AU2007296429A1 - Compositions and methods to prevent cancer with cupredoxins - Google Patents

Compositions and methods to prevent cancer with cupredoxins Download PDF

Info

Publication number
AU2007296429A1
AU2007296429A1 AU2007296429A AU2007296429A AU2007296429A1 AU 2007296429 A1 AU2007296429 A1 AU 2007296429A1 AU 2007296429 A AU2007296429 A AU 2007296429A AU 2007296429 A AU2007296429 A AU 2007296429A AU 2007296429 A1 AU2007296429 A1 AU 2007296429A1
Authority
AU
Australia
Prior art keywords
cupredoxin
peptide
azurin
cancer
residues
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2007296429A
Inventor
Ananda Chakrabarty
Tapas Das Gupta
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.)
University of Illinois
Original Assignee
University of Illinois
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 University of Illinois filed Critical University of Illinois
Publication of AU2007296429A1 publication Critical patent/AU2007296429A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/104Pseudomonadales, e.g. Pseudomonas
    • 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

Description

WO 2008/033987 PCT/US2007/078371 COMPOSITIONS AND METHODS TO PREVENT CANCER WITH CUPREDOXINS CROSS REFERENCE TO RELATED APPLICATIONS 5 This application claims priority under 35 US.C. §§ 119 and 120, of Provisional U.S. Application Serial No. 60/844,358, filed September 14, 2006, which claims priority to U.S. Patent Application No. U.S. Patent Application No. 11/244,105, filed October 6, 2005, which claims priority to U.S. Provisional Patent Application Serial No. 60/616,782, filed October 7, 2004, and U.S. Provisional Patent Application Serial No. 60/680,500, filed May 13, 2005, 10 and is a continuation-in-part of U.S. Patent Application Serial Number 10/720,603, filed November 11, 2003. which claims priority to U.S. Provisional Patent Application Serial No. 60/414,550, filed August 15, 2003, and which is a continuation-in-part of U.S. Patent Application Serial Number 10/047,710, filed January 15, 2002, which claims priority to U.S. Provisional Patent Application Serial Number 60/269,133, filed February 15, 2001. The 15 entire content of these prior applications is fully incorporated herein by reference. STATEMENT OF GOVERNMENTAL INTEREST The subject matter of this application has been supported by research grants from the National Institutes of Health (NIH), Bethesda, Maryland, U.S.A., (Grant Numbers Al 16790 20 21, ES 04050-16, AI 45541, CA09432 and N01-CM97567). The government may have certain rights in this invention. FIELD OF THE INVENTION The present invention relates to compositions comprising variants, derivatives and 25 structural equivalents of cupredoxins that inhibit the development of premalignant lesions in mammalian cells, tissues and animals. The invention also relates to the use of cupredoxins, and variants, derivatives and structurally equialcnts of cupredoxi.nis. as chemopreventive agents in mammals to inhibit the development of premalignant lesions, and ultimately cancer. BACKGROUND 30 Cancer chemoprevention is the use of natural, synthetic or biologic chemical agents to reverse, suppress, or prevent carcinogenic progression to invasive cancer. Recent clinical trials in preventing cancer in high-risk populations suggest that chemopreventive therapy is a realistic treatment for high-rik patients. Chemopreventive therapy is based on the concepts of multifbcal field carcinogenesis and umtistep caremnogenesis. In fied carcinogenesis, WO 2008/033987 PCT/US2007/078371 generalized carcinogen exposure throughout the tissue field results in diffuse epithelia] injury in tissue and clonal proliferation of the mutated cells. These genetic mutations throughout the field increase the likelihood that one or more premalignant or malignant lesions may develop in the field. Multistep carcinogenesis in the stepwise accumulation of these genetic 5 and phenotypic alterations. Arresting one or more steps in the multistep cancinogenesis may impede or prevent the development of cancer. See generally Tsao et al, CA Cancer J Clin 54:150-180 (2004). Azurin, and other cupredoxins, are cytotoxic specifically towards cancer cells. Azurin induces apoptosis in J774 lung cancer cells. Yamada et al., PNAS 99(22):14098-14103 10 (2002). On entry into J774 lung cancer cells, azurin localizes in the cytosol and nuclear fractions, and forms a complex with tumor suppressor protein p53, thereby stabilizing it and enhancing its intracellular level. Id. The induction of azurin-mediated apoptosis is not limited to J774 cells. Azurin can also enter cancer cells such as human melanoma UISO Mel-2 or human breast cancer MCF-7 cells. Yamada et aL, Infect Immun. 70:7054-7062 15 (2002); Punj et al., Oncogene. 23:2367-2378 (2004). In both cases, azurin allowed the elevation of the intracellular p53 levels, leading to enhanced Bax formation and induction of apoptosis in such cells. Most interestingly, intraperitoneal injection of azurin in nude mice harboring xenografted Mel-2 or MCF-7 human cancers led to statistically significant regression of such cancers. Id. 20 The mouse mammary gland organ culture (MMOC) assay may be used to evaluate the inhibitory effects of potential chemopreventive agents on both hormone-induced structural differentiation of mammary glands and on the development of DMBA-induced preneoplastic hyperplastic alveolar nodule-like lesions in the gland. Mammary glands from young, virgin animals, when incubated for 6 days in the presence of insulin (1) + prolactin (P) - aldosterone 25 (A), can differentiate into fully-grown glands. These glands morphologically resemble the glands obtained from. pregnant mice. Aldosterone can be replaced by estrogen (E) progesterone (Pg) Inclusion of hydrocortisone (H) to the medium stimulates the functional differentiation of the mammary glands. Mehta and Banerjee, Acta Endocrinol. 80:501 (1975); Mehta and Moon, Breast Cancer: Treatment and Prognosis 300, 300 (Basil A Stoll 30 ed., Blackweil Press 1986). Thus, the hormone-induced structural and functional dffentiation, observed in this culture system, mimics the responses to hormones observed during various physiological stages of the animal. -2- WO 2008/033987 PCT/US2007/078371 Mice exhibit a distinct preneoplastic stage prior to cancer formation in MMOC, Such preneoplastic lesions in C3H mice are induced by murine mammary tumor virus or in BALB/c mice by DMBA. Exposure of the glands to 2 pg/ml DMBA between days 3 and 4 of growth phases followed by regression of the glands for 2-3 weeks in the medium 5 containing only insulin, results in the formation of mammary alveolar lesions (MAL). Hawthorne et al., Pharmaceutical Biology 40:70-74 (2002); Mehta et al., Methods in Cell Science 19:19-24 (1997). Furthermore, transplantation of epithelial cells. prepared from glands containing the DMBA-induced mammary lesions, into syngeneic host resulted in the development of mammary adenocarcinoma. Telang et al, PNAS 76:5886-5890 (1979). 10 Pathologically, these tumors were similar to those observed in vivo when mice of the same strain are administered DMBA. Id. DMBA-induced mammary lesion formation in MMOC can be inhibited by a variety of classes of chemopreventive agents such as retinoids. These agents include chemopreventive agents derived from the natural products such as brassinin and resveretrol, 15 thiols, antioxidants, inhibitors of orithine decarboxylase such as OFMO and deguelin, inhibitors of prostaglandin synthesis, Ca regulators, etc.. Jang et al., Science 275:218-220 (1997); Mehta, Eur. J. Cancer 36:1275-1282 (2000); Metha et aL, J. Nat]. Cancer Inst. 89:212-219 (1997). These studies clearly demonstrate that this organ culture system offers a unique model to determine the effectiveness of compounds against mammary carcinogenesis. 20 The results can be expected to closely correlate to the inhibition obtained by in vivo administration of such compounds. The MMOC may also be induced to form mammary ductal lesions (MDL). The MDL can be induced if estrogen and progesterone instead of aldosterone and hydrocortisone are included in the medium. The alveolar structures in the presence of ovarian steroids are very 25 small but the intraductal lesions are observed in histopathological sections. Mehta et alt- J. Natl. Cancer Inst. 93:1103-1106 (2001). The antiestrogens, which selectively work on ovarian hormone dependent ER+ breast cancers such as tamoxifen, inhibited MDL formation and not MAL Thus, this modified culture model in addition to conventional MAL induction protocol now can be used to evaluate effects of chemopreventive agents on both MAL and 30 MOL. What is needed is a chemopreventive agent that inhibit the development of premnalignant lesions. Such a chenopreventive agent should he able to either prevent the -3- WO 2008/033987 PCT/US2007/078371 initial development of premalignant lesions, induce cell death in premalignant lesions that form, and/or prevent the development of premalignant lesions into malignant lesions. Such chemopreventive agents would have great utility in treating, in particular, patients who are at a high risk of developing cancer, due to either the presence of high-risk features, the presence 5 of pre-malignant lesions, or the previous of cancer or premalignant lesions. SUMMARY OF THE EMBODIMENTS The present invention relates to compositions comprising peptides that may be variants, derivatives and structural equivalents of cupredoxins that inhibit the development of 10 premalignant lesions in mammalian cells, tissues and animals. Specifically, these compositions may comprise azurin from Pseudononas aeruginosa, and/or the 50-77 residue region of azurin (p28). The present invention further relates to compositions that may comprise cupredoxin(s), and/or variants, derivatives or structural equivalents of cupredoxins, that retain the ability to inhibit the development of premalignant lesions in mammalian cells, 15 tissues or animals. These compositions may be isolated peptides or pharmaceutical compositions, among others. The compositions of the invention may be used in methods to prevent the development of cancer in mammalian patients. One aspect of the invention are isolated peptides that may be a variant, derivative or structural equivalent of a cupredoxin; and may inhibit the development of premalignant 20 lesions in mammalian tissue. The cupredoxin may be azurin, pseudoazurin, plastocyanin, rusticyanin, Laz, auracyanin, stellacyanin and cucumber basic protein, and specifically may be azurin. The eupredoxin may be from an organism such as Pseudomonas aeruginosa, A lcaligenes faecalis, Ulva pertussis, A chromobacter xylosoxidan, Bordetella bronchiseptica, Methylomonas sp., Neisseria meningitidis, Neisseria gonorrhea, Pseudomonas fluoreseens, 25 Pseudomonas chlororaphis, Xyleila fastidiosa and Vibrio parahaemoltieus, and specifically may be Pseudomonas aeruginosa. In some embodiments, the peptide may be part of SEQ ID NOS: 1, 3-19, or has at least 80% amino acid sequence identity to SEQ ID NOS: i, 3-19. In some embodiments, the isolated peptide may be a truncation of a cupredoxin. The isolated peptide may be more than about 10 residues and not more than about 100 residues. 30 The isolated peptide may comprise, or alternatively consist of, Pseudom.onas aeruginosa azurn residues 50-77, Pseudomonas aeruginosa azurin residues 50-67, Pseudomonas aeruginosa azurn residues 36- 88, or SEQ ID NOS: 20-24. -4- WO 2008/033987 PCT/US2007/078371 Another aspect of the invention is a pharmaceutical composition that may comprise at least one, or two, cupredoxins or isolated peptides of the invention in a pharmaceutically acceptable carrier. The pharmaceutical composition may be formulated for intravenous administration. In some embodiments, the cupredoxin in the pharmaceutical composition 5 may be from an organism such as Pseudomonas aeruginosa, Uva pertussis, A Icaligenes faecalis, Achromobacter xylosoxidan, Bordetella bronchiseptica, Meth ylomonas sp,, Neisseria meningitidis, Neisseria gonorrhea, Pseudomonas fluorescens, Pseudononas chlororaphis, Xvlella fastidiosa and Vibrio parahaemolytieus, and specifically may be from Pseudomonas aeruginosa. The cupredoxin may be S EQ ID NOS: 1, 3-19. 10 Another aspect of the invention is a method to treat a mammalian patient by administering to the patient a therapeutically effective amount of the pharmaceutical composition of the invention. The patient may be human, and may be at a higher risk to develop cancer than the general population. In some embodiments, the cancer may be melanoma, breast, pancreas, glioblastoma, astrocytoma, lung, colorectal, neck and head, 15 bladder, prostate, skin, or cervical cancer. In some embodiments, the patient may have at least one high risk feature, premalignant lesions or have been cured of cancer or premalignant lesions. The pharmaceutical composition may be administered by intravenous injection, intramuscular injection, subcutaneous injection, inhalation, topical administration, 20 transdermal patch, suppository, vitreous injection or oral, and specifically may be administered by intravenous injection. The pharmaceutical composition may be co administered with at least one other chemopreventive drug, and specifically at about the same time as another chemopreventive drug. Another aspect of the invention is a kit comprising the pharmaceutical composition of 25 the invention in a vial. The kit may be designed for intravenous administration. Another aspect of the invention is a method to study the development of cancer comprising contacting mammalian cells with a cupredoxin or peptide of the invention and measuring the development of premalignant and malignant cells. In some embodiments, the cells may be human and/or mammary cells. In some embodiments, the cells are induced to 30 develop premalignant lesions. Another aspect of the invention is an expression vector, which encodes a peptide of the invention. -5- WO 2008/033987 PCT/US2007/078371 These and other aspects, advantages, and features of the invention will become apparent from the following figures and detailed description of the specific embodiments. BRIEF DESCRIPTION OF THE SEQUENCES 5 SEQ ID NO: 1. Amino acid sequence of azurin from Pseudomonas aeruginosa. SEQ ID NO: 2. Amino acid sequence of p28, Pseudomonas aeruginosa azurin residues 50 77. SEQ ID NO: 3. Amino acid sequence of plastocyanin from Phormidium laminosum. SEQ ID NO: 4. Amino acid sequence of rusticyanin from Thiobacillusferrooxidans. 10 SEQ ID NO: 5. Amino acid sequence of pseudoazurin from Achromobacter cycloclastes. SEQ ID NO: 6. Amino acid sequence of azurin from Alcaligenes faecalis. SEQ ID NO: 7. Amino acid sequence of azurin from A chromobacter xylosoxidans ssp. denitrificans . SEQ [D NO: 8. Amino acid sequence of azurin from Bordetella bronchiseptica. 15 SEQ ID NO: 9. Amino acid sequence of azurin from.Methylomonas sp. J. SEQ ID NO: 10. Amino acid sequence of azurin from Neisseria meningitidis Z2491. SEQ ID NO: 11. Amino acid sequence of azurin from Pseudomonasfluorescen. SEQ ID NO: 12. Amino acid sequence of azurin from Pseudomonas chlororaphis. SEQ ID NO: 13. Amino acid sequence of azurin from Xylellafastidiosa 9a5c. 20 SEQ ID NO: 14. Amino acid sequence of stellacyanin from Cucumis sativus. SEQ ID NO: 15. Amino acid sequence of auracyanin A from Chloroflexus aurantiacus. SEQ ID NO: 16. Amino acid sequence of auracyanin B from Chloroflexus aurantiacus. SEQ ID NO: 17. Amino acid sequence of cucumber basic protein from Cucumis sativus. SEQ ID NO: 18. Amino acid sequence of Laz from Neisseria gonorrhoeae F62. 25 SEQ ID NO:19. Amino acid sequence of the azurin from Vibrio parahaeiolyticus. SEQ ID NO: 20. Amino acid sequence of amino acids 57 to 89 of auracyanin B of Chloroflexus aurantiacus. SEQ ID NO: 21. Amino acid sequence of amino acids 51-77 of Pseudomonas syringe azunn. 30 SEQ ID NO: 22, Amino acid sequence of amino acids 89-115 of Neisseria meningitidis Laz. SEQ ID NO: 23. Amino acid sequence of amino acids 52-78 of Vibrio parahaemolytcus azurm. -6- WO 2008/033987 PCT/US2007/078371 SEQ ID NO: 24. Amino acid sequence of amino acids 51-77 of Bordetella bronchiseptica azurn. BRIEF DESCRIPTION OF THE FIGURES 5 Figure 1. Figure 1 depicts photographs of all of the glands evaluated for the efficacy of p28 and azurin. Figure IA shows a representative photograph of alveolar lesions in a DMBA treated gland and its comparison with a gland that was treated with DMBA along with a chemopreventive agent. Figures IB-IG show representative photographs of the effects of p28 on the development of alveolar lesions. 