Therefore, in certain embodiments of the invevntion, sumoylation and phosphorylation of Tel are blocked to achieve enhanced Bcl-xl repression.
[0060] In the upper left picture of FIG. 12, the illustration of FIG. 11 is essentially repeated, but by comparison in the bottom right picture, there is an embodiment of another mechanism by which LID can repress CID. In specifc embodiments, LID directly binds to ETS and blocks CID-Tel ETS interaction.
[0061] FIG. 13 illustrates an embodiment wherein there is synergism of sumoylation and phosphorylation on negative regulation of Tel function. It also illustrates that Tel as well as ETS 1/2 have several co-factors to work together at DNA promoter level.
III. Proteinaceous Mutant Tel Compositions
[0062] In certain embodiments, the present invention concerns novel mutant Tel compositions comprising at least one proteinaceous molecule. As used herein, a "proteinaceous molecule," "proteinaceous composition," "proteinaceous compound," "proteinaceous chain" or "proteinaceous material" generally refers, but is not limited to, a protein of greater than about 400 amino acids or the full length endogenous sequence translated from a gene; a polypeptide of greater than about 350 amino acids; a polypeptide of greater than about 300 amino acids; a polypeptide of greater than about 250 amino acids; a polypeptide of greater than about 200 amino acids; a polypeptide of greater than about 150 amino acids; and/or a peptide of from about 3 to about 100 amino acids. All the "proteinaceous" terms described above may be used interchangeably herein.
[0063] In certain embodiments the size of the at least one proteinaceous molecule may comprise, but is not limited to, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59, about 60, about 61, about 62, about 63, about 64, about 65, about 66, about 67, about 68, about 69, about 70, about 71, about 72, about 73, about 74, about 75, about 76, about 77, about 78, about 79, about 80, about 81, about 82, about 83, about 84, about 85, about 86, about 87, about 88, about 89, about 90, about 91, about 92, about 93, about 94, about 95, about 96, about 97, about 98, about 99, about 100, about 110, about 120, about 130, about 140, about 150, about 160, about 170, about 180, about 190, about 200, about 210, about 220, about 230, about 240, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, or greater amino molecule residues of Tel, and any range derivable therein.
[0064] As used herein, an "amino molecule" refers to any amino acid, amino acid derivitive or amino acid mimic as would be known to one of ordinary skill in the art. In certain embodiments, the residues of the proteinaceous molecule are sequential, without any non-amino molecule interrupting the sequence of amino molecule residues. In other embodiments, the sequence may comprise one or more non-amino molecule moieties. In particular embodiments, the sequence of residues of the proteinaceous molecule may be interrupted by one or more non- amino molecule moieties.
[0065] In some embodiments of the invention, the Tel molecule is modified such that it comprises a protein transduction domain (PTD), which may also be referred to as a cell- penetrating peptide, for example. Exemplary cell penetrating peptides that are hydrophilic include the following:
[0066] Penetratin or Antennapedia PTD RQIKWFQNRRMKWKK (SEQ ID
NO:5)
[0067] TAT YGRKKRRQRRR (SEQ ID NO:6)
[0068] SynB 1 RGGRLSYSRRRFSTSTGR (SEQ ID NO:7)
[0069] SynB3 RRLSYSRRRF (SEQ ID NO: 8)
[0070] PTD-4 PIRRRKKLRRLK (SEQ ID NO:9)
[0071] PTD-5 RRQRRTS KLMKR (SEQ ID NO: 10)
[0072] FHV Coat-(35-49) RRRRNRTRRNRRRVR (SEQ ID NO: 11)
[0073] BMV Gag-(7-25) KMTRAQRRAAARRNRWTAR (SEQ ID NO: 12)
[0074] HTLV-II Rex-(4-16) TRRQRTRRARRNR (SEQ ID NO: 13)
[0075] D-Tat GRKKRRQRRRPPQ (SEQ ID NO: 14)
[0076] R9-Tat GRRRRRRRRRPPQ (SEQ ID NO: 15) [0077] Exemplary amphiphilic cell-penetrating peptides include the following:
Transportan GWTLNSAGYLLGKINLKALAALAKKIL chimera (SEQ ID
NO: 16)
[0079] MAP KLALKLALKLALALKLA (SEQ ID NO: 17)
[0080] SBP MGLGLHLLVLAAALQGAWSQPKKKRKV (SEQ ID NO: 18)
[0081] FBP GALFLGWLGAAGSTMGAWSQPKKKRKV (SEQ ID NO: 19) [0082] MPG ac-GALFLGFLGAAGSTMGAWSQPKKKRKV-cya (SEQ ID
NO:20)
[0083] MPG(ANlj¾) ac-GALFLGFLGAAGSTMGAWSQPKSKRKV-cya (SEQ ID
NO:21)
[0084] Pep-1 ac-KETWWETWWTEWSQPKKKRKV-cya (SEQ ID NO:22)
[0085] Pep-2 ac-KETWFETWFTEWSQPKKKRKV-cya (SEQ ID NO:23)
[0086] As used above, an acetyl group (ac) and a cysteamide group (Cya) are noted, by example. Cya brings more positive charge to the peptide, which in certain embodiments helps the peptide penetrate through cell membrane.
[0087] Accordingly, the term "proteinaceous composition" encompasses amino molecule sequences comprising at least one of the 20 common amino acids in naturally synthesized proteins, or at least one modified or unusual amino acid, including but not limited to those shown on Table 1 below.
Figure imgf000020_0001
TABLE 1
Modified and Unusual Amino Acids
Abbr. Amino Acid Abbr. Amino Acid
Bala β-alanine, β -Amino-propionic acid AHyl allo-Hydroxylysine
Abu 2-Aminobutyric acid 3Hyp 3-Hydroxyproline
4Abu 4- Aminobutyric acid, piperidinic acid 4Hyp 4-Hydroxyproline
Acp 6-Aminocaproic acid Ide Isodesmosine
Ahe 2-Aminoheptanoic acid Alle allo-Isoleucine
Aib 2-Aminoisobutyric acid MeGly N-Methylglycine,
sarcosine
Baib 3-Aminoisobutyric acid Mefle N-Methylisoleucine
Apm 2-Aminopimelic acid MeLys 6-N-Methyllysine
Dbu 2,4-Diaminobutyric acid MeVal N-Methylvaline
Des Desmosine Nva Norvaline
Dpm 2,2'-Diaminopimelic acid Nle Norleucine
Dpr 2,3-Diaminopropionic acid Orn Ornithine
EtGly N-Ethylglycine
[0088] In certain embodiments the proteinaceous composition comprises at least one protein, polypeptide or peptide. In further embodiments the proteinaceous composition comprises a biocompatible protein, polypeptide or peptide. As used herein, the term "biocompatible" refers to a substance that produces no significant untoward effects when applied to, or administered to, a given organism according to the methods and amounts described herein. Organisms include, but are not limited to mammals, such as humans, dogs, cats, horses, pigs, goats, and sheep. Such untoward or undesirable effects are those such as significant toxicity or adverse immunological reactions. In preferred embodiments, biocompatible protein, polypeptide or peptide containing compositions will generally be mammalian proteins or peptides or synthetic proteins or peptides each essentially free from toxins, pathogens and harmful immunogens.
[0089] Proteinaceous compositions may be made by any technique known to those of skill in the art, including the expression of proteins, polypeptides or peptides through standard molecular biological techniques, the isolation of proteinaceous compounds from natural sources, or the chemical synthesis of proteinaceous materials. The nucleotide and protein, polypeptide and peptide sequences for various genes have been previously disclosed, and may be found at computerized databases known to those of ordinary skill in the art. One such database is the National Center for Biotechnology Information's GenBank® and GenPept databases. The coding regions for these known genes may be amplified and/or expressed using the techniques disclosed herein or as would be known to those of ordinary skill in the art. Alternatively, various commercial preparations of proteins, polypeptides and peptides are known to those of skill in the art.
[0090] In certain embodiments a proteinaceous compound may be purified. Generally, "purified" will refer to a specific or protein, polypeptide, or peptide composition that has been subjected to fractionation to remove various other proteins, polypeptides, or peptides, and which composition substantially retains its activity, as may be assessed, for example, by the protein assays, as would be known to one of ordinary skill in the art for the specific or desired protein, polypeptide or peptide.
[0091] It is contemplated that virtually any protein, polypeptide or peptide containing component may be used in the compositions and methods disclosed herein. However, it is preferred that the proteinaceous material is biocompatible. In certain embodiments, it is envisioned that the formation of a more viscous composition will be advantageous in that will allow the composition to be more precisely or easily applied to the tissue and to be maintained in contact with the tissue throughout the procedure. In such cases, the use of a peptide composition, or more preferably, a polypeptide or protein composition, is contemplated. Ranges of viscosity include, but are not limited to, about 40 to about 100 poise. In certain aspects, a viscosity of about 80 to about 100 poise is preferred.
[0092] Proteins and peptides suitable for use in this invention may be autologous proteins or peptides, although the invention is clearly not limited to the use of such autologous proteins. As used herein, the term "autologous protein, polypeptide or peptide" refers to a protein, polypeptide or peptide which is derived or obtained from an organism. Organisms that may be used include, but are not limited to, a bovine, a reptilian, an amphibian, a piscine, a rodent, an avian, a canine, a feline, a fungal, a plant, or a prokaryotic organism, with a selected animal or human subject being preferred. The "autologous protein, polypeptide or peptide" may then be used as a component of a composition intended for application to the selected animal or human subject. In certain aspects, the autologous proteins or peptides are prepared, for example from whole plasma of the selected donor. The plasma is placed in tubes and placed in a freezer at about 80°C for at least about 12 hours and then centrifuged at about 12,000 times g for about 15 minutes to obtain the precipitate. The precipitate, such as fibrinogen may be stored for up to about one year (Oz, 1990).
[0093] In that the compositions of the present invention are particularly suitable for use in tissue adhesion and wound healing, preferred proteins are contemplated. Preferred protein include albumin, fibrinogen or gelatin, with albumin being most preferred.
