WO2002028418A1 - Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine - Google Patents

Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine Download PDF

Info

Publication number
WO2002028418A1
WO2002028418A1 PCT/US2001/042484 US0142484W WO0228418A1 WO 2002028418 A1 WO2002028418 A1 WO 2002028418A1 US 0142484 W US0142484 W US 0142484W WO 0228418 A1 WO0228418 A1 WO 0228418A1
Authority
WO
WIPO (PCT)
Prior art keywords
relaxin
tissue
receptor
delivery
cells
Prior art date
Application number
PCT/US2001/042484
Other languages
English (en)
Other versions
WO2002028418B1 (fr
Inventor
Edward P. Amento
Chrishan S. Samuel
Original Assignee
Molecular Medicine Research Institute
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US10/398,553 priority Critical patent/US20050032683A1/en
Application filed by Molecular Medicine Research Institute filed Critical Molecular Medicine Research Institute
Priority to AU1185502A priority patent/AU1185502A/xx
Priority to JP2002532242A priority patent/JP2004510743A/ja
Priority to AU2002211855A priority patent/AU2002211855B2/en
Priority to IL15525001A priority patent/IL155250A0/xx
Priority to CA2425712A priority patent/CA2425712C/fr
Priority to NZ525389A priority patent/NZ525389A/en
Priority to EP01979945A priority patent/EP1326627A4/fr
Publication of WO2002028418A1 publication Critical patent/WO2002028418A1/fr
Publication of WO2002028418B1 publication Critical patent/WO2002028418B1/fr
Priority to US11/781,872 priority patent/US7833526B2/en
Priority to US12/945,685 priority patent/US8119136B2/en
Priority to US13/399,620 priority patent/US9534034B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/64Relaxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2221Relaxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/06Antiabortive agents; Labour repressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/12Drugs for genital or sexual disorders; Contraceptives for climacteric disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out

Definitions

  • Apoptosis is required to remove existing cells, including immature or damaged cells. Apoptosis naturally occurs in virtually all tissues of the body. Apoptosis plays a critical role in tissue homeostasis, that is, it ensures that the number of new cells produced are correspondingly offset by an equal number of cells that die. For example, the cells in the intestinal lining divide so rapidly that the body must eliminate cells after only three days in order to prevent the overgrowth of the intestinal lining.
  • the disruption of the genetic program by either abnormally increasing or decreasing rate of cell proliferation and/or apoptosis can result in abnormal tissue development. For example, decreases in cell proliferation below normal levels can lead to immature tissues and other tissue abnormalities. Increases in cell proliferation above normal levels are thought to be major events in the development of neoplasia and cancer, as well as other cell proliferative disorders. Abnormal increases in apoptosis can also lead to precancerous lesions.
  • Precancerous lesions include lesions of the breast (that can develop into breast cancer), lesions of the prostate (that can develop into prostate cancer) or skin (that can develop into malignant melanoma or basal cell carcinoma), colonic adenomatous polyps (that can develop into colon cancer), and other such neoplasia. Such lesions exhibit a strong tendency to develop into malignant tumors or cancer.
  • Precancerous lesions can result from an accumulation of insults to existing cells in various tissues of the body.
  • insults can include exposure to sunlight, radiation, mutagens and carcinogens normally found in the diet, chemicals such as pesticides, herbicides, preservatives, and the like.
  • These insults can result in the accumulation of mutations in the cells, which can lead to hyperplastic conditions (i.e., abnormal increases in cell number), such as, for example, hyperplasia of liver, kidney, spleen, thymus, intestine, lung or prostate tissues.
  • hyperplastic conditions i.e., abnormal increases in cell number
  • the down-regulation of apoptosis can also lead to the accumulation of cells in these hyperplastic conditions.
  • An abnormal increase in apoptosis can interfere with normal development and/or differentiation of tissues. For example, apoptosis is required during pregnancy and for maturation of the male reproductive tract tissues. An abnormal increase in apoptosis can also interfere with the formation of new cells and tissues, thereby preventing normal tissue maturation or development.
  • the present invention relates to the discovery that relaxin is associated with the development of body tissues. Knockouts of the gene encoding relaxin result in various abnormalities in the development of various body tissues, including smaller size of, increased collagen deposition in, and immaturity of such tissues. Conversely, relaxin-responsive cell accumulation leads to abnormalities in body tissues.
  • the present invention provides methods of modulating apoptosis in tissues by administering a relaxin agonist or a relaxin antagonist.
  • the present invention provides methods for modulating apoptosis in a subject by administering to a subject in need thereof an effective amount of a relaxin agonist for a period of time sufficient to decrease apoptosis in cells expressing a relaxin receptor.
  • Tissues which are typically affected by the administration of the relaxin agonist useful in methods according to the present invention include, for example, liver, kidney, spleen, thymus, brain, heart, intestine, skin, lung, the male reproductive tract, and the female reproductive tract.
  • Male reproductive tract tissues include, for example, prostatic, epididymal, seminiferous tissues, tissues of the testes, and the like.
  • Female productive tract tissues include, for example, uterus, cervix, the interpubic ligament, connective tissues within the pelvic girdle, and the like.
  • the administration of the relaxin agonist typically reduces the number of apoptotic cells.
  • the relaxin agonist stimulates maturation of the tissue.
  • a relaxin agonist can stimulate maturation of male reproductive tract tissue, such as prostatic tissue, epididymal tissue, seminiferous tissue, testicular tissue or sperm.
  • the maturation results in an increase in cell number in an under-developed testes, such as, for example, an increase in the number of mature testicular cells.
  • maturation of male reproductive tract tissue results in an increase in the number of viable sperm cells, as compared with tissue not contacted with the relaxin agonist.
  • fibrosis can be reduced by the relaxin agonist, and/or excessive collagen deposition can be reduced.
  • the subject in need of administration of the relaxin agonist can have a relaxin-deficient condition, such as, for example, immature tissue, excessive collagen deposition and/or a low sperm count.
  • a relaxin-deficient condition such as, for example, immature tissue, excessive collagen deposition and/or a low sperm count.
  • the immature tissue can be immature male productive tissue (e.g., underdeveloped testes). Such immature tissue can be present in an otherwise mature or immature animal.
  • the relaxin agonist can be relaxin, a relaxin analog, a small molecule relaxin effector or a relaxin nucleic acid.
  • the relaxin is typically vertebrate relaxin and more typically is human relaxin.
  • compositions comprising a relaxin agonist useful in the methods according to the present invention can be formulated for administration, for example, by infusion, injection, oral delivery, nasal delivery as well as intrapulmonary, rectal, transdermal, interstitial or subcutaneous delivery. Such compositions also can be formulated for delayed release of the relaxin agonists into the tissues and circulation of the subject. Particular embodiments comprise compositions formulated for infusion or injection, or by intrapulmonary, subcutaneous or transdermal delivery.
  • nucleic acids encoding relaxin agonists can formulated for administration to the subject in a vector encoding the relaxin agonist.
  • the vector can be an expression vector which expresses the relaxin agonist in the cells.
  • relaxin or relaxin analog nucleic acids can be formulated for delivery to the subject.
  • the subjects can be pre-pubescent or post-pubescent.
  • the present invention provides methods for modulating apoptosis in a subject in need thereof by administering an effective amount of a relaxin antagonist for a period of time sufficient to increase apoptosis in a cell population expressing a relaxin receptor.
  • the relaxin antagonist inhibits binding of relaxin to relaxin receptor.
  • the relaxin antagonist reduces relaxin-associated tissue remodeling.
  • Tissues which are typically affected by the administration of the relaxin antagonist useful in methods according to the present invention include, for example, liver, kidney, spleen, thymus, brain, heart, intestine, skin, lung, the male reproductive tract, the female reproductive tract, and the like.
  • Male reproductive tract tissues include, for example, prostatic, epididymal, seminiferous tissues, tissues of the testes, and the like.
  • the male reproductive tract tissue can be prostatic tissue, and can be mature or immature.
  • Suitable target female productive tract tissues include, for example, uterus, cervix, the interpubic ligament, connective tissues within the pelvic girdle, and the like.
  • the cell population can comprise cells expressing a relaxin receptor such as, for example, fibroblasts, osteoblasts, monocytes epithelial cells, endothelial cells, and the like.