10 Figure 2. Figure 2 depicts a graph showing the efficacy of p28 against DMBA-induced mammary alveolar lesions. Figure 3. Figure 3 depicts photographs of representative sections of ductal lesions and effect of p28. Figure 4. Figure 4 depicts a graph showing the efficacy of p28 against DMBA-induced 15 ductal lesions DETAILED DESCRIPTION OF THE INVENTION Definitions As used herein, the term "cell" includes either the singular or the plural of the term, 20 unless specifically described as a "single cell." As used herein, the terms "polypeptide," "peptide," and "protein" are used interchangeably to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical analogue of a corresponding naturally occurring amino acid. The terms also apply to naturally occurring 25 amino acid polymers. The terms polypeptidee" "peptide," and "protein" are also inclusive of modifications including, but not limited to, giycosylation, lipid attachment, sulfation, gamma carboxylation of giutamic acid residues, hydroxvlation and ADP-ribosylation. It will be appreciated that polvpeptides are not always entirely linear. For instance, polypeptides may be branched as a result of ubiquitination and they may be circular (with or without 30 branching), generally as a result of post-translation events, including natural processing event and events brought about by human manipulation which do not occur naturally. Circular, hranched and branched circular polypeptides mnay be synthesized by non-ranslation natural process and by entirely synthetic methods as well. -7- WO 2008/033987 PCT/US2007/078371 As used herein, the term "pharmacologic activity" means the effect of a drug or other chemical on a biological system. The effect of chemical may be beneficial (therapeutic) or harmful (toxic). The pure chemicals or mixtures may be of natural origin (plant, animal, or mineral) or may be synthetic compounds. 5 As used herein, the term "premalignant" means precancerous, or before abnormal cells divide without control. As used herein, the term "lesion" means an area of abnormal tissue. As used herein, the term "pathological condition" includes anatomic and physiological deviations from the normal that constitute an impairment of the normal state of 10 the living animal or one of its parts, that interrupts or modifies the performance of the bodily functions, and is a response to various factors (as malnutrition, industrial hazards, or climate), to specific infective agents (as worms, parasitic protozoa, bacteria, or viruses), to inherent defects of the organism (as genetic anomalies), or to combinations of these factors. As used herein, the term "condition" includes anatomic and physiological deviations 15 from the normal that constitute an impairment of the normal state of the living animal or one of its parts, that interrupts or modifies the performance of the bodily functions. As used herein, the term "suffering from" includes presently exhibiting the symptoms of a pathological condition, having a pathological condition even without observable symptoms, in recovery from a pathological condition, or recovered from a pathological 20 condition. As used herein, the term "chemoprevention" is the use of drugs. vitamins, or other agents to try to reduce the risk of, or delay the development or recurrence of, cancer. A used herein, the term "treatment" includes preventing, lowering, stopping, or reversing the progression or severity of the condition or symptoms associated with a 25 condition being treated. As such, the term treatmentn includes medical, therapeutic. and/or prophylactic administration, as appropriate. Treatment may also include preventing or lessening the development of a condition, such as cancer. As used herein, the term "inhibit cell growth" means the slowing or ceasing of cell division and/or cell expansion. This term also includes the inhibition of cell development or 30 increases in cell death. A therapeuticallyy effective amount" is an amount effective to prevent, lower, stop or reverse the development of, or to partial or totally alleviate the existing symptoms o a -8- WO 2008/033987 PCT/US2007/078371 particular condition for which the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art. The term "substantially pure," as used herein, when used to modify a protein or other cellular product of the invention, refers to, for example. a protein isolated from the growth 5 medium or cellular contents, in a form substantially free of, or unadulterated by, other proteins and/or other compounds. The term "substantially pure" refers to a factor in an amount of at least about 75%. by dry weight, of isolated fraction. or at least "75% substantially pure." More specifically, the term "substantially pure" refers to a compound of at least about 85%, by dry weight, of isolated fraction, or at least "85% substantially pure." 10 Most specifically, the term "substantially pure" refers to a compound of at least about 95%, by dry weight, of isolated fraction, or at least "95% substantially pure." The term "substantially pure" may also be used to modify a synthetically-made protein or compound of the invention, where, for example, the synthetic protein is isolated from the reagents and by products of the synthesis reaction(s). 15 The term "pharmaceutical grade," as used herein, when referring to a peptide or compound of the invention, is a peptide or compound that is isolated substantially or essentially from components which normally accompany the material as it is found in its natural state, including synthesis reagents and by-products, and substantially or essentially isolated from components that would impair its use as a pharmaceutical. For example, a 20 "pharmaceutical grade" peptide may be isolated from any carcinogen. In some instances, "pharmaceutical grade" may be modified by the intended method of administration, such as "intravenous pharmaceutical grade," in order to specify a peptide or compound that is substantially or essentially isolated from any substance that would render the composition unsuitable for intravenous administration to a patient. For example, an "intravenous 25 pharmaceutical grade" peptide may be isolated from detergents, such as S.DS, and anti bacterial agents, such as azide. The terms "isolated," "purified" or "biologically pure" refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state. Thus, isolated peptides in accordance with the invention 30 preferably do not contain materials normally associated with the peptides in their in situ environment. An "isolated" region of a polypeptide refers to a region that does not include the whole sequence of the polypeptide from which the region was derived. An "isolated" -9- WO 2008/033987 PCT/US2007/078371 nucleic acid, protein, or respective fragment thereof has been substantially removed from its in vivo environment so that it may be manipulated by the skilled artisan, such as but not limited to, nucleotide sequencing, restriction digestion, site-directed mutagenesis, and subeloning into expression vectors for a nucleic acid fragment as well as obtaining the protein 5 or protein fragment in substantially pure quantities. The term "variant" as used herein with respect to a peptide, refers to amino acid sequence variants which may have amino acids replaced, deleted, or inserted as compared to the wild-type polypeptide. Variants may be truncations of the wild-type peptide. A "deletion" is the removal of one or more amino acids from within the polypeptide, which a 10 "truncation" is the removal of one or more amino acids from one or both ends of the polypeptide. Thus, a variant peptide may be made by manipulation of genes encoding the polypeptide. A variant may be made by altering the basic composition or characteristics of the polypeptide, but not at least some of its pharmacologic activities. For example, a "variant" of azurin can be a mutated azurin that retains its ability to inhibit the development 15 of premalignant mammalian cells. In some cases, a variant peptide is synthesized with non natural amino acids, such as c-(3,5-dinitrobenzoyl)-Lys residues. Ghadiri & Fernholz. J. Am. Chem. Soc., 112:9633-9635 (1990). In some embodiments, the variant has not more than 20 amino acids replaced, deleted or inserted compared to wild-type peptide or part thereof In some embodiments, the variant has not more than 15 amino acids replaced, deleted or 20 inserted compared to wild-type peptide or part thereof. In some embodiments, the variant has not more than 10 amino acids replaced, deleted or inserted compared to wild-type peptide or part thereof In some embodiments, the variant has not more than 6 amino acids replaced, deleted or inserted compared to wild-type peptide or part thereof. In some embodiments, the variant has not more than 5 amino acids replaced, deleted or inserted compared to wild-type 25 peptide or part thereof. In some embodiments, the variant has not more than 3 amino acids replaced, deleted or inserted compared to wild-type peptide or part thereof The term "amino acid," as used erin, means an amino acid moiety that comprises any naturally-occurring or non-naturally occurring or synthetic amino acid residue, i.e., any moiety comprising at least one carboxyl and at least one amino residue directly linked by one. 30 two three or more carbon atoms. typically one (a) carbon atom. The term "derivative" as used herein with respect to a peptide refers to a peptide that is derived from the subject peptide. A derivation includes chemical modifications of the -10- WO 2008/033987 PCT/US2007/078371 peptide such that the peptide still retains some of its fundamental activities. For example. a "derivative" of azurin can, for example, be a chemically modified azurin that retains its ability to inhibit angiogenesis in mammalian cells. Chemical modifications of interest include, but are not limited to, amidation, acetylation, sulfation, polyethylene glycol (PEG) 5 modification, phosphorylation or glycosylation of the peptide. In addition, a derivative peptide may be a fusion of a polypeptide or fragment thereof to a chemical compound, such as but not limited to, another peptide, drug molecule or other therapeutic or pharmaceutical agent or a detectable probe. The term "percent (%) amino acid sequence identity" is defined as the percentage of 10 amino acid residues in a polypeptide that are identical with amino acid residues in a candidate sequence when the two sequences are aligned. To determine % amino acid identity, sequences are aligned and if necessary, gaps are introduced to achieve the maximum % sequence identity; conservative substitutions are not considered as part of the sequence identity. Amino acid sequence alignment procedures to determine percent identity are well 15 known to those of skill in the art. Often publicly available computer software such as BLAST, BLAST2, ALIGN2 or Megalign (DNASTAR) software is used to align peptide sequences. In a specific embodiment, Blastp (available from the National Center for Biotechnology Information, Bethesda MD) is used using the default parameters of long complexity filter, expect 10, word size 3, existence 11 and extension 1. 20 When amino acid sequences are aligned, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) can be calculated as: 25 % amino acid sequence identity = X/Y*100 where X is the number of amino acid residues scored as identical matches by the sequence alignment program's or algorithm's alignment of A and B and Y is the total number of amino acid residues in B. 30 If the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acd sequence identity of A to B will not equal the % amino acid sequence identity of B to A. When comparing Ionger sequene s to shorter sequences, the -11- WO 2008/033987 PCT/US2007/078371 shorter sequence will be the "B" sequence. For example, when comparing truncated peptides to the corresponding wild-type polypeptide, the truncated peptide will be the "B" sequence. General 5 The present invention provides compositions comprising cupredoxin, and variants, derivatives and structural equivalents of cupredoxins, and methods to prevent the development of cancer in mammals. The invention also provides to variants, derivatives and structural equivalents of cupredoxin that retain the ability to prevent the development of cancer or the re-occurrence of cancer in mammals. Most particularly, the invention provides 10 compositions comprising Pseudomonas aeruginosa azurin, variants, derivatives and structural equivalents of azurin, and their use to treat patients, and particularly patients at a higher risk of developing cancer than the general population. Finally, the invention provides methods to study the development of cancer in mammalian cells, tissues and animals by contacting the cells with a cupredoxin, or variant, derivative or structural equivalent thereof, 15 before or after inducing premalignant lesions, and observing the development of premalignant and/or malignant cells. Previously, it was know that a redox protein elaborated by Pseudomonas aerugisnosa, the cupredoxin azurin, selectively enters J774 lung cancer cells but not normal cells, and induces apoptosis. Zaborina et al., Microbiology 146:2521-2530 (2000). Azurin can also 20 selectively enter and kill human melanoma UISO-Mel-2 or human breast cancer MCF-7 cells. Yamada et al., PNAS 99:14098-14103 (2002); Punj et al., Oncogene 23:2367-2378 (2004). Azurin from P. aeruginosa preferentially enters J774 murine reticulum cell sarcoma cells, forms a complex with and stabilizes the tumor suppressor protein p53, enhances the intracellular concentration of p53, and induces apoptosis. Yamada et al, Infection and 25 Immunity 70:7054-7062 (2002). Detailed studies of various domains of the azurin molecule showed that amino acids 50-77 (p28) (SEQ ID NO: 2) represented a protein transduction domain (PTD) critical for internalization and subsequent apoptotic activity. Yamada et al, Cell. Microbial. 7:1418-1431 (2005). It is now known that azurin, and peptides derived from azurin. such as p28, have 30 chemopreventive properties. It is now known that azuin, and p28, prevent to formation of premalignant preneoplastic lesions in mouse mammary gland organ culture. in a mouse mammary gland organ culture modelWi azurin at 50 g/m was foundu to ihbtthe formation -12- WO 2008/033987 PCT/US2007/078371 of alveolar lesions by 67%. Likewise. p28 at 25 pg'ml was found to inhibit the formation of alveolar lesions by 67%. See Example 1. Further, azurin at 50 pg/ml was found to inhibit the formation of ductal lesions by 79%. and p28 at 25 pg/ml inhibited the formation of ductal lesions by 71%. See Example 1. Confocal microscopy and FAC showed that azurin and p28 5 entered normal murine mammary epithelial cells (MM3MG) and mammary cancer cells (4Tl). P28 also entered human umbilical vein endothelial cells (HUVEC) in a temperature, time and concentration dependent manner and inhibited capillary tube formation of HUVEC plated on Matrigel in a dose dependent manner. It is therefore now known that azurin and variants of azurin may be used to inhibit the formation of premalignant preneoplastic lesions, 10 and thus the development of cancer, and specifically breast cancer, in mammalian patients. Due to the high degree of structural similarity between cupredoxins, it is likely that other cupredoxins will inhibit the formation of premalignant lesions in mammals as well as azurin. Such cupredoxins may be found in, for example, bacteria or plants. Several cupredoxins are known to have pharmacokinetic activities similar to those of azurin from 15 Pseudomonas aeruginosa. For example, rusticyanin from Thiobacillusferrooxidans can also enter macrophages and induce apoptosis. Yamada et al., Cell Cycle 3:1182-1187 (2004); Yamada et al., Cell. Micro. 7:1418-1431 (2005). Plastocyanin from Phormidium laminosum and pseudoazurin form Achrornobacter cycloclastes also are cytotoxic towards macrophages. U.S. Pat. Pub. No. 20060040269, published Feb. 23, 2006. It is therefore contemplated that 20 other cupredoxins may be used in the compositions and methods of the invention. Further, variants, derivatives, and structural equivalents of cupredoxins that retain the ability to inhibit the formation of cancer in mammals may also be used in the compositions and methods of the invention. These variants and derivatives may include, but are not limited to, truncations of a cupredoxin, conservative substitutions of amino acids and proteins modifications such as 25 PEGylation and all-hydrocarbon stabling of a-he] ices. Compositions of the Invention The invention provides for peptides that are variants, derivatives or structural equivalents of cupredoxin that inhibit the development of premalignant lesions in mammalian 30 cells, tissues and animals. The invention further provides for peptides that are variants, derivatives or structural equivalents of cupredoxin that inhibit the development of cancer in rmam malian cells, tissues and animals. In some embodiments, the peptide is isolated, In -13- WO 2008/033987 PCT/US2007/078371 some embodiments, the peptide is substantially pure or pharmaceutical grade. In other embodiments, the peptide is in a composition that comprises, or consists essentially of, the peptide. In another specific embodiment, the peptide is non-antigenic and does not raise an immune response in a mammal, and more specifically a human. In some embodiments, the 5 peptide is less that a full-length cupredoxin, and retains some of the pharmacologic activities of the cupredoxins. Specifically, in some embodiments, the peptide may retain the ability to inhibit the development of premalignant lesions in the mouse mammary gland organ culture. The invention also provides compositions comprising at least one peptide that is a cupredoxin, or variant, derivative or structural equivalent of a cupredoxin, specifically in a 10 pharmaceutical composition. In specific embodiments, the pharmaceutical composition is designed for a particular mode of administration, for example, but not limited to, oral, intraperitoneal, or intravenous. Such compositions may be hydrated in water, or may be dried (such as by lyophilization) for later hydration. Such compositions may be in solvents other than water, such as but not limited to, alcohol. 