[0094] To select other proteins, polypeptides, peptides and the like for use in the methods and compositions of the present invention, one would preferably select a proteinaceous material that possesses one or more of the following characteristics: it forms a solution with a high percentage of protenaceous material solubilized; it possesses a high viscosity (i.e. about 40 to about 100 poise); it has the correct molecular charge to bind the dye if it is a non-covalent mixture (i.e. anionic protein and cationic dye, or cationic protein and anionic dye); it has the correct amino-acids present to form covalent cross-links (i.e. one or more tyrosines, histidines, tryptophans and/or methionines); and/or it is biocompatible (i.e. from mammalian origin for mammals, preferably from human origin for humans, from canine origin for canines, etc. ; it is autologous; it is non-allergenic, and/or it is non-immunogenic).
IV. [0095] Biological Functional Equivalents
[0096] As modifications and/or changes may be made in the structure of the Tel polypeptides and and/or polynucleotides according to the present invention, while obtaining molecules having similar or improved characteristics, such biologically functional equivalents are also encompassed within the present invention.
A. [0097] Modified Polynucleotides and Polypeptides [0098] The biological functional equivalent may comprise a Tel polynucleotide that has been engineered to contain distinct sequences while at the same time retaining the capacity to encode the respective mutant protein. This can be accomplished to the degeneracy of the genetic code, i.e., the presence of multiple codons, which encode for the same amino acids. In one example, one of skill in the art may wish to introduce a restriction enzyme recognition sequence into a polynucleotide while not disturbing the ability of that polynucleotide to encode a mutant Tel protein.
[0099] In one example, a polynucleotide may encode a biologically functionally equivalent polypeptide wherein certain amino acids may be substituted for other amino acids without appreciable loss of repressor activity and, in certain cases, with enhanced repressor activity. In some instances these are "conservative" changes that do not disrupt the biological activity of the protein, as the structural change is not one that impinges of the protein's ability to carry out its designed function. In other cases, the change(s) is not conservative but still allows the polypeptide to retain functional activity. It is thus contemplated by the inventors that various changes may be made in the sequence of genes and proteins disclosed herein, while still fulfilling the goals of the present invention.
[0100] In terms of functional equivalents, it is well understood by the skilled artisan that, inherent in the definition of a "biologically functional equivalent" protein and/or polynucleotide, is the concept that there is a limit to the number of changes that may be made within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity. Biologically functional equivalents are thus defined herein as those proteins (and polynucleotides) wherein selected amino acids (or codons) may be substituted. Functional activity includes at least the ability to repress transcription of a polynucleotide, such as Bcl-xl, for example.
[0101] In general, the shorter the length of the molecule, the fewer changes that can be made within the molecule while retaining function. Longer domains may have an intermediate number of changes. The full-length protein will have the most tolerance for a larger number of changes. However, it must be appreciated that certain molecules or domains that are highly dependent upon their structure may tolerate little or no modification.
[0102] Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and/or the like. An analysis of the size, shape and/or type of the amino acid side-chain substituents reveals that arginine, lysine and/or histidine are all positively charged residues; that alanine, glycine and/or serine are all a similar size; and/or that phenylalanine, tryptophan and/or tyrosine all have a generally similar shape. Therefore, based upon these considerations, arginine, lysine and/or histidine; alanine, glycine and/or serine; and/or phenylalanine, tryptophan and/or tyrosine; are defined herein as biologically functional equivalents.
[0103] To effect more quantitative changes, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and/or charge characteristics, these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine ( 0.4); threonine ( 0.7); serine ( 0.8); tryptophan ( 0.9); tyrosine ( 1.3); proline ( 1.6); histidine ( 3.2); glutamate ( 3.5); glutamine ( 3.5); aspartate ( 3.5); asparagine ( 3.5); lysine ( 3.9); and/or arginine ( 4.5).
[0104] The importance of the hydropathic amino acid index in conferring interactive biological function on a protein is generally understood in the art (Kyte & Doolittle, 1982, incorporated herein by reference). It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index and/or score and/or still retain a similar biological activity. In making changes based upon the hydropathic index, the substitution of amino acids whose hydropathic indices are within +2 is preferred, those which are within +1 are particularly preferred, and/or those within +0.5 are even more particularly preferred.
[0105] It also is understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biological functional equivalent protein and/or peptide thereby created is intended for use in immunological embodiments, as in certain embodiments of the present invention. U.S. Patent 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and/or antigenicity, i.e., with a biological property of the protein.
[0106] As detailed in U.S. Patent No. 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ± 1); glutamate (+3.0 ± 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine ( 0.4); proline (-0.5 ± 1); alanine ( 0.5); histidine ( 0.5); cysteine ( 1.0); methionine ( 1.3); valine ( 1.5); leucine ( 1.8); isoleucine ( 1.8); tyrosine ( 2.3); phenylalanine ( 2.5); tryptophan ( 3.4). In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within +2 is preferred, those which are within +1 are particularly preferred, and/or those within +0.5 are even more particularly preferred.
B. [0107] Altered Amino Acids
[0108] The present invention, in many aspects, relies on the synthesis of peptides and polypeptides in cyto, via transcription and translation of appropriate polynucleotides. These peptides and polypeptides will include the twenty "natural" amino acids, and post-translational modifications thereof. However, in vitro peptide synthesis permits the use of modified and/or unusual amino acids. A table of exemplary, but not limiting, modified and/or unusual amino acids is provided herein.
C. [0109] Mimetics
[0110] In addition to the biological functional equivalents discussed above, the present inventors also contemplate that structurally similar compounds may be formulated to mimic the key portions of peptide or polypeptides of the present invention. Such compounds, which may be termed peptidomimetics, may be used in the same manner as the peptides of the invention and, hence, also are functional equivalents.
[0111] Certain mimetics that mimic elements of protein secondary and tertiary structure are described in Johnson et al. (1993). The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and/or antigen. A peptide mimetic is thus designed to permit molecular interactions similar to the natural molecule.
[0112] Some successful applications of the peptide mimetic concept have focused on mimetics of β-turns within proteins, which are known to be highly antigenic. Likely β turn structure within a polypeptide can be predicted by computer-based algorithms, as discussed herein. Once the component amino acids of the turn are determined, mimetics can be constructed to achieve a similar spatial orientation of the essential elements of the amino acid side chains. [0113] Other approaches have focused on the use of small, multidisulfide- containing proteins as attractive structural templates for producing biologically active conformations that mimic the binding sites of large proteins. Vita et al. (1998). A structural motif that appears to be evolutionarily conserved in certain toxins is small (30-40 amino acids), stable, and high permissive for mutation. This motif is composed of a beta sheet and an alpha helix bridged in the interior core by three disulfides.
[0114] Beta II turns have been mimicked successfully using cyclic L-pentapeptides and those with D-amino acids. Weisshoff et al. (1999). Also, Johannesson et al. (1999) report on bicyclic tripeptides with reverse turn inducing properties.
[0115] Methods for generating specific structures have been disclosed in the art. For example, alpha-helix mimetics are disclosed in U.S. Patents 5,446,128; 5,710,245; 5,840,833; and 5,859,184. These structures render the peptide or protein more thermally stable, also increase resistance to proteolytic degradation. Six, seven, eleven, twelve, thirteen and fourteen membered ring structures are disclosed.
[0116] Methods for generating conformationally restricted beta turns and beta bulges are described, for example, in U.S. Patents 5,440,013; 5,618,914; and 5,670,155. Beta- turns permit changed side substituents without having changes in corresponding backbone conformation, and have appropriate termini for incorporation into peptides by standard synthesis procedures. Other types of mimetic turns include reverse and gamma turns. Reverse turn mimetics are disclosed in U.S. Patents 5,475,085 and 5,929,237, and gamma turn mimetics are described in U.S. Patents 5,672,681 and 5,674,976.
V. [0117] Combination Treatments
[0118] In order to increase the effectiveness of the compositions of the Tel mutant protein of the invention, it may be desirable to combine these compositions with other agents effective in the treatment of hyperproliferative disease, such as anti-cancer agents. An "anticancer" agent is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, and/or increasing the lifespan of a subject with cancer. More generally, these other compositions would be provided in a combined amount effective to kill or inhibit proliferation of the cell. This process may involve contacting the cells with the expression construct and the agent(s) or multiple factor(s) at the same time. This may be achieved by contacting the cell with a single composition or pharmacological formulation that includes both agents, or by contacting the cell with two distinct compositions or formulations, at the same time, wherein one composition includes the expression construct and the other includes the second agent(s).