  • the relaxin antagonist can be, for example, a relaxin binding agent, a relaxin receptor binding agent, a relaxin antisense nucleic acid, and the like.
  • the relaxin binding agent can be, for example, an anti-relaxin antibody, a soluble relaxin receptor, a small molecule relaxin antagonist, and the like.
  • the relaxin receptor binding agent can be, for example, an anti-relaxin receptor antibody, a relaxin analog, a small molecule relaxin receptor antagonist, and the like.
  • Antibodies that bind relaxin or relaxin receptor can include, for example, polyclonal antibodies, monoclonal antibodies, an Fab, Fab', an F(ab') 2 , an Fv, a single heavy chain, a chimeric antibody, and the like.
  • Compositions comprising a relaxin antagonist useful in the methods of the present invention can be formulated for administration, for example, by infusion, injection, oral delivery, nasal delivery as well as intrapulmonary, rectal, transdermal, interstitial or subcutaneous delivery.
  • Compositions can also be formulated for delayed release of the relaxin antagonist into the tissues and circulation of the subject.
  • Particular embodiments comprise compositions formulated for infusion or injection or by intrapulmonary, subcutaneous or transdermal delivery.
  • nucleic acids encoding relaxin antagonists can also be administered to the subject in a vector encoding the relaxm antagonist.
  • the vector is an expression vector which expresses the relaxin antagonist in the cell population.
  • relaxin or relaxin receptor antisense nucleic acids can be delivered directly to the subject, according to any of the methods described above.
  • administration of the relaxin antagonist increases apoptosis to reduce unwanted cell accumulation.
  • the unwanted cells can be hyperplasia, hypertrophy, cancer or neoplasia.
  • nucleic acid refers to a polymer composed of a multiplicity of nucleotide units (ribonucleotide or deoxyribonucleotide or related structural variants) linked via phosphodiester bonds.
  • a nucleic acid can be of substantially any length, typically from about six (6) nucleotides to about 10 9 nucleotides, or larger.
  • nucleic acids are as follows: the left-hand end of single-stranded nucleic acid is the 5' end; the left-hand direction of double-stranded nucleic acid is referred to as the 5' direction. The direction of 5' to 3' addition of nascent RNA transcripts is referred to as the transcription direction. Sequence regions on the DNA strand having the same sequence as the RNA and which are 5' to the 5' end of the RNA transcript are referred to as "upstream sequences;" sequence regions on the DNA strand having the same sequence as the RNA and which are 3' to the 3' end of the coding RNA transcript are referred to as "downstream sequences".
  • Nucleic acids include RNA, niRNA, cDNA, genomic DNA, synthetic forms, and mixed polymers, both sense and antisense strands, and can also be chemically or biochemically modified or can contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those skilled in the art.
  • Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, and carbamates), charged linkages (e.g., phosphorothioates and phosphorodithioates), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine and psoralen), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids).
  • uncharged linkages e.g., methyl phosphonates, phosphotriesters, phosphoamidates, and carbamates
  • charged linkages e.g., phosphorothioates and phosphorodithioates
  • pendent moieties e.g., polypeptides
  • intercalators e.g
  • the commonly used one- and three-letter abbreviations for amino acids are used herein (see, e.g., Alberts et al., Molecular Biology of the Cell, Garland Publishing, Inc., New York (3d ed. 1994)).
  • a “conservative substitution” of a particular amino acid sequence refers to substitution of those amino acids that are not critical for polypeptide activity or substitution of amino acids with other amino acids having similar properties (e.g., acidic, basic, positively or negatively charged, polar or non-polar, and the like) such that the substitution of even critical amino acids does not substantially alter activity.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). (See also Creighton, Proteins, W. H. Freeman and Company (1984), which is incorporated by reference herein.)
  • individual substitutions, deletions or additions that alter, add or delete a single amino acid or a small percentage of amino acids in an encoded sequence are also "conservative substitutions.”
  • relaxin analog refers to a modified relaxm polypeptide that increases or decreases the functional activity of the molecule or its interaction with a relaxin receptor.
  • soluble in the context of a polypeptide, refers to the ability of the polypeptide to be dissolved in (i.e., to be molecularly or ionically dispersed in) an aqueous solution, such as water, blood or plasma.
  • small molecule effector in the context of relaxin or a relaxin receptor, refers to an agent that binds to a relaxin, or to a relaxin receptor, and stimulates the activity of relaxin or relaxin receptor.
  • small molecule antagonist in the context of a relaxin or a relaxin receptor, refers to an agent that binds to a relaxin, or to a relaxin receptor, and reduces or inhibits the activity of relaxin or relaxin receptor.
  • tissue refers to a population of cells, generally consisting of cells of the same kind that perform the same, or a similar, function.
  • a tissue can be part of an organ or bone or it can be a loose association of cells, such as cells of the immune system.
  • tissue in the context of body tissue, refers to the developmental progression and/or differentiation of the tissue towards its full developed state.
  • tissue in the context of body tissue, refers to the achievement of full development, differentiation and/or growth of the tissue.
  • tissue in the context of body tissue, refers to a tissue that has not fully developed and/or differentiated.
  • pre-pubescent male refers to a male that has not completed the process of puberty.
  • post-pubescent male refers to a male that has completed the process of puberty.
  • puberty refers to the sequence of events by which a child becomes a young adult, characterized by the beginning of gametogenesis, secretion of gonadal hormones, development of secondary sexual characteristics, and reproductive function.
  • tissue refers to an increase in the number of normal (i.e., non-mutant, non-malignant) cells in the tissue.
  • biologically active and “functionally active” refer to the ability of a molecule (e.g., a relaxin agonist or antagonist) to bind to a relaxin or relaxin receptor and to stimulate or inhibit apoptosis, cell accumulation/ and/or tissue maturation, in a body tissue.
  • relaxin-deficient condition refers to a disease, disorder or condition of a subject, in which relaxin levels, or relaxin-receptor levels, in the relevant tissue, cell(s), or in the subject, are below normal.
  • relaxin-responsive refers to an increase in cell number, or in the state of maturity (i.e., tissue development and/or differentiation), in response to the binding of relaxin to a relaxin receptor.
  • relaxin-associated refers to a property, condition or response of a cell or tissue by which the cell or tissue is affected, directly or indirectly, by an increase or decrease in functionally active relaxin levels, or in functionally active relaxin receptor.
  • hypoplasia or “hyperplastic tissue” refer to an increase in the number of cells in a tissue.
  • hypertrophy or “hypertrophic” refer to an increase in the size of a tissue.
  • cancer and “malignancy” generally refer to the various types of malignant neoplasms, most of which invade surrounding tissues.
  • cancer and malignant relate to cells or tissue having properties of cancer or a malignancy.
  • tissue remodeling refers to the formation of new cells or tissues and the destruction of existing cells through the apoptotic pathway, in response to the signals mediated by relaxin or a relaxin-receptor.
  • apoptosis refers to a regulated network of biochemical events which lead to a selective form of cell suicide, and is characterized by readily observable morphological and biochemical phenomena, such as the fragmentation of the deoxyribonucleic acid (DNA), condensation of the chromatin, which may or may not be associated with endonuclease activity, chromosomemigration, margination in cell nuclei, the formation of apoptotic bodies, mitochondrial swelling, widening of the mitochondrial cristae, opening of the mitochondrial permeability transition pores and/or dissipation of the mitochondrial proton gradient.
  • DNA deoxyribonucleic acid
  • condensation of the chromatin which may or may not be associated with endonuclease activity
  • chromosomemigration chromosomemigration
  • margination in cell nuclei the formation of apoptotic bodies
  • mitochondrial swelling widening of the mitochondrial cristae
  • the present invention provides methods for the administration of relaxin agonists or antagonists for the modulation of apoptosis.
  • the relaxin agonists and antagonists can be used to reduce, manage, treat or prevent relaxin-associated abnormalities.
  • Relaxin-associated abnormalities include diseases, disorder or conditions of a subject that are associated with increased or decreased levels of apoptosis, as compared with levels of apoptosis in a normal subject.
  • Relaxin- associated abnormalities include, for example, relaxin-deficient conditions and relaxin- responsive conditions.
  • a relaxin agonist is administered to a subject to treat a relaxin-associated abnormality having increased apoptosis, as compared with comparable cells from a normal subject (i.e., not having the relaxin-associated abnormality).