15 Because of the high structural homology between the cupredoxins, it is contemplated that cupredoxins will have the same chernopreventive properties as azurin and p28. In some embodiments, the cupredoxin is, but is not limited to, azurin, pseudoazurin, plastocyanin, rusticyanin, auracyanin, stellacyanin, cucumber basic protein or Laz. In particularly specific embodiments, the azurin is derived from Pseudomonas aeruginosa, AlcaligenesJaecalis, 20 Achromobacter xylosoxidans ssp.denitrificans I Bordetella bronchiseptica, Methylomonas sp., Neisseria meningitidis, Neisseria gonorrhea, Pseudomonas fluorescens, Pseudomonas chlororaphis, Xylellafastidiosa, U/va pertussis or Vibrio parahaenolyticus. In a very specific embodiment, the azurin is from Pseudomonas aeruginosa. In other specific embodiments, the cupredoxin comprises an amino acid sequence that is SEQ ID NO: 1, 3-19. 25 The invention provides peptides that are amino acid sequence variants which have amino acids replaced, deleted, or inserted as compared to the wild-type cupredoxin. Variants of the invention may be truncations of the wild-type cupredoxin. In some embodiments, the peptide of the invention comprises a region of a cupredoxin that is less that the full length wild-type polypeptide. In some embodiments, the peptide of the invention comprises more 30 than about 10 residues. more than about 15 residues or more than about 20 residues of a truncated cupredoxin. In sonic embodiments, the peptid comprises not more than about 100 residues, not more than about 50 residues, not more than about 40 residues, not more than -14- WO 2008/033987 PCT/US2007/078371 about 30 residues or not more than about 20 residues of a truncated cupredoxin. In some embodiments, a cupredoxin has to the peptide, and more specifically SEQ ID NOS: 1, 3-19 as to the peptide of the invention, at least about 70% amino acid sequence identity, at least about 80% amino acid sequence identity, at least about 90% amino acid sequence identity, at least 5 about 95% amino acid sequence identity or at least about 99% amino acid sequence identity. In specific embodiments, the variant of cupredoxin comprises P. aeruginosa azurin residues 50-77 (p28, SEQ ID NO: 2), azurin residues 50-67, or azurin residues 36-88. In other embodiments, the variant of cupredoxin consists of P aeruginosa azurin residues 50 77, azurin residues 50-67, or azurin residues 36-88. In other specific embodiments, the 10 variant consists of the equivalent residues of a cupredoxin other that azurin. It is also contemplated that other cupredoxin variants can be designed that have a similar pharmcologic activity to azurin residues 50-77, or azurin residues 36-88. To do this, the subject cupredoxin amino acid sequence will be aligned to the Pseudomonas aeruginosa azurin sequence using BLAST, BLAST2, ALIGN2 or Megalign (DNASTAR), the relevant residues located on the 15 P. aeruginosa azurin amino acid sequence, and the equivalent residues found on the subject cupredoxin sequence, and the equivalent peptide thus designed. In one embodiment of the invention, the cupredoxin variant contains at least amino acids 57 to 89 of auracyanin B of Chloroflexus aurantiacus (SEQ ID NO: 20). In another embodiment of the invention, the cupredoxin variant contains at least amino acids 51-77 of 20 Pseudomonas syringac azurin (SEQ ID NO: 21). In another embodiment of the invention, the cupredoxin variant contains at least amino acids 89-115 of Neisseria meningitidis Laz (SEQ ID NO: 22). In another embodiment of the invention, the cupredoxin variant contains at least amino acids 52-78 of Vbrio parahaemolyticus azurin (SEQ ID NO: 23). In another embodiment of the invention, the cupredoxin variant contains at least amino acids 51-77 of 25 Bordeella bronchiseptica azurin (SEQ ID NO: 24), The variants may also include peptides made with synthetic amino acids not naturally occurring. For example, non-naturally occurring amino acids may be integrated into the variant peptide to extend or optimize the half-life of the composition in the bloodstream. Such variants include, but are not limited to, D,L-peptides (diastereomer). (for example 30 Futaki et al., J. Biol. Chem. 276(8):5836-40 (2001); Papo et l, Cancer Res. 64(16):5779-86 (2004)- Miller et a! Bioche. Pharrnacol. 36(1):16976, (1987).; peptides containing unusual amino acids (fir example Lee ei a,, J. Pept. Res. 63(2):69-84 (2004)) olefincontainig non -15- WO 2008/033987 PCT/US2007/078371 natural amino acid followed by hydrocarbon stapling Qjr example Schafmeister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Walenski et al., Science 305:1466-1470 (2004)), and peptides comprising E43,5-dinitrobenzoyl)-Lys residues. In other embodiments, the peptide of the invention is a derivative of a cupredoxin. 5 The derivatives of cupredoxin are chemical modifications of the peptide such that the peptide still retains some of its fundamental activities. For example, a "derivative" of azurin can be a chemically modified azurin that retains its ability to inhibit the development of premalignant lesions in mammalian cells, tissues or animals. Chemical modifications of interest include., but are not limited to, hydrocarbon stabling, amidation, acetylation, sulfation, polyethylene 10 glycol (PEG) modification, phosphorylation and glycosylation of the peptide. In addition, a derivative peptide maybe a fusion of a cupredoxin, or variant, derivative or structural equivalent thereof to a chemical compound, such as but not limited to, another peptide, drug molecule or other therapeutic or pharmaceutical agent or a detectable probe. Derivatives of interest include chemical modifications by which the half-life in the bloodstream of the 15 peptides and compositions of the invention can be extended or optimized, such as by several methods well known to those in the art, including but not limited to, circularized peptides (16r example Monk et al., BioDrugs 19(4):261-78, (2005); DeFreest et al., J. Pept. Res. 63(5):409-19 (2004)), N- and C- terminal modifications (for example Labrie et al., Clin. Invest. Med. 13(5):275-8, (1990)), and olefin-containing non-natural amino acid followed by 20 hydrocarbon stapling (for example Schafmreister c aL, J. Am. Chem. Soc. 122:5891-5892 (2000); Walenski et at, Science 305:1466-1470 (2004)). In another embodiment, the peptide is a structural equivalent of a cupredoxin. Examples of studies that determine significant structural homology between cupredoxins and other proteins include Toth et al. (Developmental Cell 1:82-92 (2001)). Specifically, 25 significant structural homology between a cupredoxin and the structural equivalent may be determined by using the VAST algorithm. Gibrat et al- Curr Opin Struct Biol 6:37 7-385 (1996); Madej et al, Proteins 23:356-3690 (1995). In specific embodiments, the VAST p value from a structural comparison of a cupredoxin to the structural equivalent may be less than about 10 less than about 10-, or less than about 10. In other embodiments, 30 significant structural homology between a cupredoxin and the structural equivalent may be determined by usig the DALI algorithm. Ho m & Sande, J. Mol. Bio. 233:123-138 (1993). -16- WO 2008/033987 PCT/US2007/078371 In specific embodiments, the DALI Z score for a pairwise structural comparison is at least about 3.5. at least about 7.0, or at least about 10.0. It is contemplated that the peptides of the composition of invention may be more than one of a variant, derivative and/or structural equivalent of a cupredoxin. For example. the 5 peptides may be a truncation of azurin that has been PEGylated, thus making it both a variant and a derivative. In one embodiment, the peptides of the invention are synthesized with a,a disubstituted non-natural amino acids containing olefin-bearing tethers, followed by an all hydrocarbon "staple" by ruthenium catalyzed olefin metathesis. Scharmeister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Walensky et al., Science 305:1466-1470 (2004). 10 Additionally, peptides that are structural equivalents of azurin may be fused to other peptides, thus making a peptide that is both a structural equivalent and a derivative. These examples are merely to illustrate and not to limit the invention. Variants, derivatives or structural equivalents of cupredoxin may or may not bind copper. In some embodiments, the cupredoxin, or variant, derivative or structural equivalent 15 thereof has some of the pharmacologic activities of the P. aeruginosa azurin, and specifically p28. In a specific embodiment, the cupredoxins and variants, derivatives and structural equivalents of cupredoxins that may inhibit prevent the development of premalignant lesions in mammalian cells, tissues or animals, and specifically but not limited to, mammary gland cells. The invention also provides for the cupredoxins and variants, derivatives and structural 20 equivalents of cupredoxins that may have the ability to inhibit the development of mammalian premalignant lesions, and specifically but not limited to, melanoma, breast, pancreas, glioblastoma, astrocytoma, lung, colorectal, neck and head, bladder, prostate, skin and cervical cancer cells. Inhibition of the development of cancer cells is any decrease, or lessening of the rate of increase, of the development of premalignant lesions that is 25 statistically significant as compared to control treatments, Because it is now known that cupredoxins can inhibit the development of premalignant lesions and ultimately cancer in mammalian cells, tissues or animals, and specifically breast cells, and more specifically. mouse mammary gland cells, it is now possible to design variants and derivatives of cupredoxins that retain this chemopreventive 30 activity. Such variants, derivatives and structural equivalents can be made by, for example, creating a "library" of various variants, derivatives and structural equivalents of cupredoxins and cupredoxin derived peptides and te testinge each for chemoprevenftiv activity, andI -17- WO 2008/033987 PCT/US2007/078371 specifically chemopreventive activity in the mouse mammary gland organ culture using one of many methods known in the art, such the exemplary method in Example 1. It is contemplated that the resulting variants, derivatives and structural equivalents of cupredoxins with chemopreventive activity may be used in the methods of the invention, in place of or in 5 addition to azurin or p 2 8. In some specific embodiments, the variant, derivative or structural equivalent of cupredoxin may inhibit the development of 7,12-dimethylbenz (a) anthracene (DMBA) induced premalignant lesions in a mouse mammary gland organ culture (MMOC) to a degree that is statistically different from a non-treated control. A peptide can be tested for this 10 activity by using the MMOC model system is described in Example 1, or as in Mehta et aL (J Natl Cancer Inst 93:1103-1106 (2001)) and Mehta et al. (Meth Cell Sci 19:19-24 (1997)). Other methods to determine whether cancer development is inhibited another are well known in the art and may be used as well. In some specific embodiments, the variant, derivative or structural equivalent of 15 cupredoxin inhibits the development of mammary alveolar lesions (MAL) in the a MMOC model to a degree that is statistically different from a non-treated control. In some specific embodiments, the variant, derivative or structural equivalent of cupredoxin inhibits the development of mammary ductal lesions (MDL) in the a MMOC model to a degree that is statistically different from a non-treated control. A peptide can be tested for these activities 20 by using the MMOC model system induced to form premalignant lesions by DMBA, as described in Example 1. Evaluation of development of premalignant lesions in a MMOC model system may be determined by morphometic analysis, or histopathological analysis, as provided in Example 1. 25 Cupredoxins These small blue copper proteins (cupredoxins) are electron transfer proteins (10-20 kDa) that participate in bacterial electron transfer chains or are of unknown function. The copper ion is solely bound by the protein matrix. A special distorted trigonal planar arrangement to two histidine and one cysteine ligands around the copper gives rise to very 30 peculiar electronic properties of the metal site and an intense blue color. A number of cupredoxins have been crystallographicaily characterized at medium to high resolution -18s- WO 2008/033987 PCT/US2007/078371 The cupredoxins in general have a low sequence homology but high structural homology. Gough & Clothia, Structure 12:917-925 (2004); De Rienzo et al, Protein Science 9:1439-1454 (2000). For example, the amino acid sequence of azurin is 31% identical to that of auracyanin B, 16.3% to that of rusticyanin, 20.3 % to that of plastocyanin, and 17.3% to 5 that of pseudoazurin. See, Table I. However, the structural similarity of these proteins is more pronounced. The VAST p value for the comparison of the structure of azurin to auracyanin B is 104, azurin to rustievanin is 10-, azurin to plastocyanin is 10,' and azurin to psuedoazurin is 104. All of the cupredoxins possess an eight-stranded Greek key beta-barrel or beta 10 sandwich fold and have a highly conserved site architecture. De Rienzo et al, Protein Science 9:1439-1454 (2000). A prominent hydrophobic patch, due to the presence of many long chain aliphatic residues such as methionines and leucines, is present around the copper site in azurins, amicyanins, cyanobacterial plastocyanins, cucumber basic protein and to a lesser extent, pseudoazurin and eukaryotic plastocyanins. Id. Hydrophobic patches are also 15 found to a lesser extent in stellacyanin and rusticyanin copper sites, but have different features. Id. Table 1. Sequence and structure alignment of azurin (1JZG) from P. acruginosa to other proteins using VAST algorithm. PDB Alignment % aa P-value2 Score3 RMSD Description length identi iAOZ A 2 82 18.3 10 e-7 12.2 1.9 Ascorbate oxidase IQHQ_A 113 31 1Oe-7.4 12.1 1.9 AuracyaninB IV54 B 1 79 20.3 1Oe-6.0 11.2 2.1 Cytocrome c oxidase 1GY2 A 92 16.3 1Oe-5.0 11.1 1.8 Rusticyanin 3MSP A 74 8.1 lOe-6.7 10.9 2.5 Motile Major Sperm Protein 5 IlUZ 74 20.3 10e-5.6 10.3 2.3 Plastoevanin IKGY E 90 5.6 10e-4,6 10.1 3.4 Ephrinb2 IPMY 75 17.3 IOe-4.1 9.8 2.3 Pseudoazurin 20 Aligned Length: The number of equivalent pairs of C-alpha atoms superimposed between the two structures, i.e. how many residues have been used to calculate the 3D superposition. P-VAL: The VAST p value is a measure of the significance of the comparison, 25 expressed as a probahility. For example, if the p value is 0.001, then the odds are 1000 to 1 -19- WO 2008/033987 PCT/US2007/078371 against seeing a match of this quality by pure chance. The p value from VAST is adjusted for the effects of multiple comparisons using the assumption that there are 500 independent and unrelated types of domains in the MMDB database. The p value shown thus corresponds to the p value for the pairwise comparison of each domain pair, divided by 500. 5 3 Score: The VAST structure-similarity score. This number is related to the number of secondary structure elements superimposed and the quality of that superposition. Higher VAST scores correlate with higher similarity. 4 RMSD: The root mean square superposition residual in Angstroms. This number is calculated after optimal superposition of two structures, as the square root of the mean square 10 distances between equivalent C-alpha atoms. Note that the RMSD value scales with the extent of the structural alignments and that this size must be taken into consideration when using RMSD as a descriptor of overall structural similarity. C elegans major sperm protein proved to be an ephrin antagonist in oocyte maturation. Kuwabara, Genes and Development 17:155-161 (2003). 