[0119] Tumor cell resistance to chemotherapy and radiotherapy agents represents a major problem in clinical oncology. One goal of current cancer research is to find ways to improve the efficacy of cancer therapy by combining it with other cancer therapy. In the context of the present invention, it is contemplated that Tel mutant therapy of the present invention could be used similarly in conjunction with chemotherapeutic, radiotherapeutic, immunotherapeutic, hormonal, or gene therapy intervention, for example.
[0120] In embodiments wherein the individual being treated has a solid tumor, such as a thoracic malignancy (such as lung cancer), the other cancer therapy may include surgery, radiation, immunotherapy, hormonal therapy, or chemotherapy, including carboplatin, cisplatin, docetaxel, erlotinib, etoposide, gemcitabine, irinotecan, paclitaxel, pemetrexed, topotecan, vinorelbine, and/or combinations thereof. In embodiments wherein the individual being treated has mesothelioma, for example, the other cancer therapy may include surgery, radiation, immunotherapy, hormonal therapy, or chemotherapy, such as cisplatin, raltitrexed, pemetrexed, gemcitabine, vinorelbine, carboplatin, and/or combinations thereof.
[0121] The present invention may be used simultaneously or may precede or follow the other agent treatment by intervals ranging from minutes to weeks. In embodiments where the other agent and Tel mutant are applied separately to the cell, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the other agent and Tel mutant would still be able to exert an advantageously combined effect on the cell. In such instances, it is contemplated that one may contact the cell with both modalities within about 12-24 h of each other and, more preferably, within about 1-12 h of each other. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several d (2, 3, 4, 5, 6 or 7) to several wk (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations. [0122] Various combinations may be employed, Tel mutant therapy is "A" and the secondary agent, such as radiation therapy or chemotherapy, for example, is "B":
[0123] A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B
[0124] B/B/B/A B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A
[0125] B/A/B/A B/A/A/B A/A/A/B B/A/A/A A/B/A/A A/A/B/A
[0126] Administration of the therapeutic Tel mutants of the present invention to a patient will follow general protocols for the administration of chemotherapeutics, taking into account any toxicity. It is expected that the treatment cycles would be repeated as necessary. It also is contemplated that various standard therapies, as well as surgical intervention, may be applied in combination with the described hyperproliferative cell therapy.
A. Chemotherapy
[0127] Cancer therapies also include a variety of combination therapies with both chemical-and radiation-based treatments. Combination chemotherapies include, for example, cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrexate, or any analog or derivative variant of the foregoing. One or more chemotherapy agents may be included in the chemotherapy.
B. [0128] Radiation Therapy
[0129] Other factors that cause DNA damage and have been used extensively include what are commonly known as γ-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other forms of DNA damaging factors are also contemplated such as microwaves and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
[0130] The terms "contacted" and "exposed," when applied to a cell, are used herein to describe the process by which a therapeutic construct and a chemotherapeutic or radiotherapeutic agent are delivered to a target cell or are placed in direct juxtaposition with the target cell. To achieve cell killing or stasis, both agents are delivered to a cell in a combined amount effective to kill the cell or prevent it from dividing.
C. [0131] Immunotherapy
[0132] Immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells. The immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell. The antibody alone may serve as an effector of therapy or it may recruit other cells to actually effect cell killing. The antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent. Alternatively, the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target. Various effector cells include cytotoxic T cells and NK cells.
[0133] Immunotherapy, thus, could be used as part of a combined therapy, in conjunction with the present therapy. The general approach for combined therapy is discussed below. Generally, the tumor cell must bear some marker that is amenable to targeting, i.e., is not present on the majority of other cells. Many tumor markers exist and any of these may be suitable for targeting in the context of the present invention. Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and pi 55.
D. [0134] Genes
[0135] In yet another embodiment, the secondary treatment is a gene therapy in which a therapeutic polynucleotide is administered before, after, or at the same time as the Tel mutant. A variety of proteins are encompassed within the invention, including antisense directed against inducers of cellular proliferation; inhibitors of cellular proliferation, such as tumor suppressors, including p53, pl6, C-CAM, Rb, APC, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, zacl, p73, VHL, MMAC1 / PTEN, DBCCR- 1, FCC, rsk-3, p27, p27/pl6 fusions, p21/p27 fusions, anti-thrombotic genes (e.g., COX-1, TFPI), PGS, Dp, E2F, ras, myc, neu, raf, erb, fms, trk, ret, gsp, hst, abl, El A, p300, genes involved in angiogenesis (e.g. , VEGF, FGF, thrombospondin, BAI- 1, GDAIF, or their receptors) and MCC; and regulators of programmed cell death, such as members of the Bcl-2 family, ICE-like proteases, and so forth.
E. [0136] Surgery
[0137] Approximately 60% of persons with cancer will undergo surgery of some type, which includes preventative, diagnostic or staging, curative and palliative surgery. Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene therapy, immunotherapy and/or alternative therapies.
[0138] Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed. Tumor resection refers to physical removal of at least part of a tumor. In addition to tumor resection, treatment by surgery includes laser surgery, cryosurgery, electro surgery, and miscopically controlled surgery (Mohs' surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue.
[0139] Upon excision of part of all of cancerous cells, tissue, or tumor, a cavity may be formed in the body. Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy. Such treatment may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. These treatments may be of varying dosages as well.
F. [0140] Other agents
[0141] It is contemplated that other agents may be used in combination with the present invention to improve the therapeutic efficacy of treatment. These additional agents include immunomodulatory agents, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adehesion, or agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers. Immunomodulatory agents include tumor necrosis factor; interferon alpha, beta, and gamma; IL- 2 and other cytokines; F42K and other cytokine analogs; or MIP-1 , MIP-lbeta, MCP-1, RANTES, and other chemokines. It is further contemplated that the upregulation of cell surface receptors or their ligands such as Fas / Fas ligand, DR4 or DR5 / TRAIL would potentiate the apoptotic inducing abililties of the present invention by establishment of an autocrine or paracrine effect on hyperproliferative cells. Increases intercellular signaling by elevating the number of GAP junctions would increase the anti-hyperproliferative effects on the neighboring hyperproliferative cell population. In other embodiments, cytostatic or differentiation agents can be used in combination with the present invention to improve the anti-hyerproliferative efficacy of the treatments. Inhibitors of cell adehesion are contemplated to improve the efficacy of the present invention. Examples of cell adhesion inhibitors are focal adhesion kinase (FAKs) inhibitors and Lovastatin. It is further contemplated that other agents that increase the sensitivity of a hyperproliferative cell to apoptosis, such as the antibody c225, could be used in combination with the present invention to improve the treatment efficacy.
[0142] Hormonal therapy may also be used in conjunction with the present invention or in combination with any other cancer therapy previously described. The use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or estrogen. This treatment is often used in combination with at least one other cancer therapy as a treatment option or to reduce the risk of metastases.
VI. [0143] Pharmaceutical Preparations
[0144] Pharmaceutical compositions of the present invention comprise an effective amount of one or more Tel mutant compositions of the invention dissolved or dispersed in a pharmaceutically acceptable carrier. The phrases "pharmaceutical or pharmacologically acceptable" refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate. The preparation of an pharmaceutical composition that contains at least one composition of the present invention or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference. Moreover, for animal (e.g. , human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards. [0145] As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329, incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the pharmaceutical compositions is contemplated.
[0146] The composition of the present invention may comprise different types of carriers depending on whether it is to be administered in solid, liquid or aerosol form, and whether it need to be sterile for such routes of administration as injection. The present invention can be administered intravenously, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, topically, intramuscularly, subcutaneously, mucosally, orally, topically, locally, inhalation (e.g. , aerosol inhalation), injection, infusion, continuous infusion, localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g. , liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, incorporated herein by reference).