  • relaxin-associated abnormalities can include, for example, immature body tissue, increased collagen deposition, fibrosis (i.e., the presence of abnormal amounts of fibrous tissue, as compared with normal tissue), low tissue weight, increased apoptosis of cells in a cell population in a tissue, and the like.
  • Relaxin-associated abnormalities are typically associated with decreased levels of relaxin, and/or relaxm receptor, as compared with normal cell or tissues.
  • the relaxin-associated abnormality is a relaxin- deficient condition, such as, for example, immature tissue, the presence of excessive collagen, a low sperm count, and the like.
  • the immature tissue can be, for example, immature male productive tract tissue (e.g., immature prostatic, epididymal, seminiferous or testicular tissue or sperm).
  • Immature tissue can be characterized, for example, by increased collagen deposition, low weight, and/or decreased cell number, as compared with a comparable sample of normal tissue.
  • Such tissue also can be characterized by its lack of organization, by incomplete development and/or by a lack of, or incomplete, differentiation, as compared with a comparable sample of normal tissue.
  • fibrosis can be reduced, such as, for example, dermal fibrosis, lung fibrosis, kidney fibrosis, or age-related fibrosis of these or other organs or tissues.
  • the relaxin agonist is administered in an amount effective to reduce, manage, treat or prevent the relaxin-associated abnormality.
  • administration of a relaxin agonist can decrease apoptosis of target cells, stimulate maturation, development and/or differentiation of immature tissue, increase tissue weight, increase cell number in the tissue, decrease in collagen deposition, and the like.
  • a relaxin antagonist is administered to a subject to reduce, manage, treat or prevent a relaxin- associated abnormality by increasing apoptosis in the target cells.
  • relaxin associated abnormalities can be, for example, relaxin-associated cell accumulation, decreased apoptosis relative to normal cells or tissue, hyperplasia, hypertrophy, cancer, neoplasia, and the like.
  • the relaxin antagonist can reduce relaxin-associated tissue remodeling, reduce unwanted cell accumulation, reduce or prevent hyperplasia, hypertrophy, cancer, neoplasia, and the like.
  • Body tissues having relaxin-associated or relaxin-responsive tissue abnormalities can include, for example, tissues of the brain, heart, liver, kidney, spleen, thymus, intestine, skin, lung, male reproductive tract (e.g., prostate, epididymis, seminal vesicles or testes), or female reproductive tract (e.g., the uterus, cervix, the interpubic ligament or connective tissues of the pelvic girdle).
  • male reproductive tract e.g., prostate, epididymis, seminal vesicles or testes
  • female reproductive tract e.g., the uterus, cervix, the interpubic ligament or connective tissues of the pelvic girdle.
  • a relaxin agonist is administered to a subject to treat a relaxin-associated abnormality.
  • the relaxin agonist can be, for example, a relaxin, a relaxin analog, a small molecule relaxin effector, a relaxin nucleic acid, and the like.
  • the relaxin agonist is a relaxin polypeptide or a fragment or analog of a relaxin polypeptide.
  • the term “relaxin” refers to vertebrate relaxin polypeptides, including full length relaxin polypeptide or a portion of the relaxin polypeptide that retains biological activity.
  • Relaxin has been well defined in its natural human form, animal form, and in its synthetic form. In particular, relaxin has been extensively described in U.S. Patent Nos. 5,166,191 and 4,835,251 (both of which are hereby incorporated by reference).
  • “relaxin” generally refers to the terms “relaxin,” “human relaxin,” “native relaxin,” and “synthetic relaxin” as defined in U.S. Patent No. 5,166,191 and the terms “human relaxin” and “human relaxin analogs” as defined in U.S. Patent No. 4,835,251.
  • the relaxin is human relaxin, as described in, for example, U.S. Patent Nos.
  • Relaxin can also be prepared by synthesis of the A and B chains, and purification and assembly thereof, as described in European Patent 0 251 615 (published Jan. 7, 1988, the disclosure of which is incorporated herein by reference). For in vitro assembly of relaxin, a 4:1 molar ratio of A to B chains is generally employed. The resulting product is then purified by any means known to one of ordinary skill in the art, including, for example, reverse-phase HPLC, ion exchange chromatography, gel filtration, dialysis, and the like, or any combination of such procedures. Unprocessed or partially processed forms of relaxin, such as preprorelaxin or prorelaxin, can also be used.
  • relaxin polypeptides include the HI and H2 forms of human relaxin. It has been reported that the predominant species of human relaxin is the H2 relaxin form with a truncated B chain (i.e., relaxin H2(B29 A24)), wherein the four C-terminal amino acids of the B-chain are absent so that the B-chain ends with a serine at position 29. Either this form (referred to as designated “short relaxin” or "long relaxin,” which contains a B chain of 33 amino acids) can be used. Relaxin agonists further includes analogs, such as naturally-occurring amino acid sequence variants of relaxin.
  • Relaxin analogs also include those altered by substitution, addition or deletion of one or more amino acid residues that provide for functionally active relaxin polypeptides.
  • Such relaxin analogs include, but are not limited to, those containing as a primary amino acid sequence all or part of the amino acid sequence of a relaxin polypeptide, including altered sequences in which one or more functionally equivalent amino acid residues are substituted for residues within the sequence, resulting in a silent functional change (e.g., a conservative substitution).
  • the relaxin agonist is a polypeptide consisting of or comprising a fragment of a relaxin polypeptide having at least 10 contiguous amino acids of the relaxin polypeptide.
  • the fragment contains at least 20 or 25 contiguous amino acids of the relaxin polypeptide.
  • the fragments are not larger than 20 or 30 amino acids.
  • the relaxin analog can be a polypeptide comprising regions that are substantially similar to a relaxin polypeptide or fragments thereof (e.g., in various embodiments, at least 60%, 70%, 75%, 80%, 90%, or even 95% identity or similarity over an amino acid sequence of identical size), or when compared to an aligned sequence in which the alignment is done by a computer sequence comparison/alignment program known in the art, or which coding nucleic acid is capable of hybridizing to a relaxin nucleic acid, under high stringency, moderate stringency, or low stringency conditions (infra).
  • a relaxin nucleic acid e.g., Smith and Waterman, Adv. Appl. Math. 2:482 (1981); Needleman and Wunsch, J. Mol. Biol.
  • Relaxin agonists further comprise functionally active relaxin polypeptides, analogs or fragments that bind to a relaxin receptor.
  • Relaxin agonists such as relaxin polypeptides, analogs and fragments can be produced by various methods known in the art. The manipulations which result in their production can occur at the gene or polypeptide level.
  • cloned relaxin nucleic acids can be modified by any of numerous strategies known in the art (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 3d Ed., Cold Spring Harbor Laboratory Press, New York (2001); Ausubel et al., Current Protocols in Molecular Biology, 4th ed., John Wiley and Sons, New York (1999); which are incorporated by reference herein), such as making conservative substitutions, deletions, insertions, and the like.
  • the sequence can be cleaved at appropriate sites with restriction endonuclease(s), followed by further enzymatic modification if desired, isolated, and ligated in vitro.
  • the modified nucleic acid typically remains in the proper translational reading frame, so that the reading frame is not interrupted by translational stop signals or other signals that interfere with the synthesis of the relaxin analog or fragment.
  • the relaxin nucleic acid can also be mutated in vitro or in vivo to create and/or destroy translation initiation and/or tennination sequences.
  • the relaxin nucleic acid can also be mutated to create variations in coding regions and/or to form new restriction endonuclease sites or destroy preexisting ones and to facilitate further in vitro modification.
  • Any technique for mutagenesis known in the art can be used, including but not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (see, e.g., Hutchison et al, J. Biol. Chem. 253:6551-60 (1978)), the use of TAB ® linkers (Pharmacia), and the like. (See generally Sambrook et al, supra; Ausubel et al, supra.)
  • relaxin analogs are prepared from relaxin- encoding nucleic acids that are altered to introduce aspartic acid codons at specific position(s) within at least a portion of the relaxin coding region.
  • the resulting analogs can be treated with dilute acid to release a desired analog, thereby rendering the protein more readily isolated and purified.