15 Azurin The azurins are copper containing proteins of 128 amino acid residues which belong to the family of cupredoxins involved in electron transfer in certain bacteria. The azurins include those from P. aeruginosa (PA) (SEQ ID NO: I), A, xylosoxidans, and A. 20 denitrificans. Murphy et al, J. Mol. Biol. 315:859-871 (2002). The amino acid sequence identity between the azurins varies between 60-90%, these proteins showed a strong structural homology. All azurins have a characteristic p-sandwich with Greek key motif and the single copper atom is always placed at the same region of the protein. In addition, azurins possess an essentially neutral hydrophobic patch surrounding the copper site. Id. Plastocyanins The plastocyanins are soluble proteins of cyanobacteria, algae and plants that contain one molecule of copper per molecule and are blue in their oxidized form. They occur in the chloroplast, where they function as electron carriers. Since the determination of the structure 30 of poplar plastocyanin in 1978, the structure of algal (Scenedesmus, Enteromorpha. Chlamydomonas) and plant (French bean) plastocyanins has been determined either by erystallographic or NMR methods, and the poplar structure has been refined to 1.33 A resolution. SE I)D NO: 3 shows the amino acid sequnc of platocyanin from Phormidium -20- WO 2008/033987 PCT/US2007/078371 laminosum. a thermophilic cyanobacterium. Another plastocyanin of interest is from Ulva pertussis. Despite the sequence divergence among plastocyanins of algae and vascular plants (e.g, 62% sequence identity between the Chlamydomonas and poplar proteins), the three 5 dimensional structures are conserved (e.g., 0.76 A nns deviation in the C alpha positions between the Chlamvdomonas and Poplar proteins). Structural features include a distorted tetrahedral copper binding site at one end of an eight-stranded antiparallel beta-barrel, a pronounced negative patch, and a flat hydrophobic surface. The copper site is optimized for its electron transfer function, and the negative and hydrophobic patches are proposed to be 10 involved in recognition of physiological reaction partners. Chemical modification, cross linking, and site-directed mutagenesis experiments have confirmed the importance of the negative and hydrophobic patches in binding interactions with cytochrome f , and validated the model of two functionally significant electron transfer paths involving plastocyanin. One putative electron transfer path is relatively short (approximately 4 A) and involves the 15 solvent-exposed copper ligand His-87 in the hydrophobic patch, while the other is more lengthy (approximately 12-15 A) and involves the nearly conserved residue Tyr-83 in the negative patch. Redinbo ct aL, J. Bioenerg. Biomembr. 26:49-66 (1994). Rusticyanins 20 Rusticyanins are blue-copper containing single-chain polypeptides obtained from a Thiobacillus (now called Acidithiobacillus). The X-ray crystal structure of the oxidized form of the extremely stable and highly oxidizing cupredoxin rusticyanin from Thiobacillus ferrooxidans (SEQ ID NO: 4) has been determined by multiwavelength anomalous diffraction and refined to 1 .9A resolution. The rusticyanins are composed of a core beta 25 sandwich fold composed of a six- and a seven-stranded b-sheet. Like other cupredoxins., the copper ion is coordinated by a cluster of four conserved residues (His 85, Cys138, Hisl43, Metl48) arranged in a distorted tetrahedron. Walter, R.L e al, J Mol. Biol. 263:730-51 (1996). 30 Pseudoazurins The pseudoazurins are a family of blue-copper containing single-chain polypeptide. The amno acid sequence of pseudoazurin obtained from Achromobcter cyclolasies is shown in SEQ ID NO: 5. The X-rav structure analysis of pseudoazurin shows that it has a -21- WO 2008/033987 PCT/US2007/078371 similar structure to the azurins although there is low sequence homology between these proteins. Two main differences exist between the overall structure of the pseudoazurins and azurins. There is a carboxy terminus extension in the pseudoazurins. relative to the azurins, consisting of two alpha-helices. In the mid-peptide region azurins contain an extended loop, 5 shortened in the pseudoazurins, which forns a flap containing a short a-helix. The only major differences at the copper atom site are the conformation of the MET side-chain and the Met-S copper bond length, which is significantly shorter in pseudoazurin than in azurin. Phytocyanins 10 The proteins identifiable as phytocyanins include, but are not limited to, cucumber basic protein, stellacyanin, mavicyanin, umecyanin, a cucumber peeling cupredoxin, a putative blue copper protein in pea pods, and a blue copper protein from Arabidopsis thaliana. In all except cucumber basic protein and the pea-pod protein, the axial methionine ligand normally found at blue copper sites is replaced by glutamine. 15 Auracyanin Three small blue copper proteins designated auracyanin A, auracyanin B-1, and auracyanin B-2 have been isolated from the thermophilic green gliding photosynthetic bacterium Chloroflexus aurantiacus. The two B forms are glycoproteins and have almost 20 identical properties to each other, but are distinct from the A form. The sodium dodecyl sulfate-polyacrylamide gel electrophoresis demonstrates apparent monomer molecular masses as 14 (A), 18 (B-2), and 22 (B-1) kDa. The amino acid sequence of auracyanin A has been determined and showed auracyanin A to be a polypeptide of 139 residues. Van Dreissche et al- Protein Science 25 8:947-957 (1999). His58, Cysl23, Hisl28, and Met132 are spaced in a way to be expected if they are the evolutionary conserved metal ligands as in the known small copper proteins plastocyanin and azurin. Secondary structure prediction also indicates that auracyanin has a general beta-barrel structure similar to that of azurin from Pseudononas aeruginosa and plastocyanin from poplar leaves. However, auracyanin appears to have sequence 30 characteristics of both small copper protein sequence classes. The overall similarity with a consensus sequence of azurin is roughly the same as that with a consensus sequence of piastoyanin, namely 305%. The N-terninal sequence region 1-18 of auracvain is WO 2008/033987 PCT/US2007/078371 remarkably rich in glycine and hydroxy amino acids. Id. See exemplary amino acid sequence SEQ ID NO: 15 for chain A of auracyanin from Chloroflexus aurantiacus (NCBI Protein Data Bank Accession No. AAM12874), The auracyanin B molecule has a standard cupredoxin fold. The crystal structure of 5 auracyanin B from Chloroflexus aurantiacus has been studied. Bond et al., J. Mol. Biol. 306:47-67 (2001). With the exception of an additional N-terminal strand, the molecule is very similar to that of the bacterial cupredoxin, azurin. As in other cupredoxins, one of the Cu ligands lies on strand 4 of the polypeptide, and the other three lie along a large loop between strands 7 and 8. The Cu site geometry is discussed with reference to the amino acid 10 spacing between the latter three ligands. The crystallographically characterized Cu-binding domain of auracyanin B is probably tethered to the periplasmic side of the cytoplasmic membrane by an N-terminal tail that exhibits significant sequence identity with known tethers in several other membrane-associated electron-transfer proteins. The amino acid sequences of the B forms are presented in McManus et al. J. Biol. Chem. 267:6531-6540 (1992). See 15 exemplary amino acid sequence SEQ ID NO: 16 for chain B of auracyanin from Chloroflexus aurantiacus (NCBI Protein Data Bank Accession No. IQHQA). Stellacyanin Stellacyanins are a subclass of phytocyanins, a ubiquitous family of plant 20 cupredoxins. An exemplary sequence of a stellacyanin is included herein as SEQ ID NO: 14. The crystal structure of umecyanin, a stellacyanin from horseradish root (Koch et al, J. Am. Chem. Soc. 127:158-166 (2005)) and cucumber stellacyanin (Hart el al, Protein Science 5:2175-2183 (1996)) is also known. The protein has an overall fold similar to the other phytocyanins. The ephrin B2 protein ectodomain tertiary structure bears a significant 25 similarity to stellacyanin. Toth et al, DeveL opmental Cell 1:83-92 (2001). An exemplary amino acid sequence of a stellacyanin is found in the National Center for Biotechnology Information Protein Data Bank as Accession No. I JER, SEQ ID NO: 14. Cucumber basic protein 30 An exemplary amino acid sequence from a cucumber basic protein is included herein as SEQ ID NO: 17. The crystal structure of the cucunmer hasic protein (CBP), a type I blue copper protein, has been refined at -. 8 A resolution. The molecule resembles other blue ropper proteins in having a Creek key beta-barrei structure, except that the barrel is open on -23- WO 2008/033987 PCT/US2007/078371 one side and is better described as a "beta-sandwich" or "beta-taco". Guss et al. I Mol. Biol. 262:686-705 (1996). The ephrinB2 protein ectodomian tertiary structure bears a high similarity (rms deviation 1.5k for the 50 a carbons) to the cucumber basic protein. Toth et al, Developmental Cell 1:83-92 (2001). 5 The Cu atom has the normal blue copper NNSS' co-ordination with bond lengths Cu N(His39) 1.93 A, Cu-S(Cys79)= 2.16 A, Cu-N(His84) = 1.95 A, Cu-S(Met89)= 2.61 A. A disulphide link, (Cys52)-S-S-(Cys85). appears to play an important role in stabilizing the molecular structure. The polypeptide fold is typical of a sub-family of blue copper proteins (phytocyanins) as well as a non-metalloprotein, ragweed allergen Ra3, with which CBP has a 10 high degree of sequence identity. The proteins currently identifiable as phytocyanins are CBP, stellacyanin, mavicyanin, umecyanin, a cucumber peeling cupredoxin, a putative blue copper protein in pea pods, and a blue copper protein from Arabidopsis thaliana. In all except CBP and the pea-pod protein, the axial methionine ligand nonnally found at blue copper sites is replaced by glutamine. An exemplary sequence for cucumber basic protein is found in 15 NCBI Protein Data Bank Accession No. 2CBP, SEQ ID NO: 17. Methods of Use The invention provides methods to prevent de novo malignancies in otherwise healthy patients comprising administering to the patient at least one peptide that is a cupredoxin, or 20 variant, derivative or structural equivalent thereof, as described above. Chemopreventive therapies are based on the hypothesis that the interruption of processes involved in cancergenesis will prevent the development of cancer. The cupredoxin Pseudomonas acruginosa azurin and the truncated azurin peptide p28 are now known to inhibit the development of premalignant lesions, either by inhibiting the initial formation of 25 premalignant lesions, or killing or inhibiting the growth of premalignant lesions that are present. It therefore contemplated that a cupredoxin, or variant, derivative or structural equivalent thereof, as described above, with the ability to inhibit the development of premalignant lesions, may be used in chemopreventive therapies in otherwise healthy patients. Such otherwise healthy patients are, in some embodiments, patients at a higher risk 30 to develop cancer than those in the general population. Cancers that may be prevented by treatment with the compositions of the invention include, but are not limited to, melanoma, breast, pancreas, glioblastoma, astrocytoma, lung, colorectal, neck and head, bladder, -24- WO 2008/033987 PCT/US2007/078371 prostate, skin, and cervical cancer. In some embodiments, the patient may be human. In other embodiments, the patient is not human. The invention further includes methods to study the development of cancer comprising contacting mammalian cells before or after induction with a carcinogen with a 5 composition comprising cupredoxin, or a variant, derivative or structural equivalent thereof and observing the development of the cells. In some embodiments, the cells are mouse mammary gland cells, while in others they are other cells that may become malignant in mammals. Patients at a higher at risk to develop cancer than the general population may be 10 patients with high risk features, patients with premalignant lesions, and patients that have been cured of their initial cancer or definitively treated for their premalignant lesions. See generally Tsao et aL, CA Cancer J Clin 54:150-180 (2004). High risk features may be behavioral, genetic, environmental or physiological factors of the patient. Behavioral factors that predispose a patient to various forms of cancer include, but are not limited to, smoking, 15 diet, alcohol consumption, hormone replacement therapy, higher body mass index, nulliparity, betal nut use, frequent mouthwash use, exposure to human papillomavirus, childhood and chronic sun exposure, early age of first intercourse, multiple sexual partners, and oral contraceptive use. Genetic factors that predispose a patient to various forms of cancer include, but are not limited to, a family history of cancer, gene carrier status of BRCA1 20 and BR CA2, prior history of breast neoplasia, familial adenomatous polyposis (FAP). hereditary nonpolyposis colorectal cancer (HNPCC), red or blond hair and fair-skinned phenotype, xeroderma pigmentosum, and ethnicity. Environmental features that predispose a patient to various forms of cancer include, but are not limited to, exposure to radon, polycyclic aromatic hydrocarbons, nickel, chromate, arsenic, asbestos, chloromethyl ethers, 25 benzo[ajpyrene, radiation, and aromatic amines from rubber or paint occupational exposure. Other miscellaneous factors that predispose a patient to various forms of cancer include, but are not limited to, chronic obstructive pulmonary disease with airflow obstruction, chronic bladder infections. schistosomiasis, older age, and immunocompromised status. Additionally. patients at a higher risk of developing cancer may be determined by the 30 use of various risk models that have been developed for certain kinds of cancer. For example, patients prdsposed to breast cancer may be determined using the Gail risk model, -25-.
WO 2008/033987 PCT/US2007/078371 orthe Claus model, among others. See Gail et al, J Natl Cancer Inst 81:1879-1886 (1989); Cuzick, Breast 12:405-411 (2003); Huang et al, Am J Epidemiol 151:703-714 (2000). Patients with premalignant lesions are at a higher risk to develop cancer than the general population. The presence of premalignant lesions in or on a patient may be 5 determined by many methods that are well known to those in the art. Intermediate markers or biomarkers that originate from premalignant lesions may be measured in a patient to determine if the patient harbors premalignant lesions. Chromosomal abnormalities occur in tumor cells and the adjacent histologicially normal tissues in the majority of cancer patients. Progression in chromosomal abnormalities parallels the phenotypic progression from 10 premalignant lesion to invasive cancer. Thiberville et al, Cancer Res. 55:5133-5139 (1995). Therefore, chromosomal abnormalities associated with cancer may be used as intermediate markers to detect premalignant lesions in a patient. Common chromosomal abnormalities associated with cancer include, but are not limited to, allelic deletions or loss of heterozygosity (LOH) in tumor suppressor genes such as 3p (FHIT and others), 9 p (9p21 for 15 pI 6 X, p15NK4B and p R9F"), ]7p (]7p13 forp53 gene and others) and 13q (13 14 for retinoblastoma gene Rb and others). Deletions in 3p and 9 p are associated with smoking and the early stages of lung cancer. Mao et al., J. NatI. Cancer Inst. 89:857-862 (1997). Deletions affecting 3p, Sq, 8p, 1 7 p and 18 q are common change in epithelial cancers. See generally Tsao et al., CA Clin. Cancer J. Clin. 54:153 (2004). Other chromosomal mutations 20 associated with cancer include those which activate oncogenes. Oncogenes whose presence may be used as intermediate markers include, but are not limited to, Ras, c-myc, epidemral growth factor. erb-B2 and cyclins E, DI and .B1. See generally id. at 154. Other intermediate markers may be the products of genes up-regulated in premalignant cells and cancer cells. Genes that may be up-regulated in premalignant cells 25 include, but are not limited to, cyclooxygenases COX-1 and COX-2, telomerase. Other biomarkers of cancer cells, and some premali gant cells, include. but are not limited to, p53, epidermal growth factor receptor (GFR), proliferating cell nuclear antigen (PCNA), RAS. COX-2, Ki-67, DNA aneuploidy, DNA polymerasc-a, ER. Her2neu, E-cadherin, RARp, hTERT p/6 , FHIT ( 3 pl4), Bel-2, VEGF-R, HPV infection, LOH 9 p21, LOH 17p, p 30 AKT, hnRNP A2/B1, RAF, Mye, c-KIT, cyclin D1, E and B1, IGF , bi-2, p16, LOH p2.3, LOH 3 p 2 5, LOH 9p21, LOH 17p13, LOH 13q, LH 8p, hMSH2, APC, DCC, DPC4, JV18, BAX, PSA, GSTP, N'F-kB, AP, D3S2, HPV infection. LO4 3,4 LOH 4 -26- WO 2008/033987 PCT/US2007/078371 LOH Sp. bladder tumor antigen (BTA), BTK TRAK (Alidex, Inc., Redmond WA), urinary tract matrix protein 22, fibrin degradation product, autodrine motility factor receptor, BCLA 4, cytokeratin 20, hyaluronic acid, CYFRA 21-1, BCA, beta-human chorionic gonadotropin., and tissue polypeptide antigen (TPA). See generally id, at 155-157. 