[0147] The composition of the present invention may be formulated into a composition in a free base, neutral or salt form. Pharmaceutically acceptable salts, include the acid addition salts, e.g. , those formed with the free amino groups of a proteinaceous composition, or which are formed with inorganic acids such as for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric or mandelic acid. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as for example, sodium, potassium, ammonium, calcium or ferric hydroxides; or such organic bases as isopropylamine, trimethylamine, histidine or procaine. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as formulated for parenteral administrations such as injectable solutions, or aerosols for delivery to the lungs, or formulated for alimentary administrations such as drug release capsules and the like.
[0148] Further in accordance with the present invention, the composition of the present invention suitable for administration is provided in a pharmaceutically acceptable carrier with or without an inert diluent. The carrier should be assimilable and includes liquid, semisolid, i.e., pastes, or solid carriers. Except insofar as any conventional media, agent, diluent or carrier is detrimental to the recipient or to the therapeutic effectiveness of a the composition contained therein, its use in administrable composition for use in practicing the methods of the present invention is appropriate. Examples of carriers or diluents include fats, oils, water, saline solutions, lipids, liposomes, resins, binders, fillers and the like, or combinations thereof. The composition may also comprise various antioxidants to retard oxidation of one or more component. Additionally, the prevention of the action of microorganisms can be brought about by preservatives such as various antibacterial and antifungal agents, including but not limited to parabens (e.g., methylparabens, propylparabens), chlorobutanol, phenol, sorbic acid, thimerosal or combinations thereof.
[0149] In accordance with the present invention, the composition is combined with the carrier in any convenient and practical manner, i.e., by solution, suspension, emulsification, admixture, encapsulation, absorption and the like. Such procedures are routine for those skilled in the art.
[0150] In a specific embodiment of the present invention, the composition is combined or mixed thoroughly with a semi- solid or solid carrier. The mixing can be carried out in any convenient manner such as grinding. Stabilizing agents can be also added in the mixing process in order to protect the composition from loss of therapeutic activity, i.e., denaturation in the stomach. Examples of stabilizers for use in an the composition include buffers, amino acids such as glycine and lysine, carbohydrates such as dextrose, mannose, galactose, fructose, lactose, sucrose, maltose, sorbitol, mannitol, etc.
[0151] In further embodiments, the present invention may concern the use of a pharmaceutical lipid vehicle compositions that include one or more compositions of the present invention, one or more lipids, and an aqueous solvent. As used herein, the term "lipid" will be defined to include any of a broad range of substances that is characteristically insoluble in water and extractable with an organic solvent. This broad class of compounds are well known to those of skill in the art, and as the term "lipid" is used herein, it is not limited to any particular structure. Examples include compounds which contain long-chain aliphatic hydrocarbons and their derivatives. A lipid may be naturally occurring or synthetic (i.e., designed or produced by man). However, a lipid is usually a biological substance. Biological lipids are well known in the art, and include for example, neutral fats, phospholipids, phosphoglycerides, steroids, terpenes, lysolipids, glycosphingolipids, glycolipids, sulphatides, lipids with ether and ester-linked fatty acids and polymerizable lipids, and combinations thereof. Of course, compounds other than those specifically described herein that are understood by one of skill in the art as lipids are also encompassed by the compositions and methods of the present invention.
[0152] One of ordinary skill in the art would be familiar with the range of techniques that can be employed for dispersing a composition in a lipid vehicle. For example, the composition of the present invention may be dispersed in a solution containing a lipid, dissolved with a lipid, emulsified with a lipid, mixed with a lipid, combined with a lipid, covalently bonded to a lipid, contained as a suspension in a lipid, contained or complexed with a micelle or liposome, or otherwise associated with a lipid or lipid structure by any means known to those of ordinary skill in the art. The dispersion may or may not result in the formation of liposomes.
[0153] The actual dosage amount of a composition of the present invention administered to an animal patient can be determined by physical and physiological factors such as body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject.
[0154] In certain embodiments, pharmaceutical compositions may comprise, for example, at least about 0.1% of an active compound. In other embodiments, the an active compound may comprise between about 2% to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein. Naturally, the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half-life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
[0155] In other non-limiting examples, a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100 microgram/kg/body weight, about 200 microgram/kg/body weight, about 350 microgram/kg/body weight, about 500 microgram/kg/body weight, about 1 milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc., can be administered, based on the numbers described above.
A. [0156] Alimentary Compositions and Formulations
[0157] In preferred embodiments of the present invention, the Tel mutant compositions of the present invention are formulated to be administered via an alimentary route. Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract. Specifically, the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft- shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
[0158] In certain embodiments, the active compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like (Mathiowitz et ah, 1997; Hwang et ah, 1998; U.S. Pat. Nos. 5,641,515; 5,580,579 and 5,792, 451, each specifically incorporated herein by reference in its entirety). The tablets, troches, pills, capsules and the like may also contain the following: a binder, such as, for example, gum tragacanth, acacia, cornstarch, gelatin or combinations thereof; an excipient, such as, for example, dicalcium phosphate, mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate or combinations thereof; a disintegrating agent, such as, for example, corn starch, potato starch, alginic acid or combinations thereof; a lubricant, such as, for example, magnesium stearate; a sweetening agent, such as, for example, sucrose, lactose, saccharin or combinations thereof; a flavoring agent, such as, for example peppermint, oil of wintergreen, cherry flavoring, orange flavoring, etc. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar, or both. When the dosage form is a capsule, it may contain, in addition to materials of the above type, carriers such as a liquid carrier. Gelatin capsules, tablets, or pills may be enterically coated. Enteric coatings prevent denaturation of the composition in the stomach or upper bowel where the pH is acidic. See, e.g., U.S. Pat. No. 5,629,001. Upon reaching the small intestines, the basic pH therein dissolves the coating and permits the composition to be released and absorbed by specialized cells, e.g., epithelial enterocytes and Peyer's patch M cells. A syrup of elixir may contain the active compound sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compounds may be incorporated into sustained-release preparation and formulations.
[0159] For oral administration the compositions of the present invention may alternatively be incorporated with one or more excipients in the form of a mouthwash, dentifrice, buccal tablet, oral spray, or sublingual orally- administered formulation. For example, a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution). Alternatively, the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically- effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants. Alternatively the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
[0160] Additional formulations which are suitable for other modes of alimentary administration include suppositories. Suppositories are solid dosage forms of various weights and shapes, usually medicated, for insertion into the rectum. After insertion, suppositories soften, melt or dissolve in the cavity fluids. In general, for suppositories, traditional carriers may include, for example, polyalkylene glycols, triglycerides or combinations thereof. In certain embodiments, suppositories may be formed from mixtures containing, for example, the active ingredient in the range of about 0.5% to about 10%, and preferably about 1% to about 2%.
B. [0161] Parenteral Compositions and Formulations
[0162] In further embodiments, the Tel mutant composition of the invention may be administered via a parenteral route. As used herein, the term "parenteral" includes routes that bypass the alimentary tract. Specifically, the pharmaceutical compositions disclosed herein may be administered for example, but not limited to intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally (see U.S. Pat. Nos. 6,613,308, 5,466,468, 5,543,158; 5,641,515; and 5,399,363, each specifically incorporated herein by reference in its entirety).
[0163] Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Patent 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy injectability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (i.e., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
[0164] For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in isotonic NaCl solution and either added hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