  • Other relaxin analogs are disclosed in U.S. Patent Nos. 4,656,249; 5,179,195; 5,945,402; 5,811,395; and 5,795,807 (the disclosures of which are incorporated by reference herein).
  • relaxin polypeptide sequence can also be made at the polypeptide level. Included within the scope of the invention are relaxin polypeptides, analogs or fragments that are differentially modified during or after synthesis (e.g., in vivo or by in vitro translation). Such modifications include conservative substitution, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule, another polypeptide or other cellular ligand, and the like.
  • any of numerous chemical modifications can be carried out by known techniques, including, but not limited to, specific chemical cleavage (e.g., by cyanogen bromide), enzymatic cleavage (e.g., by trypsin; chymotrypsin, papain, V8 protease, and the like); modification by, for example, NaBH , acetylation, formylation, oxidation and reduction, metabolic synthesis in the presence of tunicamycin, and the like.
  • Relaxin polypeptides, analogs and fragments can be purified from natural sources by standard methods such as those described herein (e.g., immunoaffmity purification).
  • Relaxin polypeptides, analogs and fragments can also be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, sizing column chromatography, high pressure liquid chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of polypeptides.
  • Relaxin polypeptides can be synthesized by standard chemical methods known in the art (see, e.g., Hunkapiller et al., Nature 310:105-11 (1984); Stewart and Young, Solid Phase Peptide Synthesis, 2 nd Ed., Pierce Chemical Co., Rockford, IL, (1984); the disclosures of which are inco ⁇ orated by reference herein).
  • analogs of relaxin polypeptides can be chemically synthesized.
  • a peptide corresponding to a fragment of a relaxin polypeptide, which comprises a desired domain, or which mediates a desired activity in vivo can be synthesized by use of chemical synthetic methods using, for example, an automated peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the relaxin polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, the D-isomers of the common amino acids, ⁇ -amino isobutyric acid, 4- aminobutyric acid, 2-amino butyric acid, ⁇ -amino hexanoic acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, selenocysteine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids, N ⁇ - methyl amino acids, and amino acid analogs in general.
  • the amino acid can be D (dextrorotary) or L (le
  • the relaxin agonist is a chimeric, or fusion, protein comprising a relaxin polypeptide, or fragment thereof (typically consisting of at least a domain or motif of the relaxin polypeptide, or at least 10 contiguous amino acids of the relaxin polypeptide), joined at its amino- or carboxy-terminus via a peptide bond to an amino acid sequence of a different protein.
  • a chimeric protein is produced by recombinant expression of a nucleic acid encoding the chimeric polypeptide.
  • the chimeric product can be made by ligating the appropriate nucleic acid sequences, encoding the desired amino acid sequences, to each other in the proper reading frame and expressing the chimeric product by methods commonly known in the art.
  • the chimeric product can be made by protein synthetic techniques (e.g. , by use of an automated peptide synthesizer).
  • the fusion protein is a relaxin-ubiquitin fusion protein.
  • U.S. Patent No. 5,108,919 discloses methods for preparing a fusion protein of a relaxin chain and ubiquitin.
  • the relaxin analog, or fragment is functionally active (i.e., capable of exhibiting one or more functional activities associated with a full-length, wild-type relaxin polypeptide).
  • analogs or fragments that retain a desired relaxin property of interest e.g., binding to a relaxin binding partner (e.g., relaxin receptor) and/or modulation (e.g., inhibition) of apoptosis
  • a desired relaxin property of interest e.g., binding to a relaxin binding partner (e.g., relaxin receptor) and/or modulation (e.g., inhibition) of apoptosis
  • a specific embodiment relates to a relaxin analog or fragment that bind to a relaxin receptor and induces a relaxin-associated decrease in apoptosis.
  • Analogs or fragments of relaxin can be tested for the desired activity by procedures known in the art, including but not limited to the functional assays described herein.
  • the invention provides relaxin nucleic acid sequences for expression of relaxin polypeptide, fragments and analogs in vivo or in vitro.
  • the relaxin nucleic acid can be a vertebrate or mammalian relaxin, including, for example, human, mouse, rat, pig, cow, dog, or monkey relaxin.
  • the relaxin nucleic acids can comprise genomic nucleic acids, cDNA, the relaxin coding region or a fragment thereof. Relaxin nucleic acids further include mRNAs corresponding to the relaxin locus. Relaxin nucleic acids can also include analogs (e.g., nucleotide sequence variants), such as those encoding other possible codon choices for the same amino acid or conservative amino acid substitutions thereof, such as naturally occurring allelic variants.
  • nucleic acid sequences that encode substantially the same amino acid sequence as a relaxin cDNA or open reading frame, can be used in the practice of the present invention.
  • nucleic acid sequences include, but are not limited to, nucleic acid sequences comprising all or portions of a relaxin gene which is altered by the substitution of different codons that encode the same or a functionally equivalent amino acid residue (e.g., a conservative substitution) within the sequence, thus producing a silent change.
  • the invention further provides relaxin nucleic acid fragments of at least 6 contiguous nucleotides (e.g., a hybridizable portion); in other embodiments, the nucleic acids comprise at least 8 contiguous nucleotides, at least contiguous 25 nucleotides, at least contiguous 50 nucleotides, at least 100 nucleotides, or at least, 150 nucleotides, or more of a relaxin sequence. In another embodiment, the nucleic acids are smaller than 150 nucleotides in length.
  • the relaxin nucleic acids can be single or double-stranded.
  • nucleic acid encoding a fragment of a relaxin polypeptide is construed as referring to a nucleic acid encoding only the recited fragment or portion of the relaxin polypeptide and not the other contiguous portions of the relaxin polypeptide as a contiguous sequence. Fragments of relaxin nucleic acids encoding one or more relaxin domains are also provided.
  • Relaxin nucleic acids can be inserted into an appropriate vector, such as an expression vector (i.e., a vector which contains the necessary elements for the transcription and translation of the inserted polypeptide-coding sequence).
  • an expression vector i.e., a vector which contains the necessary elements for the transcription and translation of the inserted polypeptide-coding sequence.
  • the necessary transcriptional and translational signals can also be supplied by the native relaxin gene and/or its flanking regions.
  • a variety of host- vector systems can be utilized to express the relaxin nucleic acid sequences.
  • mammalian cell systems infected with virus e.g., vaccinia virus, adeno virus, adeno-associated virus, and the like
  • insect cell systems infected with virus e.g., baculo virus
  • microorganisms such as yeast containing yeast vectors, or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA.
  • the expression elements of vectors vary in their strengths and specificities. Depending on the host- vector system utilized, any one of a number of suitable transcription and translation elements can be used.
  • human relaxin nucleic acids, or a nucleic acid sequence encoding a functionally active portion of human relaxin is expressed in yeast or bacteria.
  • a fragment of relaxin comprising a domain of the relaxin polypeptide is expressed.
  • any of the methods known in the art for the insertion of nucleic acids into a vector can be used to construct expression vectors containing a chimeric gene consisting of appropriate transcriptional/translational control signals and relaxin nucleic acid sequences. These methods include in vitro recombinant DNA and synthetic techniques and in vivo recombinants (genetic recombination). Expression of nucleic acid sequence encoding a relaxin polypeptide, analog or fragment can be regulated by a second nucleic acid sequence so that the relaxin polypeptide, analog or fragment is expressed in a host transformed with the recombinant DNA molecule. For example, expression of a relaxin polypeptide can be controlled by any promoter/enhancer element known in the art.
  • Promoters which can be used to control relaxm gene expression include, but are not limited to, the SV40 early promoter region (Benoist and Chambon, Nature 290:304-10 (1981)), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al, Cell 22:787-97 (1980)), the he ⁇ es thymidine kinase promoter (Wagner et al, Proc. Natl. Acad. Sci.
  • the promoter of the photosynthetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al, Nature 310:115-20 (1984)), promoter elements from yeast or other fungi such as the Gall and Gal4 promoters, the ADH (alcohol dehydrogenase) promoter, the PGK (phosphoglycerol kinase) promoter, the alkaline phosphatase promoter, and the like.
  • mice mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells
  • the albumin gene control region which is active in liver
  • the alpha-fetoprotein gene control region which is active in liver
  • the tissue specific promoter is the prostate specific antigen promoter.