5 Patients that have been cured of their initial cancers or have been definitively treated for their premalignant lesions are also at a higher risk to develop cancer than the general population. A second primary tumor refers to a new primary cancer in a person with a history of cancer. Second primary tumors are the leading cause of mortality in head and neck cancer. Id. at 150. A second primary tumor is distinct from a metastasis in that the former 10 originates de novo while the later originates from an existing tumor. Patients that have been cured of cancer or premalignant lesions of the breast, head and neck, lung, and skin are at a particularly high risk to develop second primary tumors. The compositions comprising a cupredoxin or variant, derivative or structural equivalent thereof can be administered to the patient by many routes and in many regimens 15 that will be well known to those in the art. In specific embodiments, the cupredoxin, or variant, derivative or structural equivalent thereof is administered intravenously, intramuscularly, subcutaneously, topically, orally, or by inhalation. The compositions may be administered to the patient by any means that delivers the peptides to the site in the patient that is at risk of developing cancer. In specific embodiments, the cupredoxin or variant, 20 derivative or structural equivalent thereof is administered intraveneously. In one embodiment, the methods may comprise co-administering to a patient one unit dose of a composition comprising a cupredoxin or a variant, derivative or structural equivalent of cupredoxin and one unit dose of a composition comprising another chemopreventive drug, in either order, administered at about the same time, or within about a 25 given time following the administration of the other, for example. about one minute to about 6o minutes following the administration of the other drug, or about I hour to about 12 hours following the administration of the other drug. Chemopreventive drugs of interest include., but are not limited to, tamoxifen. aromatase inhibitors such as letrozole and anastrozole (Arimidex*, retinoids such as N-[4-hydroxyphenyl] retinamide (4-HPR, fenretinide), 30 nonsteriodal antiinflammatorv agents (NSAIDs) such as aspirin and sulindac, celecoxib (COX-2 inhibitor), detluoromethylornthing (DFMO), ursodeoxycholic acid, 3-hydroxy-3 methylglutaryl coenzyme A reductase inhibitors, FKI-785 (ER inhibitor), hevacizumab -27 - WO 2008/033987 PCT/US2007/078371 (antibody to VEGF-receptor), cetuximab (antibody to EGFR), retinol such as vitamin A, beta-carotene, 13-cis retinoic acid, isotretinoin and retinyl palmitate, a-tocopherol, interferon, oncolytic adenovirus dl1520 (ONYX-015), gefitinib, etretinate, finasteride, indole-3-carbinol, resveratrol, chlorogenic acid, raloxifene, and oltipraz. 5; Pharmaceutical Compositions Comprising Cupredoxin, Or Variant, Derivative Or Structural Equivalent Thereof Pharmaceutical compositions comprising cupredoxin or variant, derivative or structural equivalents thereof, can be manufactured in any conventional manner, e.g., by conventional mixing, dissolving, granulating, dragee-making, emulsifying, encapsulating, entrapping, or 10 lyophilizing processes. The substantially pure or pharmaceutical grade cupredoxin or variants, derivatives and structural equivalents thereof can be readily combined with a pharmaceutically acceptable carrier well-known in the art. Such carriers enable the preparation to be formulated as a tablet, pill, dragee, capsule, liquid, gel, syrup, slurry, suspension, and the like. Suitable carriers or excipients can also include, for example, fillers 15 and cellulose preparations. Other excipients can include, for example, flavoring agents, coloring agents, detackifiers, thickeners, and other acceptable additives, adjuvants, or binders. In some embodiments, the pharmaceutical preparation is substantially free of preservatives. In other embodiments, the pharmaceutical preparation may contain at least one preservative. General methodology on pharmaceutical dosage forms is found in Ansel et al, 20 Pharmaceutical Dosage Forms and Drug Delivery Systems (Lippencott Williams & Wilkins, Baltimore MD (1999)), The composition comprising a cupredoxin or variant, derivative or structural equivalent thereof used in the invention may be administered in a variety of ways, including by injection (e.g,. intradermal, subcutaneous, intramuscular, intraperitoneal and the like), by 25 inhalation, by topical administration, by suppository, by using a transdermal patch or by mouth. General information on drug delivery systems can be found in Ansel et at, id. In some embodiments, the composition comprising a cupredoxin or variant, derivative or structural equivalent thereof can be formulated and used directly as injectibles. for subcutaneous and intravenous injection, among others. The injectable formulation, in 30 particular, can advantageously be used to treat patients that are appropriate for chemopreventive therapy. The composition comprising a cupredoxin or variant, derivative or -28- WO 2008/033987 PCT/US2007/078371 structural equivalent thereof can also be taken orally after mixing with protective agents such as polypropylene glycols or similar coating agents. When administration is by injection, the cupredoxin or variant, derivative or structural equivalent thereof may be formulated in aqueous solutions, specifically in physiologically 5 compatible buffers such as Hanks solution, Ringer's solution, or physiological saline buffer. The solution may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the cupredoxin or variant, derivative or structural equivalent thereof may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen free water, before use. In some embodiments, the pharmaceutical composition does not 10 comprise an adjuvant or any other substance added to enhance the immune response stimulated by the peptide. In some embodiments, the pharmaceutical composition comprises a substance that inhibits an immune response to the peptide. When administration is by intravenous fluids, the intravenous fluids for use administering the cupredoxin or variant, derivative or structural equivalent thereof may be composed of 15 crystalloids or colloids. Crystalloids as used herein are aqueous solutions of mineral salts or other water-soluble molecules. Colloids as used herein contain larger insoluble molecules, such as gelatin. Intravenous fluids may be sterile. Crystalloid fluids that may be used for intravenous administration include but are not limited to, normal saline (a solution of sodium chloride at 0.9% concentration), Ringer's lactate or 20 Ringer's solution, and a solution of 5% dextrose in water sometimes called D5W, as described in Table 2. Table 2. Composition of Common Crystalloid Solutions Solution Other Name [Na t ] [ClI [Glucose] D5W 5% Dextrose 0 0 252 2/3 & 1/3 3.3% Dextrose 51 51 168 0.3% saline Half-normal 0.45% NaCl 77 77 0 saline Normal saline 0.9% NaCl 154 154 0 Ringer' Ringer's 130 109 0 lactate* soltion -29- WO 2008/033987 PCT/US2007/078371 *Ringer's lactate also has 28 mmol/L lactate, 4 mmol/L K and 3 mmol/L Ca2 When administration is by inhalation, the cupredoxin or variant, derivative or structural equivalent thereof may be delivered in the form of an aerosol spray from 5 pressurized packs or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin, for use in an inhaler or insufflator may be formulated containing a powder mix of the proteins and a suitable powder 10 base such as lactose or starch, When administration is by topical administration, the cupredoxin or variant, derivative or structural equivalent thereof may be formulated as solutions, gels, ointments, creams, jellies, suspensions, and the like, as are well known in the art. In some embodiments, administration is by means of a transdermal patch. When administration is by suppository 15 (e.g., rectal or vaginal), cupredoxin or variants and derivatives thereof compositions may also be formulated in compositions containing conventional suppository bases. When administration is oral, a cupredoxin or variant, derivative or structural equivalent thereof can be readily formulated by combining the cupredoxin or variant, derivative or structural equivalent thereof with pharmaceutically acceptable carriers well 20 known in the art. A solid carrier, such as mannitol, lactose, magnesium stearate, and the like may be employed; such carriers enable the cupredoxin and variants, derivatives or structural equivalent thereof to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. For oral solid formulations such as, for example, powders, capsules and tablets, suitable excipients include 25 fillers such as sugars, cellulose preparation, granulating agents, and binding agents. Other convenient carriers, as well-know n in the art, also include multiv talent carriers, such as bacterial capsular polysaccharide. a dextran or a genetically engineered vector. In addition, sustained-release formulations that include a cupredoxin or variant, derivative or structural equivalent thereof allow for the release of cupredoxin or variant, derivative or 30 structural equivalent thereof over extended periods of time, such that without the sustained release formulation, the cupredoxin or variant, derivative or structural equivalent thereof -30- WO 2008/033987 PCT/US2007/078371 would be cleared from a subject's system. and/or degraded by, for example, proteases and simple hydrolysis before eliciting or enhancing a therapeutic effect. The half-life in the bloodstream of the peptides of the invention can be extended or optimized by several methods well known to those in the art. The peptide variants of the 5 invention may include, but are not limited to, various variants that may increase their stability, specific activity, longevity in the bloodstream, and/or decrease immunogenicity of the cupredoxin, while retaining the ability of the peptide to inhibit the development of premalignant lesions in mammalian cells, tissues and animals. Such variants include, but are not limited to, those which decrease the hydrolysis of the peptide, decrease the 10 deamidation of the peptide, decrease the oxidation, decrease the immunogenicity, increase the structural stability of the peptide or increase the size of the peptide. Such peptides also include circularized peptides (see Monk et al., BioDrugs 19(4):261-78, (2005); DeFreest et al, J. Pept. Res. 63(5):409-19 (2004)), D,L-peptides (diastereomer), Futaki et al, J. Biol. Chem. Feb 23;276(8):5836-40 (2001); Papo et al., Cancer Res. 64(16):5779-86 15 (2004); Miller et al., Biochem. Pharmacol. 36(1):169-76, (1987)); peptides containing unusual amino acids (see Lee et al., J. Pept. Res. 63(2):69-84 (2004)), N- and C- terminal modifications (see Labrie et al., Clin. Invest. Med. 13(5):275-8, (1990)), hydrocarbon stapling (see Schafneister et al., J. Am. Chem. Soc. 122:5891-5892 (2000); Walenski et al., Science 305:1466-1470 (2004)) and PEGylation. 20 In various embodiments, the pharmaceutical composition includes carriers and excipients (including but not limited to buffers, carbohydrates, mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, bacteriostats, chelating agents, suspending agents, thickening agents and/or preservatives), water, oils, saline solutions, aqueous dextrose and glycerol solutions, other pharmaceutically acceptable auxiliary 25 substances as required to approxima te physioogical conditions, such as suffering agents, tonicity adjusting agents, wetting agents and the like. It will be recognized that, while any suitable carrier known to those of ordinary skill in the art may be employed to administer the compositions of this invention, the tpe of carrier will vary depending on the mode of administration. Compounds may also be encapsulated within liposomes using well-known 30 technology. Biodegradable microspheres may also, be employed as carriers for the pharmaceutical compositions of this invention. Suitable biodegradable microspheres are -31- WO 2008/033987 PCT/US2007/078371 disclosed, for example, in U.S. Patent Nos. 4.897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763; 5,814,344 and 5,942,252. The pharmaceutical compositions may be sterilized by conventional, well-known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration. Administration of Cupredoxin or Variant, Derivative or Structural Equivalent Thereof The cupredoxin or variant, derivative or structural equivalent thereof can be 10 administered formulated as pharmaceutical compositions and administered by any suitable route, for example, by oral, buccal, inhalation, sublingual, rectal, vaginal, transurethral, nasal, topical, percutaneous, i.e., transdermal or parenteral (including intravenous, intramuscular, subcutaneous and intracoronary) or vitreous administration. The pharmaceutical formulations thereof can be administered in any amount effective to achieve its intended 15 purpose. More specifically, the composition is administered in a therapeutically effective amount. In specific embodiments, the therapeutically effective amount is generally from about 0.01-20 mg/day/kg of body weight. The compounds comprising cupredoxin or variant, derivative or structural equivalent thereof are useful for the prevention of cancer, alone or in combination with other active 20 agents. The appropriate dosage will, of course, vary depending upon, for example, the compound of cupredoxin or variant, derivative or structural equivalent thereof employed, the host, the mode of administration and the nature and severity of the potential cancer. However, in general, satisfactory results in humans are indicated to be obtained at daily dosages from about 0.01-20 mg/kg of body weight. An indicated daily dosage in humans is 25 in the range from about 0.7 mg to about 1400 mg of a compound of cupredoxin or variant, derivative or structural equivalent thereof conveniently administered, for example, in daily doses, weekly doses, monthly doses, and/lor continuous dosing. Daily doses can be in discrete dosages from I to 12 times per day. Alternatively, doses can be administered every other day, every third day, every fourth day, every fifth day, every sixth day, every week, and 30 similarly in day increments up to 31 days or over. Alternatively, dosing can be continuous using patches, i.v. administration and the like. -32- WO 2008/033987 PCT/US2007/078371 The exact formulation, route of administration. and dosage is determined by the attending physician in view of the patient's condition. Dosage amount and interval can be adjusted individually to provide plasma levels of the active cupredoxin or variant, derivative or structural equivalent thereof which are sufficient to maintain therapeutic effect. Generally, 5 the desired cupredoxin or variant, derivative or structural equivalent thereof is administered in an admixture with a pharmaceutical carrier selected with regard to the intended route of administration and standard pharmaceutical practice. In one aspect, the cupredoxin or variant, derivative or structural equivalent thereof is delivered as DNA such that the polypeptide is generated in situ. In one embodiment, the 10 DNA is "naked," as described, for example, in Ulmer et aL, (Science 259:1745-1749 (1993)) and reviewed by Cohen (Science 259:1691-1692 (1993)). The uptake of naked DNA may be increased by coating the DNA onto a carrier, e.g., biodegradable beads, which are then efficiently transported into the cells. In such methods, the DNA may be present within any of a variety of delivery systems known to those of ordinary skill in the art, including nucleic 15 acid expression systems, bacterial and viral expression systems. Techniques for incorporating DNA into such expression systems are well known to those of ordinary skill in the art. See, e.g., W090/11092, W093/24640, WO 93/17706, and U.S. Pat. No. 5,736,524. Vectors, used to shuttle genetic material from organism to organism, can be divided into two general classes: Cloning vectors are replicating plasmid or phage with regions that 20 are essential for propagation in an appropriate host cell and into which foreign DNA can be inserted; the foreign DNA is replicated and propagated as if it were a component of the vector. An expression vector (such as a plasmid, yeast, or animal virus genome) is used to introduce foreign genetic material into a host cell or tissue in order to transcribe and translate the foreign DNA, such as the DNA of a cupredoxin. In expression vectors, the introduced 25 DNA is operably-linked to elements such as promoters that signal to the host cell to highly transcribe the inserted DNA, Some promoters are exceptionally useful, such as inducible promoters that control gene transcription in response to specific factors. Operably-linking a cupredoxin and variants and derivatives thereof polynucleotide to an inducible promoter can control the expression of the cupredoxin and variants and derivatives thereof in response to 30 specific factors. Examples of classic inducible promoters include those that are responsive to a-interferon, heat shock, heavy metal ions, and steroids such as lucocortieoids (Kaulfman. Methods Enzyvmot~ 185:487-511 (1990)) and tetracycline. Other desirable inducible -33- WO 2008/033987 PCT/US2007/078371 promoters include those that are not endogenous to the cells in which the construct is being introduced, but, are responsive in those cells when the induction agent is exogenously supplied. In general,. useful expression vectors are often plasmids. However, other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses 5 and adeno-associated viruses) are contemplated. Vector choice is dictated by the organism or cells being used and the desired fate of the vector. In general, vectors comprise signal sequences, origins of replication, marker genes, polylinker sites, enhancer elements, promoters, and transcription termination sequences. 