[0165] Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. A powdered composition is combined with a liquid carrier such as, e.g., water or a saline solution, with or without a stabilizing agent.
C. [0166] Miscellaneous Pharmaceutical Compositions and Formulations
[0167] In other preferred embodiments of the invention, the Tel mutant compound may be formulated for administration via various miscellaneous routes, for example, topical (i.e., transdermal) administration, mucosal administration (intranasal, vaginal, etc.) and/or inhalation. [0168] Pharmaceutical compositions for topical administration may include the active compound formulated for a medicated application such as an ointment, paste, cream or powder. Ointments include all oleaginous, adsorption, emulsion and water-solubly based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only. Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin. Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram. Possible bases for compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base. Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the active ingredient and provide for a homogenous mixture. Transdermal administration of the present invention may also comprise the use of a "patch". For example, the patch may supply one or more active substances at a predetermined rate and in a continuous manner over a fixed period of time.
[0169] In certain embodiments, the pharmaceutical compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described e.g., in U.S. Pat. Nos. 5,756,353 and 5,804,212 (each specifically incorporated herein by reference in its entirety). Likewise, the delivery of drugs using intranasal microparticle resins (Takenaga et ah, 1998) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 5,725, 871, specifically incorporated herein by reference in its entirety) are also well-known in the pharmaceutical arts. Likewise, transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U.S. Pat. No. 5,780,045 (specifically incorporated herein by reference in its entirety).
[0170] The term aerosol refers to a colloidal system of finely divided solid of liquid particles dispersed in a liquefied or pressurized gas propellant. The typical aerosol of the present invention for inhalation will consist of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent. Suitable propellants include hydrocarbons and hydrocarbon ethers. Suitable containers will vary according to the pressure requirements of the propellant. Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms. VII. Kits of the Invention
[0171] Any of the compositions described herein may be comprised in a kit. In a non-limiting example, one or more Tel mutants are comprised in a kit and are housed in suitable container means.
[0172] The kits may comprise a suitably aliquoted Tel mutant of the present invention. When there are multiple components, the components of the kits may be packaged either in aqueous media or in lyophilized form, for example. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there are more than one component in the kit, the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present invention also will typically include a means for containing the Tel mutant and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.
[0173] When the components of the kit are provided in one and/or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. The Tel mutants may also be formulated into a syringeable composition, in which case, the container means may itself be a syringe, pipette, and/or other such like apparatus, from which the formulation may be applied to an infected area of the body, injected into an individual, and/or even applied to and/or mixed with the other components of the kit.
[0174] However, the components of the kit may be provided as dried powder(s). When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
[0175] The kits of the present invention will also typically include a means for containing the vials in close confinement for commercial sale, such as, e.g., injection and/or blow-molded plastic containers into which the desired vials are retained.
[0176] Irrespective of the number and/or type of containers, the kits of the invention may also comprise, and/or be packaged with, an instrument for assisting with the injection/administration and/or placement of the ultimate therapeutic composition within the body of an animal. Such an instrument may be a syringe, pipette, forceps, and/or any such medically approved delivery vehicle.
VIII. [0177] Examples
[0178] The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
EXAMPLE 1
EXEMPLARY MATERIALS AND METHODS
[0179] Cell lines and Reagents: The human mesothelioma cell line 145 and the human lung cancer cell line HI 299 were maintained in RPMI 1640 medium (Thermo Scientific Waltham, MA) supplemented with 10% Fetal Bovine Serum (Invitrogen, Carlsbad, CA). A549 human lung adenocarcinoma cells were maintained in F-12K medium (ATCC, Manassas, VA). Tel cDNA was purchased from Origene Corp (Rockville, MD). All Tel mutants were generated through standard DNA mutagenesis. Anti-Bcl-xl antibodies and all anti-MAP kinase antibodies were purchased from Cell Signaling Technology (Beverly, MA). All antibodies used to detect ETS proteins were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-Actin monoclonal antibody was purchased from Sigma- Aldrich (St. Louis, MO).
[0180] Immunoprecipitations: For Tel phosphorylation studies, cancer cells were transfected with wild type and mutant Tel cDNAs using Lipofectamine 2000 (Invitrogen) in a 10 cm dish and cultured for 24 hours in serum-containing media. The transfected cells were serum starved overnight and exposed to lOOng/ml of EGF or HGF (Invitrogen) for 20 minutes. The cells were then lysed using RIPA lysis buffer. Proteins were immunoprecipitated using antibodies against Tel, Ets-2 and PU. l with the Catch and Release v 2.0 kit (Millipore, Billerica, MA) overnight and equal amounts were resolved on a 4-20 % Tris-glycine gel. The signals were visualized using the ECL system. [0181] Chromatin immunoprecipitation assay: 145 and A549 cells were plated at a density of 5 X 105 per well in a 6 well plate and transfected with Tel and Tel S213A expressing plasmids using Lipofectamine 2000. The cells were serum- starved with or without a lOOng/ml HGF or EGF simulation for 20 minutes. The chromatin was crosslinked by treating the transfected cells with 1% formaldehyde for 15 minutes, sonicated to an average length of 1000 bp and immunoprecipitated with Tel antibody (Santa Cruz) at 4oC overnight. The immunoprecipitated chromatin was then reverse crosslinked and PCR amplified with primers specific for BCL-XL promoter (forward 5 '-GCCTAAGGCGGATTTGA ATGTAG-3 ' (SEQ ID NO: l); reverse 5 '-GAAGGGAGAGAAAGAGATTC AGGAA-3 '(SEQ ID NO:2)).
[0182] Luciferase assay: A549, H1299 and 145 cells were transfected in triplicate in a 24 well plate using Lipofectamine 2000 with 200 ng of Tel or Tel S213A cDNA and pXL plasmid generated by cloning the human Bcl-xl promoter into the pGL2 vector (Promega, Madison, WI). A plasmid expressing β-Gal (25 ng) was used as an internal control for transfection efficiency. The transfected cells were cultured in serum containing media for 24 hours and then subjected to serum starvation and stimulation by HGF/EGF for 20 minutes. Luciferase and β- Galactosidase activities were measured and the luciferase values were normalized to the β- Galactosidase control.
[0183] Immunofluorescence studies: Cells transfected with Tel or Tel S213A cDNAs were cultured in chamber slides at a density of 105 cells. The transfected cells were then serum-starved overnight and stimulated with EGF for 20 minutes. The cells were later fixed in 2.5% formaldehyde and immunostained using a primary Tel antibody and a fluorescein isothiocyanate (FITC)-conjugated secondary antibody plus a 5 ng/ml concentration of Hoechst dye and visualized using fluorescence microscopy.
[0184] XTT assays: A549 and I 45 cells were seeded in 96 well plates at 5xl03 cells per well and then transfected with various amounts of Tel and Tel S213A using Lipofectamine 2000. The transfected cells were cultured for a further 48 hours and assayed following the standard protocol provided with an XTT kit (Roche, Indianapolis, IN).
[0185] Clonogenic assays: Clonogenic assays were performed to determine the cytotoxic effects of Tel and the Tel S213A. I 45 and A549 cells were transfected with Tel and Tel S213A at various concentrations by electroporation using nucleofector solution L (Lonza Group, Basel, Switzerland). lxlO4 cells were then plated per 10 mm dish in triplicate for each concentration and cultured for 2 weeks. The cells were then fixed using 2.5% formaldehyde and stained with 1% crystal violet.
[0186] Western blotting: Western blotting was performed using a standard protocol. Briefly, I 45, H1299 and A549 cells were transfected with Tel, Tel S213A and Tel S213A/K11R cDNA and cultured for 48 hours. The cells were then lysed and equal amounts of total protein were resolved on 4-20 % Tris-glycine gels. The resolved proteins were then transferred onto a nitrocellulose membrane and the membranes were incubated with antibodies raised against Bcl2 family proteins. The ECL system was used to visualize the signals.
[0187] Flow cytometry: Samples (add treatment) were run on an Acuri C6 flow cytometer (Accuri Cyto meters Inc, Ann Arbor, MI ) using a 488nm laser and fluorescence emissions for FITC (Annexin V) and PI (propidium iodide) were measured. The data were then analyzed using FlowJo software (Tree Star Corp, Ashland, OR).
[0188] Immunohistochemistry: Tissue arrays (US Biomax, Rockville, MD) were deparaffinized in xylene substitute and rehydrated in PBS. Antigen retrieval was performed with citrate buffer (pH 6.5) for 20 minutes at 99°C, followed by the block of endogenous peroxidase activity. Sections were incubated with blocking serum in PBS containing 5% bovine serum albumin, followed by incubation with rabbit anti-human Bcl-xl polyclonal antibody or with rabbit anti-phosphorylated human EGFR polyclonal antibody for 24 hours, followed by incubation with a biotinylated goat secondary anti-rabbit antibody (1:200 dilution, Vector Laboratories, Burlingame, CA). Immunoreactive signals were detected using a streptavidin- biotin-peroxidase complex (Vector Laboratories), according to the manufacturer's recommended procedures. All of the slides were counterstained with hematoxylin (Sigma- Aldrich).