  • a vector is used that comprises a promoter operably linked to a relaxin nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic or drug resistance marker).
  • an expression construct can be made by subcloning a relaxin nucleic acid into a restriction site of the pRSECT expression vector.
  • a construct allows for the expression of a relaxin polypeptide, analog or fragment under the control of the T7 promoter with a histidine amino terminal flag sequence for affinity purification of the expressed polypeptide.
  • a vector is used that comprises the prostate specific antigen promoter operably linked to a relaxin nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an drug resistance marker).
  • Expression vectors containing relaxin nucleic acids can be identified by general approaches well known to the skilled artisan, including: (a) nucleic acid hybridization, (b) the presence or absence of "marker" gene function, (c) expression of inserted sequences, (d) by polymerase chain reaction (PCR), and the like.
  • PCR polymerase chain reaction
  • the presence of a relaxin nucleic acid inserted in an expression vector can be detected by nucleic acid hybridization using probes comprising sequences that are homologous to an inserted relaxin nucleic acid.
  • the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, colorimetric change, and the like) caused by the insertion of the relaxin nucleic acids into the vector.
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, colorimetric change, and the like
  • recombinant expression vectors can be identified by assaying the relaxin polypeptide, analog or fragment expressed by the recombinant. Such assays can be based, for example, on the physical or functional properties of the relaxin polypeptide, analog or fragment in in vitro assay systems (e.g., binding with anti-relaxin antibody, binding to relaxin receptor, and the like).
  • recombinant expression vectors can be identified by polymerase chain reaction. (See, e.g., U.S. Patent Nos. 4,683,202, 4,683,195 and 4,889,818; Gyllensten et al, Proc. Natl. Acad. Sci.
  • recombinant vectors can be propagated and prepared in quantity.
  • the vectors which can be used include, but are not limited to the following vectors or their analogs: human or animal viruses such as vaccinia virus or adenovirus; insect viruses such as baculovirus; yeast vectors; bacteriophage vectors (e.g., lambda); and plasmid and cosmid DNA vectors; to name but a few.
  • a host cell strain can be chosen that modulates the expression of the inserted nucleic acids, or modifies or processes the relaxin, relaxin analog or fragment in the specific fashion desired.
  • Expression from certain promoters can be elevated in the presence of certain inducers; thus, expression of the relaxin polypeptide, analog or fragment can be controlled.
  • different host cells having characteristic and specific mechanisms for the translational and post- translational processing, and modification (e.g., glycosylation and/or phosphorylation) can be used. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the polypeptide, analog or fragment expressed.
  • expression in a bacterial system can be used to produce an unprocessed core protein product.
  • Expression in mammalian cells can be used to ensure "native" processing of mammalian relaxin polypeptides, or of analogs or fragments.
  • different vector/host expression systems can affect processing reactions to different extents.
  • relaxin antagonists are provided for the modulation of apoptosis.
  • Relaxin antagonists can include, for example, relaxin binding agents, relaxin receptor binding agents, antisense nucleic acids, and the like.
  • Relaxin antagonists can comprise antibodies that immunospecifically- recognize relaxin or a relaxin receptor polypeptide and that stimulate apoptosis, and/or reduce or inhibit relaxin-associated cell accumulation in cell populations or tissues.
  • Anti-relaxin and anti-relaxin receptor antibodies include, but are not limited to, polyclonal antibodies, monoclonal antibodies, chimeric antibodies (e.g., fully humanized antibodies or human chimeric antibodies), single chain antibodies, antibody fragments (e.g., Fab, F(ab'), F(ab') 2 , Fv, or hypervariable regions), single heavy chains, and an Fab expression library.
  • polyclonal and/or monoclonal antibodies to full length, vertebrate or mammalian relaxin or relaxin receptor polypeptide are produced and selected for those antibodies that selectively bind to relaxin or a relaxin receptor polypeptides, and thereby functionally inactivate such polypeptides.
  • antibodies to a domain of a vertebrate relaxin polypeptide, or a relaxin receptor polypeptide are produced.
  • fragments of a vertebrate relaxin polypeptide, or a relaxin receptor polypeptide, which are identified as hydrophilic are used as immunogens for antibody production and selected for immunospecific binding to such a polypeptide and inhibition of its biological activity.
  • adjuvants can be used to increase the immunological response, depending on the host species, including but not limited to Freund's adjuvant (complete or incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Corynebacterium parvum bacille Calmette-Guerin
  • any technique which provides for the production of antibody molecules by continuous cell lines in culture can also be used.
  • Such techniques include, for example, the hybridoma technique originally developed by Kohler and Milstein (Nature 256:495-97 (1975)), as well as the trioma technique, the human B-cell hybridoma technique (see, e.g., Kozbor et al, Immunology Today 4:72 (1983)), and the EBV-hybridoma technique to produce human monoclonal antibodies (see, e.g. , Cole et al. , In Monoclonal Antibodies and Cancer Therapy, Alan R.
  • Mammalian antibodies can be used and can be obtained by using hybridomas (see, e.g., Cote et al, Proc. Natl. Acad. Sci. USA 80:2026-30 (1983)) or by transforming human B cells with EBV virus in vitro (see, e.g., Cole et al. (1985), supra). Selection of hybridomas producing antibodies with appropriate biological function are well known in the art or are described herein below. Human monoclonal antibodies can also be prepared by preparing hybridomas from animals having a human immune system that have been immunized by injection with the native relaxin or relaxin receptor polypeptide. (See, e.g., U.S. Patent No. 6,114,598 and 6,111, 166, which are inco ⁇ orated by reference herein.)
  • chimeric or “humanized” antibodies can be prepared.
  • Such chimeric antibodies are typically prepared by splicing the non-human genes for an antibody molecule specific for a relaxin or receptor polypeptide together with genes from a human antibody molecule of appropriate activity.
  • the antigen binding regions e.g., an F(ab') , F(ab'), Fv, or hypervariable region(s)
  • the antibodies are fully humanized.
  • a human monoclonal antibody or portions thereof can be identified by first screening a human B-cell cDNA library for DNA molecules that encode antibodies that specifically bind to a relaxin or a relaxin receptor polypeptide according to the method generally set forth by Huse et al. (Science 246:1275-81 (1989)). The DNA molecule can then be cloned and amplified to obtain sequences that encode the antibody (or binding domain) of the desired specificity.
  • Phage display technology offers another technique for selecting antibodies that bind to relaxin or relaxin receptor polypeptides, fragments or analogs thereof. (See, e.g., International Patent Publications WO 91/17271 and WO 92/01047; and Huse et al, supra.)
  • techniques for the production of single chain antibodies can be adapted to produce relaxin- or relaxin receptor-specific single chain antibodies.
  • An additional aspect of the invention utilizes the techniques described for the construction of a Fab expression library (see, e.g., Huse et al. (1989), supra) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity for relaxin polypeptides, fragments, or analogs thereof and biological activity.
  • Antibody fragments which contain the idiotype of the molecule can be generated by known techniques.
  • such fragments include, but are not limited to, an F(ab') fragment which can be produced by pepsin digestion of the antibody molecule, Fab' fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragment, Fab fragments which can be generated by treating the antibody molecule with papain and a reducing agent, and Fv fragments.
  • Recombinant Fv fragments can also be produced in eukaryotic cells using, for example, the methods described in U.S. Patent No. 5,965,405.
  • antibodies which recognize a specific domain of a relaxin or relaxin receptor polypeptide can be used to assay hybridomas for a product (e.g., antibody) that binds to a relaxin or relaxin receptor fragment containing that domain.
  • a product e.g., antibody
  • For selection of an antibody that specifically binds to a first relaxin or relaxin receptor polypeptide, but which does not specifically bind a second, different relaxin polypeptide one can select on the basis of antibody-positive binding to the first polypeptide and a lack of antibody binding to the second, different polypeptide.
  • the relaxin antagonist is a relaxin binding agent comprising a soluble relaxin receptor, or a fragment or analog thereof, that binds relaxin.
  • soluble relaxin receptor refers to a relaxin receptor polypeptide that is not bound to a cell membrane.
  • the relaxin receptor is approximately 200 kilodaltons. (See Palejwala et al, Endocrinology 139(3):1208-12 (1998), the disclosure of which is inco ⁇ orated by reference herein.)