10 Kits Comprising Cupredoxin, or Variant, Derivative Or Structural Equivalent Thereof In one aspect, the invention provides regimens or kits comprising one or more of the following in a package or container: (1) a pharmacologically active composition comprising at least one cupredoxin or variant, derivative or structural equivalent thereof; (2) an additional 15 chemopreventive drug, (3) apparatus to administer the biologically active composition to the patient, such as a syringe, nebulizer etc.. When a kit is supplied, the different components of the composition may be packaged in separate containers, if appropriate, and admixed immediately before use. Such packaging of the components separately may permit long-term storage without losing the active 20 components' functions. The reagents included in the kits can be supplied in containers of any sort such that the life of the different components are preserved and are not adsorbed or altered by the materials of the container. For example, sealed glass ampules may contain lyophilized cupredoxin and variants, derivatives and structural equivalents thereof, or buffers that have 25 been packaged under a neutral, non-reacting gas, such as nitrogen. Ampules may consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, etc., ceramic, metal or any other material typically employed to hold similar reagents. Other examples of suitable containers include simple bottles that may be fabricated from similar substances as ampules, and envelopes, that may comprise foil-lined interiors, such as 30 aluminum or an alloy. Other containers include test tubes, vials, flasks, bottles, syringes, or th cLike. Contaners may have a sterile access port, such as a bottle having a stopper that can be pie-rced by a hyodermie injection needle. Other containers may hve~ two compartmnents -34- WO 2008/033987 PCT/US2007/078371 that are separated by a readily removable membrane that upon removal permits the components to be mixed. Removable membranes may be glass, plastic, rubber, etc. Kits may also be supplied with instructional materials. Instructions may be printed on paper or other substrate, and/or may be supplied as an electronic-readable medium, such as a 5 floppy disc. CD-ROM, DVD-ROM, Zip disc, videotape, audiotape, flash memory device etc. Detailed instructions may not be physically associated with the kit instead, a user may be directed to an internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail. 10 Modification of Cupredoxin and Variants, Derivatives and Structural Equivalents Thereof Cupredoxin or variant, derivative or structural equivalents thereof may be chemically modified or genetically altered to produce variants and derivatives as explained above. Such variants and derivatives may be synthesized by standard techniques. 15 In addition to naturally-occurring allelic variants of cupredoxin, changes can be introduced by mutation into cupredoxin coding sequence that incur alterations in the amino acid sequences of the encoded cupredoxin that do not significantly alter the ability of cupredoxin to inhibit the development of premalignant lesions. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the cupredoxin 20 without altering pharmacologic activity, whereas an "essential" amino acid residue is required for such pharmacologic activity. For example, amino acid residues that are conserved among the cupredoxins are predicted to be particularly non-amenable to alteration, and thus "essential." Amino acids for which conservative substitutions that do not change the 25 pharmacologic activity of the polypeptide can be made are well known in the art. Useful conservative substitutions are shown in Table 3, "Preferred substitutions" Conservative substitutions whereby an amino acid of one class is replaced with another amino acid of the same type fall within the scope of the invention so long as the substitution does not materially alter the pharmacologic activity of the compound. -35- WO 2008/033987 PCT/US2007/078371 Table 3. Preferred substitutions Preferred Original residue Exemplary substitutions substitutions Ala (A) Val, Leu, Ile Val Arg (R) Lys, Gin, Asn Lys Asn (N) Gln, His, Lys, Arg Gin Asp (D) Glu Glu Cys (C) Ser Ser GIn (Q) Asn Asn Glu (E) Asp Asp Gly (G) Pro, Ala Ala His (H) Asn, Gln, Lys, Arg Arg Ile (1) Leu, Val, Met, Ala, Phe, Leu Norleucine Leu (L) Norleucine, Ile, Val, Met, Ala, I Phe Lys (K) Arg, Gln, Asn Arg Met (M) Leu, Phe, Ile Leu Phe (F) Leu, Val, Ile, Ala, Tyr Leu Pro (P) Ala Ala Ser (S) Thr Thr Thr (T) Ser Ser Trp (W) Tyr, Phe Tyr Tyr (Y) Trp, Phe, Thr, Ser Phe Val (V) Ile, Leu, Met, Phe, Ala, Leu Norleucine Non-conservative substitutions that affect (1) the structure of the polypeptide 5 backbone, such as a p-sheet or a-helical conformation, (2) the charge, (3) hydrophobicity, or (4) the bulk of the side chain of the target site can modify the pharmacologic activity. Residues are divided into groups based on common side-chain properties as denoted in Table 4. Non-conservative substitutions entail exchanging a member of one of these classes for another clas. Substitutions may be introduced into conservative substitution sites or more 10 specifically into non-conserved sites. -36- WO 2008/033987 PCT/US2007/078371 Table 4. Amino acid classes Class Amino acids hydrophobic Norleucine, Met, Ala, Val, Leu, Ile neutral hydrophilic Cys, Ser, Thr acidic Asp, Glu basic Asn, Gln, His, Lys, Arg disrupt chain conformation Gly, Pro aromatic Trp, Tyr, Phe The variant polypeptides can be made using methods known in the art such as 5 oligonucleotide-mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter, Biochem J. 237:1-7 (1986); Zoller and Smith, Methods Enzymol 154:329-350 (1987)), cassette mutagenesis, restriction selection mutagenesis (Wells et al, Gene 34:315-323 (1985)) or other known techniques can be performed on the cloned DNA to produce the cupredoxin variant DNA. 10 Known mutations of cupredoxins can also be used to create variant cupredoxin to be used in the methods of the invention. For example, the CI 12D and M44KM64E mutants of azurin are known to have cytotoxic and growth arresting activity that is different from the native azurin, and such altered activity can be useful in the treatment methods of the present invention. 15 A more complete understanding of the present invention can be obtained by reference to the following specific Examples. The Examples are described solely for purposes of illustration and are not intended to limit the scope of the invention. Changes in form and substitution of equivalents are contemplated as circumstances may suggest or render expedient. Although specific terms have been employed herein, such terms are intended in a 20 descriptive sense and not for purposes of limitations. Modifications and variations of the invention as hereinbefore set forth can be made without departing from. the spirit and scope thereof -37- WO 2008/033987 PCT/US2007/078371 EXAMPLES Example 1. Effect of peptide P-28 on DMBA-Induced Mammary lesions in the MMOC 5 Model The mouse mammalary gland organ culture (MMOC) model allows evaluating efficacy of potentially chemopreventive agents against development of mammary alveolar lesions (MAL) or mammary ductal lesions (MDL) in response to DMBA. DMBA under appropriate incubation conditions form either MAL or MDL based on the hormonal milieu in 10 the medium. Hawthorne et al., Pharmaceutical Biology 40: 70-74 (2002); Mehta et al., J. Natl. Cancer Inst. 93: 1103-1106 (2001). Estrogen and progesterone-treated glands in culture develop ductal lesions whereas aldosterone and hydrocortisone-treated glands form estrogen and progesterone-independent alveolar lesions. Mammary glands not exposed to a carcinogen or chemopreventive agent, undergo structural regression in the absence of growth 15 promoting hormones, whereas treatment with DMBA for the 24-hr period between days 3 and 4 prevents the regression of structures caused by deprivation of hormones. It is assumed that this is because the glands have lost normal hormonal responsiveness and now have altered their course of development. Generating mammary adenocarcinoma by transplanting transformed cells into syngeneic mice has proved the premalignant preneoplastic nature of 20 these unrepressed areas. The thoracic pair of mammary glands was excised aseptically from each Balb/c mouse, and the glands were divided into several groups. The effects of p28 were evaluated at 4 different dilutions in the medium. Carcinogen treated glands without the test agent served as a measure to determine percent incidence in the absence of a chemopreventive agent. An 25 additional control was included to serve as a positive control for chemoprevention. Azurin was included in the medium at 50 gA/ml concentration. For alveolar lesions (MAL) stained glands were evaluated for the incidence of lesions (glands containing any lesions as compared to total number of glands in a given treatment group). For the ductal lesions (MDL) similar protocol was adapted, however, as indicated below in the methods section the 30 hormonal combination is different for alveolar and ductal lesions. The glands were fixed in fornalin and then processed for histopathoiogy. The sections are stained with eosin and -38- WO 2008/033987 PCT/US2007/078371 hematoxelene and evaluated under microscope. Here the multiplicity of ductal lesions between the control and the treatment groups are compared. Organ Culture Procedure. The experimental animals used for the studies were young, virgin BALB/c female mice 3 to 4 weeks of age obtained from Charles River, Wilmington, MA. The mice were treated daily by subcutaneous injections with I pg estradiol-170 +j3 1 mg progesterone for 9 days. This treatment is a prerequisite inasmuch as animals not pretreated with steroids fail to respond to hormones in vitro. The entire culture procedure is described in detail. Jang et al., Science 275:218-220 (1997); Mehta, Eu. J. Cancer 36:1275-1282 (2000); Mehta et al., J. Natl. Cancer Inst. 89:212-219 (1997); Mehta et al., J. Natl. Cancer 10 Inst. 93:1103-1106 (2001). Briefly, the animals were killed by cervical dislocation, and the thoracic pair of mammary glands were dissected out on silk rafts and incubated for 10 days in serum free Waymouth MB752/1 medium (5-glands/5 ml/dish). The medium was supplemented with glutamine, antibiotics (penicillin and streptomycin 100 units/ml medium) and growth 15 promoting hormones, 5 pg insulin (I), 5 pg prolactin (P), I pg aldosterone (A) and 1 pg hydrocortisone (H) per ml of medium for the protocol to induce mammary alveolar lesions (MAL). For induction of ductal lesions (MDL), the medium contained 5 pg/ml, 5 pg/ml P, 0.001 pg/ml estradiol 17p and 1 pg/ml progesterone (Pg). Mehta et al., J. Natl. Cancer Inst. 93:1103-1106 (2001) The carcinogen, DMBA (2 pg/ml) was added to the medium between 20 days 3 and 4. For the present study, DMBA was dissolved in DMSO at a final concentration of 4 mg/ml, and 50 pg I was added to 100 ml medium resulting in 2 pg/ml final concentrations. The control dishes contained DMSO as vehicle. On day 4, DMBA is removed from the medium by rinsing the glands in fresh medium and transferring them to new dishes containing fresh medium without DMBA. After 10 days 25 of incubation, the glands were maintained for another 14 days in the medium containing only 1(5 pg/ml). During the entire culture period, the glands were maintained at 37 0 C under 95% 02 and 5% CO 2 environment. The chemopreventive agent was included in the medium during the first ten days of growth -promoting phase. The test peptide p28 was evaluated at 4 concentrations ranging from 12.5 g/ml to 100 pg/mi. Azurin was evaluated at 50 pg/ml in 30 the medium. The peptide was dissolved in sterile water and filtered prior to use. The medium was changed three times per week (Monday, Wednesday and Friday. t the end of the exposure, the glands were fixed in fonmalin. -39- WO 2008/033987 PCT/US2007/078371 Results were analyzed by Chi-square analysis and Fisher's Exact Test. Morphometic Analysis of MAL. For examination of MAL, the glands were stained in alum carmine, and evaluated for the presence of the lesions. The glands were scored for the presence or absence of mammary lesions, severity of lesions per gland, and toxicity of the 5 agent. The glands stored in xylene were evaluated for the presence or absence, incidence, and severity of mammary lesions for each gland under a dissecting microscope. Mammary glands were scored as positive or negative for mammary lesions, and the percent incidence was determined as a ratio of glands exhibiting lesions and the total number of glands in that group. Dilation of ducts or disintegration of mammary structure because of treatment with 10 chemopreventive agent was considered a toxic effect. The data were subjected to statistical analysis for the incidence to determine the effectiveness of the potential chemopreventive agents. Figure IA shows a representative photograph of alveolar lesions in a DMBA treated gland and its comparison with a gland that was treated with DMBA along with a 15 chemopreventive agent. The effects of p28 on the development of alveolar lesion are shown in Figures lB- 1G and summarized in Figure 2. The peptide p28 inhibited MAL formation by 67% at 25 jg/ml concentration. Increasing concentration further up to 100 pg/ml did not enhance the efficacy of the peptide. The comparison of the peptide with azurin indicated that p28 was as effective as azurin for MAL development. Azurin at 50 pg/mil concentration 20 resulted in a 67% inhibition. Statistical analyses indicated that the effect of p28 was statistically significant compared to DMBA control at concentrations greater than 12.5 pg/ml (p<0.01, Fisher's Exact Test; Chi Square analysis). Histopathological Evaluation of MDL. For MDL, the glands were processed for histopathological evaluations. The glands were sectioned longitudinally into 5-micron 25 sections and stained with eosin hematoxeline. The longitudinal section of each gland was divided into several fields and each field was evaluated for ductal lesions. Mehta el al., J. Nat. Cancer Inst. 93:1103-1106 (2001) Briefly, the entire gland is evaluated under the scope; smaller glands will have fewer total fields as compared to larger glands. Thus, each gland will have variable number of fields. Often the number of sections through the ducts 30 also varies greatly from gland to gland. This results in the variable number from group to group Fields containing dua hyperplasia or atypia were determined and wre con mare with total nurnber of field evaluated for each gland. No discrimination is made between the -40- WO 2008/033987 PCT/US2007/078371 hyperplasia or atypia and severely occluded glands. Any field containing any of these histological patterns was considered positive for the lesion. The treatment groups were compared with the controls for the severity and percent inhibition was calculated. Figure 3 shows a representative ductal lesion. DMBA induces ductal lesions varying 5 from hyperplasia. atypia to complete occlusion of the ducts. A ratio of ductal lesions/total number of ductal sections was determined. Again, 12 5 g/ml concentration of p28 suppressed only 15% of the MDL formation. However, at 25 jpg/ml there was a significant inhibition of the lesions comparable to that observed with 50 pg/ml azurin. The efficacy of p28 at concentrations greater than 12.5 pg/ml was statistically significant (p<0.01, Fishers 10 Exact Test). These results are summarized in Figure 4. Often effects of chemopreventive agents can be differentiated between the MAL and MDL. For example tamoxifen inhibited the development of MDL but not MAL, It is interesting to note that azurin and p28 inhibited both estrogen and progesterone-dependent ductal lesions as well as independent alveolar lesions. 15 100011 This example indicates that both p28 and azurin can prevent the development of precancerous lesions in breast tissue. Thus, p28 and azurin may be used as chemopreventive agents in mammalian patients. -41-