[0189] Statistical analysis: The relationships between Bcl-xl, and EGFR were analyzed statistically using Chi-square analysis. Differences between Tel and Tel S213A in colony formation and XTT assays were measured statistically using the student t-test. EXAMPLE 2
THE ACTIVATION OF RECEPTOR KINASES INDUCES THE PHOSPHORYLATION
OF TEL
[0190] To evaluate whether c-Met and EGFR activated the same MAPK signal transduction pathways, the phosphorylation status was evaluated of all three major families of mitogen- activated protein kinases (MAPKs) and other intracellular kinases, such as Akt, GSK-3, and p70 S6 kinase with Proteome Profiler™ Human Phospho-MAPK Arrays. The array signals, shown in FIG. 1A, indicated that kinase activities from growth factor stimulated cells were much stronger than those from serum-starved 145 cells. There were no significant differences found between the c-Met and EGFR groups. The activation of MAPKs was further confirmed by western blotting.
[0191] To examine how the activation of growth factor receptors may affect ETS functions, the phosphorylation of the ETS-2, PU. l and Tel proteins was analyzed in 145 cells under conditions of serum starvation or HGF stimulation by immunoprecipitation and western blot analysis. Whereas the levels of total ETS proteins were observed to be equivalent in the cells, the levels of phosphorylated ETS-2, PU.l and Tel were clearly elevated (FIG. IB). Similar results were observed in lung cancer cells upon EGF stimulation. The inventors then mapped the serine sites that were phosphorylated after growth factor exposure. FIG. 1C demonstrated that only S213A mutation prevented Tel being phosphorylated.
EXAMPLE 3
TEL PHOSPHORYLATION NEGATIVELY REGULATES THE TEL-BCL-XL PROMOTER INTERACTION UPON THE GROWTH FACTOR STIMULATION OF
CELLS
[0192] The effects of Tel on the Bcl-xl promoter were analyzed by CHIP assay (FIG. 2). Compared with the unstimulated samples, HGF or EGF stimulation resulted in a significantly decreased PCR signal from the chromatin precipitated from Tel wide-type cDNA transfected cells. In contrast, HGF or EGF stimulation have little impact on PCR signals from Tel S213A plasmids transfected cells. This result proves that phosphorylation is the key modification to prevent Tel's binding to Bcl-xl promoter DNA. EXAMPLE 4
HGF FAILS TO CAUSE THE RELOCALIZATION OF TEL S213A TO THE
CYTOPLASM
[0193] To better understand the impact of S213 phosphorylation on the repression of the Bcl-xl promoter by Tel, the subcellular distribution of wild- type Tel and Tel S213A was compared using fluorescent microscopy. I 45 cells were transfected with wild-type Tel and Tel S213A and then subjected to overnight serum starvation at 48 hours after transfection. Twenty minutes after the HGF stimulation of serum-starved I 45 cells, the Tel S213A mutant was retained in the nucleus, whereas wild-type Tel proteins showed increased cytoplasmic accumulation (FIG. 3A).
[0194] To further determine whether Tel S213A is a stronger repressor of Bcl-xl promoter activity than wild type Tel, Bcl-xl promoter construct XLp-Luc, Tel and Tel S213A cDNA expression vectors were co-transfected into A549 and I 45 cells. As shown in FIG. 3B, Bcl-xl promoter activity is significantly decreased in serum-starved Tel transfected 145 cells in comparison with the I 45 cells under growth factor stimulation. In contrast, Tel S213A expression leads to decreased Bcl-xl expression under both growth factor stimulated and growth factor deprived conditions. These results indicate that phosphorylation at S213 is critical for Tel localization and its regulation of Bcl-xl transcriptional activities.
EXAMPLE 5
TEL S213A IS A MORE POTENT REPRESSOR OF BCL-XL EXPRESSION THAN
WILD TYPE TEL
[0195] To investigate whether serum starvation enhances the repressive function of Tel upon Bcl-xl expression, GFP, wild type Tel and Tel S213A cDNAs were expressed in 145 cells under normal growth or serum starvation conditions, or under growth factor stimulation for 24 hours. Bcl-xl expression was found to be significantly decreased in the serum-starved 145 GFP transfected cells in comparison with the I45/GFP cells cultured under normal growth conditions. This was also the result for the Tel transfected cells. In contrast, the repression of Bcl-xl by Tel S213A was unaffected by either serum starvation or growth factor stimulation (FIG. 4A). [0196] The expressions of Bcl-2 family proteins were evaluated by western blotting following the transfection of cancer cells with Tel, Tel S213A and control expression vector. As shown in FIG. 4B, the expression of Bcl-xl was decreased below basal levels after Tel cDNA transfection. Bcl-xl expression was further reduced when Tel S213A was expressed in both A549 and 145 cells, whereas the Bcl-2 levels remained unchanged. Mcl-1 was found to be upregulated in these cells on a western blot suggesting that an apoptosis "defense mechanism" operates in these tumor cells (Cao et ah, 2007; Xu et ah, 2010).
EXAMPLE 6
TEL S213A IS MORE POTENT INDUCER OF APOPTOSIS THAN WILD TYPE TEL
[0197] The short term cell killing effects of Tel and Tel S213A were evaluated in vitro in A549 and 145 cells, which were transfected with 0.2 μg Tel and Tel S213A in 96 well plates. The cell viability was determined 48 hours after transfection via an XTT assay. The transfection of Tel S213A resulted in a significant loss of cell viability (FIG. 5A). Colony formation assays were performed to further measure the growth inhibitory effects of Tel and Tel S213A upon tumor cells. The results further demonstrated that Tel S213A is a far stronger repressor of cell growth than Tel (FIG. 5B).
[0198] To determine whether Tel or Tel S213A induced cell growth inhibition via an apoptotic response, A549 cells were transfected with both for 48 hours, and the apoptosis were evaluated using an Annexin V binding assay and Flow Cytometry. The results shown in FIG. 5C indicate that the forced expression of Tel or Tel S213A causes a dramatic increase in the proportion of apoptotic cells. Tel transfection resulted in 19+3 % apoptosis, whilst the Tel S213A expressing population of apoptotic cells was 26 + 2.5 % (student t-test, P<0.05).
EXAMPLE 7
SER 213 PHOSPHORYLATION AND Kll SUMOYLATION ON TEL SYNERGISTICALLY PROMOTE ITS NUCLEAR EXPORT
[0199] To elucidate the role of the sumoylation of Tel at Kl l, in addition to the phosphorylation of Tel at Ser 213, upon its nuclear export, the possible synergistic interaction of sumoylation and phosphorylation on the repression of Bcl-xl was then measured. A549 cells were transfected with either GFP, wild-type Tel, Tel S213A, Tel Kl lR, Tel K11R/S213A and co-transfected with the Bcl-xl promoter construct, XLp-Luc. The resulting Bcl-xl promoter activity levels, shown in FIG. 6A, demonstrated that the expression of the Tel K11R/S213A mutant caused the strongest Bcl-xl repression. The similar result was obtained in measuring Bcl- xl expression levels via western blotting (FIG. 6B).
EXAMPLE 8
EGFR PHOSPHORYLATION AND BCL-XL EXPRESSION POSITIVELY
CORRELATE IN PRIMARY TUMOR SAMPLES
[0200] Given the positive association observed between EGFR activation and Bcl- xl expression in cell culture, we examined whether such a relationship existed in primary human mesothelioma and non-small cell lung adenocarcinoma samples. By immunohistochemical staining analysis using mesothelioma tissue arrays as well as non small cell lung adenocarcinoma tissue arrays, the protein expression profile of Bcl-xl was analyzed and the phosphorylated EGFR levels in 38 mesothelioma patient samples was evaluated, as well as 44 non small cell lung adenocarcinoma samples. Chi-square analysis revealed positive correlations between EGFR activation and Bcl-xl expression in both mesothelioma and lung cancer samples (FIG. 7).
EXAMPLE 9
SIGNIFICANCE OF CERTAIN EMBODIMENTS OF THE INVENTION
[0201] The current data show that the activation of EGFR and Met, both of which are amplified in lung cancer and mesothelioma, share an identical signaling pathway. In this embodiment, the inhibition of one receptor might have no effect if downstream effectors are constitutively activated or if parallel pathways are switched on (Li et ah, 2003; Hu et ah, 2008). Targeting the common downstream proteins of these receptors, such as Bcl-xl, Akt and their associated transcription factors are a viable alternative to receptor inhibition approaches, in certain embodiments of the invention.
[0202] The present findings clearly show that EGFR and Met activation lead to the phosphorylation of ETS transcription activators and repressors. As demonstrated herein, phosphorylated Tel accumulates in the cytoplasm after cell stimulation by growth factors. Once phosphorylated, Tels are removed from their cognate DNA-binding sites and their repression of Bcl-xl transcription is thereby abrogated. Activated MAP kinases can also phosphorylate ETS-2 and PU. l, which will stimulate their nuclear import or reduce their nuclear export (Petrovic et ah, 2003; Foulds et ah, 2004). The coordinate regulation of both activating and repressing ETS family members leads to rapid and robust changes in gene expression to allow the cell to survive, as illustrated in FIG. 8.
[0203] The current data also indicate that Tel Ser213 phosphorylation is the key response to Bcl-xl transcriptional repression. Tel has multiple serine and threonine sites that can potentially be phosphorylated by ERK kinase (Irvin et ah, 2003). Although in certain embodiments Ser213 (not Ser257) is the only Tel site phosphorylated by ERK in the tested cell lines, Ser 257 has been reported to be the target of p38, which is an activator of different stress signals (Hanson et ah, 2008). Hence, the phosphorylation of Tel by both ERK and p38 is a unique property of this ETS protein. Both ERK and stress signaling pathways converge via Tel in the nucleus under certain circumstances and provide further stimulation of Bcl-xl transcription in cancer cells.