  • the soluble form of the relaxin receptor retains the ability to bind vertebrate relaxin, but typically lacks transmembrane and/or cytoplasmic domains.
  • Soluble relaxin receptors can comprise additional amino acid residues, such as affinity tags, that provide for a means for purification of the polypeptide or to provide sites for attachment of the polypeptide to another polypeptide, or to immuno globulin sequences.
  • the soluble relaxin receptor can optionally contain a transmembrane domain that cannot associate with a cell membrane.
  • transmembrane domain is meant a domain of the relaxin receptor polypeptide that contains a sufficient number of hydrophobic amino acids to allow the polypeptide to insert and anchor in a cell membrane.
  • transmembrane domain that cannot associate with a cell membrane is meant a transmembrane domain that has been altered by mutation or deletion such that it is not sufficiently hydrophobic to allow insertion or other association with a cell membrane.
  • a transmembrane domain does not preclude, for example, the fusion of the relaxin receptor polypeptide, or fragment thereof, with a secretion signal sequence useful for secretion of the polypeptide from the cell.
  • Substitutions or alterations of the amino acid sequence useful to achieve an inactive transmembrane domain include, but are not limited to, deletion or substitution of amino acids within the transmembrane domain.
  • the soluble relaxin receptors include soluble, naturally-occurring amino acid sequence variants of relaxin receptor. Soluble relaxin receptors further include those altered by substitution, addition or deletion of one or more amino acid residues that provide for functionally active relaxin receptor polypeptides. Such relaxin receptors include, but are not limited to, those containing as a primary amino acid sequence of all or part of the amino acid sequence of a relaxin receptor polypeptide including sequences in which one or more functionally equivalent amino acid residues are substituted for residues within the sequence, resulting in a silent functional change (e.g., a conservative substitution).
  • the soluble relaxin receptor is a polypeptide consisting of or comprising a fragment of a relaxin receptor polypeptide having at least 10 contiguous amino acids of the relaxin receptor polypeptide. More typically, the fragment contains at least 20 or at least 50 contiguous amino acids of the relaxin receptor polypeptide. In other embodiments, the fragments are larger than 100 or even 200 amino acids.
  • the relaxin receptor polypeptide can be a polypeptide comprising regions that are substantially similar to a relaxin receptor polypeptide or fragments thereof (e.g., in various embodiments, at least 60%, 70%, 75%, 80%, 90%, or even 95% identity or similarity over an amino acid sequence of identical size), or when compared to an aligned sequence in which the alignment is done by a computer sequence comparison/alignment program known in the art, or by visual inspection.
  • a computer sequence comparison/alignment program known in the art, or by visual inspection.
  • Sequence identity or similarity can also be determined by identifying nucleic acids that are capable of hybridizing to a relaxin receptor nucleic acid, under high stringency, moderate stringency, or low stringency conditions.
  • Soluble relaxin receptors and fragments thereof can be produced by various methods known in the art. The manipulations which result in their production can occur at the gene or protein level.
  • cloned relaxin receptor nucleic acids can be modified by any of numerous strategies known in the art (see, e.g., Sambrook et al, supra; Ausubel et al, supra), such as making conservative substitutions, deletions, insertions, and the like.
  • the sequence can be cleaved at appropriate sites with restriction endonuclease(s), followed by further enzymatic modification if desired, isolated, and ligated in vitro.
  • the modified nucleic acid typically remains in the proper translational reading frame, so that the reading frame is not interrupted by translational stop signals or other signals which interfere with the synthesis of the soluble relaxin receptor or fragment thereof.
  • the relaxin receptor nucleic acid can also be mutated in vitro or in vivo to create and/or destroy translation initiation and/or termination sequences.
  • the relaxin receptor nucleic acid can also be mutated to create variations in coding regions (e.g., amino acid substitutions) and/or to form new restriction endonuclease sites or destroy preexisting ones and to facilitate further in vitro modification.
  • Any technique for mutagenesis known in the art can be used, including but not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (see, e.g., Hutchison et al, J. Biol Chem. 253:6551-60 (1978)), the use of TAB ® linkers (Pharmacia), and the like.
  • relaxin receptor polypeptide sequence can also be made at the polypeptide level. Included within the scope of the invention are relaxin receptor polypeptides that are differentially modified during or after synthesis (e.g., in vivo or by in vitro translation). Such modifications include conservative substitution, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage. to an antibody molecule, a protein or other cellular ligand, and the like.
  • any of numerous chemical modifications can be carried out by known techniques, including, but not limited to, specific chemical cleavage (e.g., by cyanogen bromide), enzymatic cleavage (e.g., by trypsin, chymotrypsin, papain, V8 protease, and the like); modification by, for example, NaBH 4 , acetylation, formylation, oxidation and reduction, metabolic synthesis in the presence of tunicamycin, and the like.
  • Relaxin receptor polypeptides and fragments thereof can be purified from natural sources by standard methods such as those described herein (e.g., immunoaffinity purification).
  • Relaxin receptor polypeptides and fragments can also be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, sizing column chromatography, high pressure liquid chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of polypeptides.
  • Relaxin receptor polypeptides and fragments thereof can be synthesized by standard chemical methods known in the art (see, e.g., Hunkapiller et al, Nature 310:105-11 (1984); Stewart and Young, Solid Phase Peptide Synthesis, 2 nd Ed., Pierce Chemical Co., Rockford, IL, (1984)).
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the relaxin receptor polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, the D-isomers of the common amino acids, ⁇ -amino isobutyric acid, 4-aminobutyric acid, 2-amino butyric acid, ⁇ -amino hexanoic acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3 -amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, selenocysteine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl
  • the soluble relaxin receptor is a chimeric, or fusion, protein comprising a relaxin receptor polypeptide, or fragment thereof
  • such a chimeric protein is produced by recombinant expression of a nucleic acid encoding the chimeric polypeptide.
  • the chimeric product can be made by ligating the appropriate nucleic acid sequences, encoding the desired amino acid sequences, to each other in the proper reading frame and expressing the chimeric product by methods commonly known in the art.
  • the chimeric product can be made by protein synthetic techniques (e.g., by use of an automated peptide synthesizer).
  • the fusion protein is a relaxin-receptor-ubiquitin fusion protein.
  • the relaxin antagonist can further be a relaxin analog, such as a relaxin polypeptide that binds to a relaxin receptor but fails to induce a response by that receptor.
  • the relaxin analog can be a competitive inhibitor of relaxin binding or a conventional antagonist of the relaxin receptor.
  • the term "relaxin" refers to vertebrate relaxin polypeptides, including full length relaxin polypeptide or a portion of the relaxin polypeptide that retains biological activity. Relaxin has been well defined in its natural human form, animal form, and in its synthetic form, hi particular, relaxin has been extensively described in U.S. Patent Nos.
  • Relaxin analogs can be prepared by modification of relaxin polypeptides such that the relaxin analog retains relaxin receptor binding activity, but does not induce a response by the relaxin receptor.
  • relaxin analogs can be amino acid sequence variants of relaxin that retain relaxin receptor binding activity, but that fail to induce a response by a relaxin receptor.
  • Relaxin analogs further include relaxm polypeptides, altered by addition or deletion of one or more amino acid residues, that retain receptor-binding function but fail to induce a response by relaxm receptor.
  • the relaxin analog is fragment of a relaxin polypeptide consisting of or comprising at least 10 contiguous amino acids of the relaxin polypeptide.
  • the fragment contains at least 20 or 40 contiguous amino acids of the relaxin polypeptide. In other embodiments, the fragments are not larger than 35 amino acids.
  • the relaxin analog can be a polypeptide comprising regions that are substantially similar to a relaxin polypeptide (e.g., in various embodiments, at least 60%, 70%, 75%, 80%, 90%, or even 95% identity or similarity over an amino acid sequence of identical size), or when compared to an aligned sequence in which the alignment is done by a computer sequence comparison/alignment program known in the art, or which coding nucleic acid is capable of hybridizing to a relaxin nucleic acid, under high stringency, moderate stringency, or low stringency conditions. (See supra)
  • Relaxin analogs can be produced by various methods known in the art. The manipulations which result in their production can occur at the gene or protein level. For example, cloned relaxin nucleic acids can be modified by any of numerous strategies known in the art (see, e.g., Sambrook et al, supra; Ausubel et al, supra), such as by making conservative or non-conservative substitutions, deletions, insertions, and the like. The sequence can be cleaved at appropriate sites with restriction endonuclease(s), followed by further enzymatic modification, if desired, isolated, and ligated in vitro.