Claims (8)

1. An isolated peptide that is a variant, derivative or structural equivalent of a cupredoxin; and that can inhibit the development of premalignant lesions in mammalian 5 tissue.
2. The isolated peptide of claim 1, wherein the cupredoxin is selected from the group consisting of azurin, pseudoazurin, plastocyanin, rusticyanin, Laz, auracyanin, stellacyanin and cucumber basic protein..
3. The isolated peptide of claim 2, wherein the cupredoxin is azurin. 10 4. The isolated peptide of claim 1, wherein the cupredoxin is from an organism selected from the group consisting of Pseudomonas aeruginosa, Alcaligenesfaecalis, Achromobacter xylosoxidan, Bordetella bronchiseptica, Methylomonas sp., Neisseria meningitidis, Neisseria gonorrhea, Pseudomonas fluorescens, Pseudomonas chlororaphis, Xylellafastidiosa and Vibrio parahaemolyticus. 15 5. The isolated peptide of claim 4, that is from Pseudomonas aeruginosa.
6. The isolated peptide of claim 1, which is part of a peptide selected from the group consisting of SEQ ID NOS: 1, 3-19.
7. The isolated peptide of claim 1, to which a sequence selected from the group consisting of SEQ ID NOS: 1, 3-19 has at least 80% amino acid sequence identity. 20 8. The isolated peptide of claim 1, which is a truncation of the cupredoxin.
9. The isolated peptide of claim 8, wherein the peptide is more than about 10 residues and not more than about 100 residues.
10. The isolated peptide of claim 8, wherein the peptide comprises a sequence selected from the group consisting of Pseudomonas aeruginosa azurin residues 50-77, 25 Pseudomonas aeruginosa azurin residues 50-67, Pseudomonas aeruginosa azurin residues
36-88, and SEQ ID NOS: 20-24. 11. The isolated peptide of claim 10, wherein the peptide consists of a sequence selected from the group consisting of Pseudomonas aeruginosa azurin residues 50-77, Pseudomonas aeruginosa azurin residues 50-67, Pseudomonas aeruginosa azurin residues 30 36-88 and SEQ ID NOS: 20-24. -42- WO 2008/033987 PCT/US2007/078371 12. The isolated peptide of claim 1, wherein the peptide comprises equivalent residues of a cupredoxin as a region of Pseudomonas aeruginosa azurin selected from the group consisting of residues 50-77, residues 50-67 and residues 36-88. 13. A pharmaceutical composition, comprising at least one cupredoxin or peptide 5 of claim 1 in a pharmaceutically acceptable carrier. 14. The pharmaceutical composition of claim 13 which comprises at least two of the cupredoxins or peptides. 15. The composition of claim 13, wherein the pharmaceutical composition is formulated for intravenous administration. 10 16. The composition of claim 13, wherein the cupredoxin is from an organism selected from the group consisting of Pseudomonas aeruginosa, Alcaligenesfaecalis, Achromobacter xylosoxidan, Bordetella bronchiseptica, Methylomonas sp., Neisseria meningitidis, Neisseria gonorrhea, Pseudomonasfluorescens, Pseudomonas chlororaphis, Xylellafastidiosa and Vibrio parahaemolyticus. 15 17. The composition of claim 16, wherein the cupredoxin is from Pseudomonas aeruginosa. 18. The composition of claim 13, wherein the cupredoxin is selected from the group consisting of SEQ ID NOS: 1, 3-19. 19. A method to treat a mammalian patient, comprising administering to the 20 patient a therapeutically effective amount of the composition of claim 13. 20. The method of claim 19, wherein the patient is human. 21. The method of claim 19, wherein the patient is at a higher risk to develop cancer than the general population. 22. The method of claim 21, wherein the cancer is selected from melanoma, 25 breast, pancreas, glioblastoma, astrocytoma, lung, colorectal, neck and head, bladder, prostate, skin, and cervical cancer. 23. The method of claim 21, wherein the patient has at least one high risk feature. 24. The method of claim 19, wherein the patient has premalignant lesions. 25. The method of claim 19, wherein the patient has been cured of cancer or 30 premalignant lesions. 26. The method of claim 19, wherein the pharmaceutical composition is administered by a mode selected from the group consisting of intravenous injection, -43- WO 2008/033987 PCT/US2007/078371 intramuscular injection, subcutaneous injection, inhalation, topical administration, transdermal patch, suppository, vitreous injection and oral. 27. The method of claim 24, wherein the mode of administration is by intravenous injection. 5 28. The method of claim 21, wherein the pharmaceutical composition is co administered with at least one other chemopreventive drug. 29. The method of claim 26, wherein the pharmaceutical composition is administered at about the same time as another chemopreventive drug. 30. A kit comprising the composition of claim 13 in a vial. 10 31. The kit of claim 30, wherein the kit is designed for intravenous administration. 32. A method to study the development of cancer comprising contacting the mammalian cells with a cupredoxin or peptide of claim 1; and measuring the development of premalignant and malignant cells. 33. The method of claim 32, wherein the cells are human cells. 15 34. The method of claim 32, wherein the cells are mammary gland cells. 35. The method of claim 32, wherein the cells are induced to develop cancer. 36. An expression vector, which encodes the peptide of claim 1. -44-
AU2007296429A 2006-09-14 2007-09-13 Compositions and methods to prevent cancer with cupredoxins Abandoned AU2007296429A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US84435806P 2006-09-14 2006-09-14
US60/844,358 2006-09-14
PCT/US2007/078371 WO2008033987A2 (en) 2006-09-14 2007-09-13 Compositions and methods to prevent cancer with cupredoxins