[0204] The identified phosphorylation sites on Tel (S213) reside within the linker inhibitory damper (LID) domain, outside its ETS DNA-binding domain. Recent study has indicated that the autoregulation of Tel-promoter interaction involves two domains, the C- terminal inhibitory domain (CID) and LID domains. CID (amino acids 423-452) negatively interferes with the Tel DNA binding domain. LID domain (amino acids 124-338) serves as negative regulator of CID (Green et ah, 2010). Decreased Tel binding to DNA due to the phosphorylation of S213 indicates that LID, once phosphorylated, in certain embodiments of the invention loses its inhibitory interaction with the CID, which would negatively regulate Tel- promoter binding.
[0205] In addition to being regulated by phosphorylation, Tel is also modified by sumoylation at Kl l and K99 (Wood et ah, 2003; Chakrabarti et ah, 2000). The present studies indicate that the double mutant Tel K11R/S213A is a stronger repressor of Bcl-xl transcription than either single mutation alone. In addition, in certain embodiments whereas both phosphorylation and sumoylation are required for Tel export, the order in which Tel is phosphorylated and sumoylated is unclear, as is whether these two types of modification function cooperatively or independently. Studies of other ETS members, such as NET and YAN, indicate that in certain aspects phosphorylation and sumoylation generally work synergistically to mediate the downregulatory nuclear export of the ETS transcriptional repressors (Tootle and Rebay, 2005).
[0206] In summary, negative regulation of Tel function following tyrosine kinase receptor activation contributes to the overexpression of the Bcl-xl protein in cancer cells. The present data thus provide new insights into the molecular regulation of multiple receptor- dependent signal transduction pathways, the phosphorylation of ETS transcriptional repressors, and the transcriptional regulation of Bcl-xl.
REFERENCES
[0207] The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
PATENTS
[0208 U.S. Patent No. 4,554,101
[0209 U.S. Patent No. 5,466,468
[0210 U.S. Patent No. 5,543,158
[0211 U.S. Patent No. 5,641,515
[0212 U.S. Patent No. 5,399,363
[0213 U.S. Patent 5,440,013
[0214 U.S. Patent 5,618,914
[0215 U.S. Patent 5,670,155
[0216 U.S. Patent 5,446,128
[0217 U.S. Patent 5,710,245
[0218 U.S. Patent 5,840,833
[0219 U.S. Patent 5,859,184 [0220] U.S. Patent 5,929,237
[0221] U.S. Patent 5,475,085
[0222] U.S. Patent 5,672,681 [0223] U.S. Patent 5,674,976 [0224] U.S. Patent No. 6,613,308
PUBLICATIONS
[0225] Bachleitner-Hofmann T, Sun MY, Chen C-T, et al. HER kinase activation confers resistance to MET tyrosine kinase inhibition in MET oncogene-addicted gastric cancer cells. Molecular Cancer Therapeutics 2008; 7: 3499-3508.
[0226] Cao X, Rodarte C, Zhang L, et al. Bcl2/bcl-xL inhibitor engenders apoptosis and increases chemosensitivity in mesothelioma. Cancer Biol Ther 2007; 6: 246-252.
[0227] Cao X, Littlejohn J, Rodarte C, et al. Up-Regulation of Bcl-xl by Hepatocyte Growth Factor in Human Mesothelioma Cells Involves ETS Transcription Factors. Am J Pathol 2009; 175: 2207-2216.
[0228] Chakrabarti SR, Sood R, Nandi S, et al. Posttranslational modification of TEL and TEL/AML1 by SUMO-1 and cell-cycle-dependent assembly into nuclear bodies. Proceedings of the National Academy of Sciences of the United States of America 2000; 97: 13281-13285.
[0229] Dai C, Li Y, Yang J, et al. Hepatocyte Growth Factor Preserves Beta Cell Mass and Mitigates Hyperglycemia in Streptozotocin-induced Diabetic Mice. Journal of Biological Chemistry 2003; 278: 27080-27087.
[0230] Dazzi H, Hasleton PS, Thatcher N, et al. Malignant pleural mesothelioma and epidermal growth factor receptor (EGF-R). Relationship of EGF-R with histology and survival using fixed paraffin embedded tissue and the F4, monoclonal antibody. Br J Cancer 1990; 61: 924-926. [0231] Foulds CE, Nelson ML, Blaszczak AG, et al. Ras/mitogen- activated protein kinase signaling activates Ets-1 and Ets-2 by CBP/p300 recruitment. Mol Cell Biol 2004; 24: 10954-10964.
[0232] Green SM, Coyne HJ, Mcintosh LP, et al. DNA Binding by the ETS Protein TEL (ETV6) Is Regulated by Autoinhibition and Self-association. Journal of Biological Chemistry 2010; 285: 18496-18504.
[0233] Guo A, Villen J, Kornhauser J, et al. Signaling networks assembled by oncogenic EGFR and c-Met. Proceedings of the National Academy of Sciences 2008; 105: 692- 697.
[0234] Hanson CA, Wood LD, Hiebert SW. Cellular stress triggers TEL nuclear export via two genetically separable pathways. Journal of Cellular Biochemistry 2008; 104: 488- 498.
[0235] Herbst RS, Shin DM. Monoclonal antibodies to target epidermal growth factor receptor-positive tumors. Cancer 2002; 94: 1593-1611.
[0236] Hu YP, Patil SB, Panasiewicz M, et al. Heterogeneity of Receptor Function in Colon Carcinoma Cells Determined by Cross-talk between Type I Insulin-like Growth Factor Receptor and Epidermal Growth Factor Receptor. Cancer Research 2008; 68: 8004-8013.
[0237] Irvin BJ, Wood LD, Wang L, et al. TEL, a Putative Tumor Suppressor, Induces Apoptosis and Represses Transcription of Bcl-XL. Journal of Biological Chemistry 2003; 278: 46378-46386.
[0238] Johannesson et al., 1999, "Bicyclic tripeptide mimetics with reverse turn inducing properties." J. Med. Chem. 42:601-608.
[0239] Jost M, Huggett TM, Kari C, et al. Epidermal Growth Factor Receptor- dependent Control of Keratinocyte Survival and Bcl-xL Expression through a MEK-dependent Pathway. Journal of Biological Chemistry 2001; 276: 6320-6326.
[0240] Lee J, Kannagi M, Ferrante RJ, et al. Activation of Ets-2 by oxidative stress induces Bcl-xL expression and accounts for glial survival in amyotrophic lateral sclerosis. The FASEB Journal 2009; 23: 1739-1749. [0241] Li B, Chang CM, Yuan M, et al. Resistance to small molecule inhibitors of epidermal growth factor receptor in malignant gliomas. Cancer Res 2003; 63: 7443-7450.
[0242] Maki K, Arai H, Waga K, et al. Leukemia-Related Transcription Factor TEL Is Negatively Regulated through Extracellular Signal-Regulated Kinase-Induced Phosphorylation. Mol Cell Biol 2004; 24: 3227-3237.
[0243] Mukohara T, Civiello G, Davis Π, et al. Inhibition of the Met Receptor in Mesothelioma. Clinical Cancer Research 2005; 11: 8122-8130.
[0244] Nelson ML, Kang H-S, Lee GM, et al. Ras signaling requires dynamic properties of Etsl for phosphorylation-enhanced binding to coactivator CBP. Proceedings of the National Academy of Sciences; 107: 10026-10031.
[0245] Petrovic N, Bhagwat SV, Ratzan WJ, et al. CD13/APN transcription is induced by RAS/MAPK-mediated phosphorylation of Ets-2 in activated endothelial cells. J Biol Chem 2003; 278: 49358-49368.
[0246] Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company,
1990.
[0247] Rogers SJ, Box C, Chambers P, et al. Determinants of response to epidermal growth factor receptor tyrosine kinase inhibition in squamous cell carcinoma of the head and neck. J Pathol 2009; 218: 122-130.
[0248] Roukens MG, Alloul-Ramdhani M, Moghadasi S, et al. Downregulation of Vertebrate Tel (ETV6) and Drosophila Yan Is Facilitated by an Evolutionarily Conserved Mechanism of F-Box-Mediated Ubiquitination. Mol Cell Biol 2008; 28: 4394-4406.
[0249] Sekharam M, Zhao H, Sun M, et al. Insulin-like Growth Factor 1 Receptor Enhances Invasion and Induces Resistance to Apoptosis of Colon Cancer Cells through the Akt/Bcl-xL Pathway. Cancer Research 2003; 63: 7708-7716.
[0250] Sevilla L, Aperlo C, Dulic V, et al. The Ets2 Transcription Factor Inhibits Apoptosis Induced by Colony-Stimulating Factor 1 Deprivation of Macrophages through a Bcl- xL-Dependent Mechanism. Mol Cell Biol 1999; 19: 2624-2634. [0251] Sevilla L, Zaldumbide A, Carlotti F, et al. Bcl-xL Expression Correlates with Primary Macrophage Differentiation, Activation of Functional Competence, and Survival and Results from Synergistic Transcriptional Activation by Ets2 and PU.l. Journal of Biological Chemistry 2001; 276: 17800-17807.
[0252] Silverman ES, Baron RM, Palmer LJ, et al. Constitutive and Cytokine- Induced Expression of the ETS Transcription Factor ESE-3 in the Lung. Am J Respir Cell Mol Biol 2002; 27: 697-704.
[0253] Sridhar SS, Seymour L, Shepherd FA. Inhibitors of epidermal-growth- factor receptors: a review of clinical research with a focus on non-small-cell lung cancer. Lancet Oncol 2003; 4: 397-406.
[0254] Tootle TL, Rebay I. Post-translational modifications influence transcription factor activity: a view from the ETS superfamily. Bioessays 2005; 27: 285-298.
[0255] Vita et al., 1998, "Novel miniproteins engineered by the transfer of active sites to small natural scaffolds." Biopolymers 47:93-100.
[0256] Wang L, Hiebert SW. TEL contacts multiple co-repressors and specifically associates with histone deacetylase-3. Oncogene 2001; 20: 3716-3725.
[0257] Wei G-H, Badis G, Berger MF, et al. Genome- wide analysis of ETS-family DNA-binding in vitro and in vivo. EMBO J; 29: 2147-2160.
[0258] Weisshoff et al., 1999, "Mimicry of beta ΙΓ -turns of proteins in cyclic pentapeptides with one and without D-amino acids." Eur. J. Biochem. 259:776-788.
[0259] Whittaker S, Marais R, Zhu AX. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene; 29: 4989-5005.
[0260] Wood LD, Irvin BJ, Nucifora G, et al. Small ubiquitin-like modifier conjugation regulates nuclear export of TEL, a putative tumor suppressor. Proc Natl Acad Sci U S A 2003; 100: 3257-3262. [0261] Xu H, Krystal GW. Actinomycin D decreases Mcl-1 expression and acts synergistically with ABT-737 against small cell lung cancer cell lines. Clin Cancer Res; 16: 4392-4400.
[0262] Although the present invention and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined by the appended claims. Moreover, the scope of the present application is not intended to be limited to the particular embodiments of the process, machine, manufacture, composition of matter, means, methods and steps described in the specification. As one of ordinary skill in the art will readily appreciate from the disclosure of the present invention, processes, machines, manufacture, compositions of matter, means, methods, or steps, presently existing or later to be developed that perform substantially the same function or achieve substantially the same result as the corresponding embodiments described herein may be utilized according to the present invention. Accordingly, the appended claims are intended to include within their scope such processes, machines, manufacture, compositions of matter, means, methods, or steps.