  • the modified nucleic acid typically remains in the proper translational reading frame, so that the reading frame is not interrupted by translational stop signals or other signals which interfere with the synthesis of the relaxin analog.
  • the relaxin nucleic acid can be mutated in vitro or in vivo to create and or destroy translation, initiation and/or termination sequences.
  • the relaxin nucleic acid can also be mutated to create variations in coding regions and/or to form new restriction endonuclease sites or destroy preexisting ones and to facilitate further in vitro modification.
  • mutagenesis Any technique for mutagenesis known in the art can be used, including but not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (see, e.g., Hutchison et al, J. Biol. Chem. 253:6551-60 (1978)), the use of TAB ® linkers (Pharmacia), and the like.
  • relaxin analogs are prepared from relaxin- encoding nucleic acids that are altered to introduce aspartic acid codons at specific position(s) within at least a portion of the relaxin coding region.
  • the resulting analogs can be treated with dilute acid to release a desired analog, thereby rendering the protein more readily isolated and purified.
  • relaxin polypeptide sequence can also be made at the polypeptide level. Included within the scope of the invention are relaxin analogs that are differentially modified during or after synthesis (e.g., in vivo or by in vitro translation). Such modifications include amino acid substitution (either conservative or non-conservative), glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule, a protein or other cellular ligand, and the like.
  • any of numerous chemical modifications can be carried out by known techniques, including, but not limited to, specific chemical cleavage (e.g., by cyanogen bromide), enzymatic cleavage (e.g., by trypsin, chymotrypsin, papain, V8 protease, and the like); modification by, for example, NaBH , acetylation, formylation, oxidation and reduction, metabolic synthesis in the presence of tunicamycin, and the like.
  • specific chemical cleavage e.g., by cyanogen bromide
  • enzymatic cleavage e.g., by trypsin, chymotrypsin, papain, V8 protease, and the like
  • Relaxin analogs can be purified from natural sources by standard methods such as those described herein (e.g., immunoaffmity purification). Relaxin analogs can also be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, sizing column chromatography, high pressure liquid chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of polypeptides. Relaxin analogs can be synthesized by standard chemical methods known in the art (see, e.g., Hunkapiller et al, Nature 310:105-11 (1984); Stewart and Young, Solid Phase Peptide Synthesis, 2 nd Ed., Pierce Chemical Co., Rockford, IL, (1984)).
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the relaxin polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, the D-isomers of the common amino acids, ⁇ -amino isobutyric acid, 4-aminobutyric acid, 2-amino butyric acid, ⁇ -amino hexanoic acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, selenocysteine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids,
  • the relaxin analog is a chimeric, or fusion, protein comprising a relaxin polypeptide (typically consisting of at least a domain or motif of the relaxin polypeptide, or at least 10 contiguous amino acids of the relaxin polypeptide) joined at its amino- or carboxy-terminus via a peptide bond to an amino acid sequence of a different protein.
  • a chimeric protein is produced by recombinant expression of a nucleic acid encoding the chimeric polypeptide.
  • the chimeric product can be made by ligating the appropriate nucleic acid sequences, encoding the desired amino acid sequences, to each other in the proper reading frame and expressing the chimeric product by methods commonly known in the art.
  • the chimeric product can be made by protein synthetic techniques (e.g., by use of an automated peptide synthesizer).
  • the fusion protein is a relaxin analog- ubiquitin fusion protein.
  • U.S. Patent No. 5,108,919 discloses methods for preparing a fusion protein of a relaxin chain and ubiquitin.
  • Nucleic Acids The invention further provides nucleic acids for use as an antagonist, or for expressing an antagonist, according to the present invention. Such nucleic acids include those encoding a soluble relaxin receptor or a relaxin analog for synthesis of soluble relaxin receptor or relaxin analog, respectively. Antisense nucleic acids are provided for inhibition of relaxin or relaxin receptor expression. Nucleic acids encoding a relaxin polypeptide or a relaxin receptor polypeptide are also provided for the preparation of antibodies (see supra). In one aspect, the invention provides nucleic acid sequences encoding a relaxin receptor or relaxin analog for expression in vivo. The relaxin receptor or relaxin analog, or antisense nucleic acids, can be expressed in vivo for gene therapy.
  • Relaxin receptor, relaxin analog or antisense nucleic acids can also be expressed in vivo or in vitro for the production of recombinant soluble relaxin receptor, relaxin analog or antisense nucleic acids for exogenous administration to a subject.
  • the nucleic acids can be vertebrate nucleic acid, including, for example, human, mouse, rat, pig, cow, dog, or monkey relaxin receptor, relaxin, or a relaxin analog derived from a vertebrate relaxin.
  • the nucleic acids can comprise genomic DNA, cDNA, or the coding region of the relaxin receptor, relaxin or a relaxin analog.
  • the nucleic acids can further include mRNAs corresponding to the relaxin receptor locus or the relaxin locus.
  • the nucleic acids also include nucleotide sequence variants, such as those encoding other possible codon choices for the same amino acid or conservative amino acid substitutions thereof, such as naturally occurring allelic variants. Due to the degeneracy of nucleotide coding sequences, other nucleic acid sequences which encode substantially the same amino acid sequence as a relaxin receptor or relaxin coding sequence, can be used in the practice of the present invention. These nucleic acid sequences include, but are not limited to, nucleotide sequences comprising all or portions of a relaxin gene which is altered by the substitution of different codons that encode the same or a functionally equivalent amino acid residue (e.g., a conservative substitution) within the sequence, thus producing a silent change.
  • nucleotide sequence variants such as those encoding other possible codon choices for the same amino acid or conservative amino acid substitutions thereof, such as naturally occurring allelic variants. Due to the degeneracy of nucleotide coding sequences, other nucleic acid sequences which
  • the invention further provides nucleic acid fragments of at least 6 contiguous nucleotides (e.g., a hybridizable portion); in other embodiments, the nucleic acids comprise at least 8 contiguous nucleotides, at least contiguous 25 nucleotides, at least contiguous 50 nucleotides, at least 100 nucleotides, 150 nucleotides or more of a relaxin sequence. In another embodiment, the nucleic acids are smaller than 100 or 150 nucleotides in length. The nucleic acids can be single or double-stranded.
  • nucleic acid encoding a fragment of a relaxin or relaxin receptor polypeptide is construed as referring to a nucleic acid encoding only the recited fragment or portion of the relaxin polypeptide and not the other contiguous portions of the relaxin receptor or relaxin polypeptide as a contiguous sequence. Fragments of the nucleic acids encoding one or more domains of relaxin or relaxin receptor are also provided.
  • Relaxin receptor, relaxin analog or antisense nucleic acids can be inserted into an appropriate vector (e.g. , an expression vector which contains the necessary elements for the transcription or transcription and translation of the inserted polypeptide-coding sequence) in either the sense or antisense orientations, as desired.
  • the necessary transcriptional and translational signals can also be supplied by the native relaxin or relaxin receptor gene and/or its flanking regions.
  • host- vector systems can be utilized to express the polypeptide-coding sequence.
  • mammalian cell systems infected with virus e.g., vaccinia virus, adenovirus, adeno-associated virus, and the like
  • insect cell systems infected with virus e.g., baculovirus
  • microorganisms such as yeast containing yeast vectors, or bacteria transformed with bacteriophage, DNA, plasmid DNA, or cosmid DNA.
  • the expression elements of vectors vary in their strengths and specificities. Depending on the host- vector system utilized, any of a number of suitable transcription and translation elements can be used.
  • the nucleic acids are expressed, or a nucleic acid sequence encoding a functionally active portion of a relaxin receptor or relaxin analog is expressed in mammalian cells, yeast or bacteria.
  • a fragment of a relaxin receptor or relaxin analog comprising a domain of the respective polypeptide is expressed.
  • Any of the methods known in the art for the insertion of nucleic acids into a vector can be used to construct expression vectors containing a chimeric gene having the appropriate transcriptional, translational control signals and/or polypeptide coding sequences. These methods include in vitro recombinant DNA and synthetic techniques and in vivo recombinants (genetic recombination).