Publications (1)

Publication Number Publication Date
AU2007296429A1 true AU2007296429A1 (en) 2008-03-20

Family

ID=39184582

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007296429A Abandoned AU2007296429A1 (en) 2006-09-14 2007-09-13 Compositions and methods to prevent cancer with cupredoxins

Country Status (13)

Country Link
EP (1) EP2061498A4 (en)
JP (2) JP2010503409A (en)
KR (1) KR20090059152A (en)
CN (1) CN101595124A (en)
AU (1) AU2007296429A1 (en)
BR (1) BRPI0714987A2 (en)
CA (1) CA2663498A1 (en)
IL (1) IL197602A0 (en)
MX (1) MX2009002787A (en)
NO (1) NO20091267L (en)
RU (1) RU2009113812A (en)
SG (1) SG174784A1 (en)
WO (1) WO2008033987A2 (en)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9096663B2 (en) 2001-02-15 2015-08-04 The Board Of Trustees Of The University Of Illinois Compositions and methods for treating conditions related to ephrin signaling with cupredoxins and mutants thereof
US7381701B2 (en) 2001-02-15 2008-06-03 The Borad Of Trustees Of The University Of Illinois Compositions and methods for treating conditions related to ephrin signaling with cupredoxins
US7556810B2 (en) 2005-07-19 2009-07-07 The Board Of Trustees Of The University Of Ilinois Compositions and methods to control angiogenesis with cupredoxins
US10675326B2 (en) 2004-10-07 2020-06-09 The Board Of Trustees Of The University Of Illinois Compositions comprising cupredoxins for treating cancer
US9598470B2 (en) 2004-10-07 2017-03-21 Craig W. Beattie Compositions and methods to prevent cancer by stabilizing P53 through non MDM2-mediated pathways
UA97466C2 (en) 2004-10-07 2012-02-27 Ананда Чакрабарти Cupredoxin derived transport agents and methods of use thereof
US9434770B2 (en) 2004-10-07 2016-09-06 Tapas K Das Gupta Modified cupredoxin derived peptides
US9968685B2 (en) 2004-10-07 2018-05-15 Brad N. Taylor Methods to treat cancer with cupredoxins
JP2009502135A (en) 2005-07-19 2009-01-29 ザ・ボード・オブ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・イリノイ Transport factors for crossing the blood-brain barrier and into brain cancer cells and methods of use thereof
MX2009005849A (en) 2006-12-04 2009-08-12 Univ Illinois Compositions and methods to treat cancer with cupredoxins and cpg rich dna.
EP2114429A4 (en) 2007-02-08 2010-05-19 Univ Illinois Compositions and methods to prevent cancer with cupredoxins
KR20200058421A (en) * 2017-09-29 2020-05-27 온크그노스틱스 게엠베하 Determination of risk for neoplasia and cancer

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7491394B2 (en) * 2001-02-15 2009-02-17 The Board Of Trustees Of The University Of Illinois Cytotoxic factors for modulating cell death
CN1506375A (en) * 2002-12-12 2004-06-23 浙江养生堂天然药物研究所有限公司 Azurin as bacterial protein with wide-spectrum antitumor function and its use and medicinal composition
UA97466C2 (en) * 2004-10-07 2012-02-27 Ананда Чакрабарти Cupredoxin derived transport agents and methods of use thereof
JP2008539795A (en) * 2005-05-20 2008-11-20 ザ・ボード・オブ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・イリノイ Compositions and methods for treating symptoms related to ephrin signaling using cupredoxins
JP2009502797A (en) * 2005-07-19 2009-01-29 ザ・ボード・オブ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・イリノイ Compositions and methods for controlling angiogenesis with cupredoxins

Also Published As

Publication number Publication date
JP2010503409A (en) 2010-02-04
KR20090059152A (en) 2009-06-10
WO2008033987A2 (en) 2008-03-20
SG174784A1 (en) 2011-10-28
WO2008033987A3 (en) 2009-04-30
RU2009113812A (en) 2010-10-20
BRPI0714987A2 (en) 2013-07-30
EP2061498A2 (en) 2009-05-27
CN101595124A (en) 2009-12-02
EP2061498A4 (en) 2009-11-04
IL197602A0 (en) 2011-08-01
MX2009002787A (en) 2009-08-27
NO20091267L (en) 2009-06-04
CA2663498A1 (en) 2008-03-20
JP2014050390A (en) 2014-03-20

Similar Documents

Publication Publication Date Title
AU2007296429A1 (en) Compositions and methods to prevent cancer with cupredoxins
US10117905B2 (en) Compositions and methods to prevent cancer with cupredoxins
US10086037B2 (en) Compositions and methods to prevent cancer with cupredoxins
US10351605B2 (en) Compositions and methods to prevent cancer by stabilizing p53 through non MDM2-mediated pathways
CA2747192A1 (en) Compositions and methods to prevent cancer with cupredoxins
US8158574B2 (en) Compositions and methods to prevent cancer with cupredoxins
KR20080019615A (en) Compositions and methods for treating malaria with cupredoxin and cytochrome
KR20080024124A (en) Compositions and methods for treating hiv infection with cupredoxin and cytochrome c

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period