Claims

CLAIMS What is claimed is:
1. A recombinant TEL polypeptide comprising an ETS DNA-binding domain (ETS) and a) a sterile alpha motif (SAM) domain having a deletion or an amino acid substitution at a lysine or a valine; and/or b) a linker inhibitory damper domain (LID) domain having a deletion or an amino acid substitution at one or more serines; and/or c) an amino acid substitution at a lysine corresponding to position 11 in SEQ ID NO:3.
2. The polypeptide of claim 1, further comprising a protein transduction domain, a nuclear localization signal, and/or a C- terminal inhibitory domain (CID) domain.
3. The polypeptide of claim 1, wherein the polypeptide has increased transcriptional repressor activity over wild-type TEL polypeptide.
4. The polypeptide of claim 1, further defined as having an amino acid substitution at the serine corresponding to position 213 in SEQ ID NO:3.
5. The polypeptide of claim 1, further defined as having an amino acid substitution at the serine corresponding to position 257 in SEQ ID NO:3.
6. The polypeptide of claim 1, further defined as having an amino acid substitution at the lysine corresponding to position 99 in SEQ ID NO:3.
7. The polypeptide of claim 1, further defined as having an amino acid substitution at the valine corresponding to position 112 in SEQ ID NO:3.
8. The polypeptide of claim 2, wherein the protein transduction domain is HIV TAT, Antennapedia, penetratin, SynBl, SynB3, PTD-4, PTD-5, FHV Coat-(35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-Tat, R9-Tat, Transportan, MAP, SBP, FBP, MPG, MPG(ANLS), Pep-1, or Pep-2.
9. A method of treating an individual for cancer, comprising the step of administering to the individual a therapeutically effective amount of a polypeptide of claim 1.
10. The method of claim 9, wherein the individual has a thoracic malignancy.
11. The method of claim 9, wherein the individual has mesothelioma.
12. The method of claim 9, further comprising an additional therapy for cancer.
13. A kit comprising the polypeptide of claim 1, said polypeptide housed in a suitable container.
14. An isolated recombinant TEL polypeptide comprising: a) an amino acid substitution at one or more sites in the polypeptide corresponding to SEQ ID NO:3, said sites selected from the group consisting of a serine corresponding to position 213, a serine corresponding to position 257, a lysine corresponding to position 11, a lysine corresponding to position 99, and a valine corresponding to position 112; or b) a deletion in the polypeptide corresponding to SEQ ID NO:3, said deletion occurring in one or more of a protein transduction domain, a TAT domain, a SAM domain, a LID domain, an ETS domain, or a CID domain, wherein the polypeptide has transcriptional repressor activity.
15. A method of treating an individual for cancer, comprising the step of administering to the individual a therapeutically effective amount of a polypeptide of claim 14.
16. The method of claim 15, wherein the individual has a thoracic malignancy.
17. The method of claim 15, wherein the individual has mesothelioma.
18. The method of claim 15, further comprising an additional therapy for cancer.
19. A kit comprising the polypeptide of claim 14, said polypeptide housed in a suitable container.
20. An isolated recombinant Tel polypeptide comprising a TAT
domain, an amino acid substitution at the valine at position 112 relative to SEQ ID NO:3, and that lacks at least part of the CID domain.
21. The polypeptide of claim 20, wherein the polypeptide comprises SEQ ID NO:43.
PCT/US2012/025608 2011-02-17 2012-02-17 Targeted cancer therapy using ets transcriptional inhibitors Ceased WO2012112869A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161443918P 2011-02-17 2011-02-17
US61/443,918 2011-02-17

Publications (2)

Publication Number Publication Date
WO2012112869A2 true WO2012112869A2 (en) 2012-08-23
WO2012112869A3 WO2012112869A3 (en) 2012-10-11

Family

ID=46673197

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/025608 Ceased WO2012112869A2 (en) 2011-02-17 2012-02-17 Targeted cancer therapy using ets transcriptional inhibitors

Country Status (1)

Country Link
WO (1) WO2012112869A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180141977A1 (en) * 2015-05-06 2018-05-24 Norwegian University Of Science And Technology (Ntnu) Anti-bacterial agents and their use in therapy

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GREEN ET AL.: 'DNA binding by the ETS protein TEL (ETV6) is regulated by autoinhibition and self-association' THE JOURNAL OF BIOLOGICAL CHEMISTRY vol. 285, no. 24, 11 June 2010, pages 18498 - 18504 *
MAKI ET AL.: 'Leukemia-related transcription factor TEL is negatively regulated through extracellular signal-regulated kinase-induced phosphorylation' MOLECULAR AND CELLULAR BIOLOGY vol. 24, no. 8, April 2004, pages 3227 - 3237 *
ROUKENS ET AL.: 'Identification of a new site of sumoylation on Tel (ETV6) uncovers a PIAS-dependent mode of regulating Tel function' MOLECULAR AND CELLULAR BIOLOGY vol. 28, no. 7, April 2008, pages 2342 - 4357 *
WOOD ET AL.: 'Small ubiquitin-like modifier conjugation regulates nuclear export of TEL, a putative tumor suppressor' PNAS vol. 100, no. 6, 18 March 2003, pages 3257 - 3262 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180141977A1 (en) * 2015-05-06 2018-05-24 Norwegian University Of Science And Technology (Ntnu) Anti-bacterial agents and their use in therapy
US10570180B2 (en) * 2015-05-06 2020-02-25 Norwegian University Of Science And Technology (Ntnu) Anti-bacterial agents and their use in therapy

Also Published As

Publication number Publication date
WO2012112869A3 (en) 2012-10-11

Similar Documents

Publication Publication Date Title
KR102494803B1 (en) Peptide having angiogenesis inhibitory activity and composition containing same
JP6030622B2 (en) MUC-1 cytoplasmic domain peptide as an inhibitor of cancer
US9527895B2 (en) CAPCNA peptide therapeutics for cancer
KR20160089523A (en) Composition for treating prostate cancer
US7745405B2 (en) Peptides selectively lethal to malignant and transformed mammalian cells
EA010291B1 (en) Method of protecting mammals against radiation
CN114026114A (en) BNIP3 peptide for the treatment of reperfusion injury
US20220048973A1 (en) B1SP Fusion Protein Therapeutics, Methods, and Uses
CN107629114B (en) Polypeptide, its derivative and its application in the preparation of anti-pulmonary fibrosis medicine
CN112292402B (en) Anti-tumor fusion protein and preparation method and application thereof
US9044421B2 (en) Treating MUC1-expressing cancers with combination therapies
WO2012061015A2 (en) L-citrulline supplementation during arginine depletion therapy with arginase
WO2019173478A2 (en) Methods of treating or preventing cancer with an agent that depletes tregs and a checkpoint inhibitor
WO2012112869A2 (en) Targeted cancer therapy using ets transcriptional inhibitors
CA2477878C (en) Peptides selectively lethal to malignant and transformed mammalian cells
AU2006235742B2 (en) Conjugate comprising P21 protein for the treatment of cancer
WO2013134116A1 (en) Hsp90 inhibitor for the treatment of cancer and inflammatory diseases
US7183257B2 (en) Use of RPL41 to treat infections and inhibit cancer
WO2014164394A1 (en) Combination anti-her2 cancer therapy using muc1 peptides and chemotherapeutics
KR102275912B1 (en) Peptide for inhibiting hsp expression and composition containing same
CN103534273B (en) Cyclized allosteric TRAIL/APO2L and coding gene and application thereof
EA022915B1 (en) Methods of treating acute myeloid leukemia and myelodysplastic syndrome
US9327013B2 (en) Activity enhancer for anticancer agent
US20130225503A1 (en) Tex14 peptides as novel antitumor agents
US20220096594A1 (en) Macrocyclic peptides for targeted inhibition of autophagy

Legal Events

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

Ref document number: 12746829

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12746829

Country of ref document: EP

Kind code of ref document: A2