  • nucleic acids encoding a relaxin receptor or relaxin analog can be regulated by a second nucleic acid sequence so that the nucleic acid or polypeptide is expressed in a host transformed with the recombinant DNA molecule.
  • expression of a nucleic acid of a relaxin receptor or relaxin analog can be controlled by any promoter/enhancer element known in the art.
  • Promoters which can be used to control expression include, but are not limited to, the SV40 early promoter region (see, e.g., Benoist and Chambon, Nature 290:304-10 (1981)), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (see, e.g., Yamamoto et al, Cell 22:787-97 (1980)), the he ⁇ es thymidine kinase promoter (see, e.g., Wagner et al, Proc. Natl. Acad. Sci.
  • the SV40 early promoter region see, e.g., Benoist and Chambon, Nature 290:304-10 (1981)
  • the promoter contained in the 3' long terminal repeat of Rous sarcoma virus see, e.g., Yamamoto et al, Cell 22:787-97 (1980)
  • the he ⁇ es thymidine kinase promoter see, e.g., Wagner et
  • the regulatory sequences of the metallothionen gene see, e.g., Brinster et al, Nature 296:39-42 (1982)
  • prokaryotic expression vectors such as the ⁇ -lactamase promoter (see, e.g., Villa- Komaroff et al, Proc. Natl. Acad. Sci. USA 75:3727-31 (1978)) or the tac promoter (see, e.g., deBoer et al, Proc. Natl Acad. Sci. USA 80:21-25 (1983)
  • plant expression vectors including the cauliflower mosaic virus 35S RNA promoter (see, e.g., Gardner et al, Nucl Acids Res.
  • promoter of the photosynthetic enzyme ribulose biphosphate carboxylase see, e.g., Herrera-Estrella et al, Nature 310:115-20 (1984)
  • promoter elements from yeast or other fungi such as the Gal7 and Gal4 promoters, the ADH (alcohol dehydrogenase) promoter, the PGK (phosphoglycerol kinase) promoter, the alkaline phosphatase promoter, and the like.
  • a vector in another embodiment, comprises a promoter operably linked to a nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic or drug resistance marker).
  • an expression construct can be made by subcloning a relaxin receptor or relaxin analog nucleic acid into a restriction site of the pRSECT expression vector. Such a construct allows for the expression of the relaxin analog or relaxin receptor polypeptide under the control of the T7 promoter with a histidine amino terminal flag sequence for affinity purification of the expressed polypeptide.
  • a vector in another specific embodiment, comprises the prostate specific antigen promoter operably linked to a relaxin analog nucleic acid, one or more origins of replication, and, optionally, one or more selectable markers (e.g., a drug resistance marker).
  • Expression vectors containing such nucleic acids can be identified by general approaches well known to the skilled artisan, including: (a) nucleic acid hybridization or polymerase chain reaction, (b) the presence or absence of "marker” gene function, (c) expression of inserted sequences, or polymerase chain reaction (PCR).
  • the presence of a nucleic acid inserted in an expression vector can be detected by nucleic acid hybridization or polymerase chain reaction using probes comprising sequences that are homologous to an inserted nucleic acid.
  • the recombinant vector/host system can be identified and selected based upon the presence or absence of certain "marker" gene functions (e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, and the like) caused by the insertion of a vector containing the relaxin receptor, relaxin or relaxin analog nucleic acids.
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, and the like
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body formation in baculovirus, and the like
  • certain "marker" gene functions e.g., thymidine kinase activity, resistance to antibiotics, transformation phenotype, occlusion body
  • Such assays can be based, for example, on the physical or functional properties of the polypeptide in in vitro assay systems (e.g., binding with anti-relaxin antibody, binding relaxin or a relaxin analog, binding to relaxin receptor, and the like).
  • binding with anti-relaxin antibody, binding relaxin or a relaxin analog, binding to relaxin receptor, and the like e.g., binding with anti-relaxin antibody, binding relaxin or a relaxin analog, binding to relaxin receptor, and the like.
  • the expression vectors which can be used include, but are not limited to the following vectors or their derivatives: human or animal viruses such as vaccinia virus or adenovirus; insect viruses such as baculovirus; yeast vectors; bacteriophage vectors (e.g., lambda), and plasmid and cosmid DNA vectors, to name but a few.
  • human or animal viruses such as vaccinia virus or adenovirus
  • insect viruses such as baculovirus
  • yeast vectors bacteriophage vectors (e.g., lambda), and plasmid and cosmid DNA vectors, to name but a few.
  • bacteriophage vectors e.g., lambda
  • plasmid and cosmid DNA vectors to name but a few.
  • PCR is used to detect the nucleic acid in the vector (see supra).
  • a host cell strain can be chosen that modulates the expression of the inserted sequences, or modifies or processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus, expression of the polypeptide can be controlled.
  • different host cells having characteristic and specific mechanisms for the translational and post-translational processing, and modification (e.g., glycosylation, phosphorylation) of polypeptides can be used. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. For example, expression in a bacterial system can be used to produce an unprocessed core protein product. Expression in mammalian cells can be used to ensure "native" processing of mammalian receptor polypeptide. Furthermore, different vector/host expression systems can affect processing reactions to different extents. Functional Assays for Relaxin and Relaxin Receptor Agonists and Antagonists
  • the activity of relaxin agonists and antagonists, and of relaxin receptor agonists and antagonists can be determined by standard assays for relaxin and/or relaxin receptor activity.
  • the activity of a relaxin agonist is assayed. For example, the ability of a relaxin agonist to decrease apoptosis, or to stimulate maturation of tissue, is assayed.
  • the ability of a relaxin antagonist to inhibit relaxin functional activity by binding to relaxin is assayed.
  • the ability of a relaxin antagonist to inhibit relaxin function, or relaxin receptor function can be assayed by, for example, adding a relaxin antagonist to a relaxin receptor assay and determining the inhibition, as compared with a control without the relaxin antagonist. Suitable measurements of relaxin antagonist activity include measuring percent inhibition, IC 50 , and the like.
  • Suitable assays for measuring relaxin or relaxin receptor agonist or antagonist activity include, for example, those described in the following references (which are inco ⁇ orated by reference herein): MacLennan et al, Ripening of the Human Cervix and Induction of Labor with Intracervical Purified Porcine Relaxin, Obstetrics & Gynecology 68:598-601 (1986); Poisner et al, Relaxin Stimulates the Synthesis and Release of Prorenin From Human Decidual Cells: Evidence For
  • the activity of an agonist or antagonist is determined by measuring the ability of the agonist or antagonist to compete with wild-type relaxin polypeptide, or relaxin receptor polypeptide, for binding to anti-relaxin antibody.
  • Various immunoassays known in the art can be used.
  • Such assays include, but are not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay) "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitin reactions, immunodiffusion assays, in situ immunoassays (using .
  • Antibody binding can be detected by measuring the amount of label on the primary antibody that is bound, or prevented from binding to, a substrate. Alternatively, primary antibody binding is detected by measuring binding of a secondary antibody or reagent to the primary antibody. The secondary antibody can also be directly labeled. Many means are known in the art for detecting binding in an immunoassay and are considered within the scope of the present invention.
  • the functional activity of an agonist or antagonist can also be determined in an in vivo system. For example, the ability of relaxin agonists or antagonists to bind, or to compete for binding to, a relaxin receptor, or to modulate apoptosis in a cell population and/or tissues can be measured.
  • the assays described above can be used to determine the activity resulting from expression of relaxin agonist or antagonists in vertebrate cells.
  • relaxin agonist or antagonist can be expressed in a heterologous system and the activity of the relaxin agonist or antagonist can be assayed as a modulator of a physiological change in that system.
  • the ability of a relaxin agonist or antagonist to modulate apoptosis can be tested in vertebrate cells (e.g. , transfected mammalian cells).
  • the invention provides methods for the administration to a subject of an effective amount of a relaxin agonist or antagonist (also referred to collectively as an "active agent").
  • a relaxin agonist or antagonist also referred to collectively as an "active agent”
  • the active agent is substantially purified prior to formulation.
  • the subject can be a human or non-human animal, a vertebrate, and is typically an animal, including but not limited to, cows, pigs, horses, chickens, cats, dogs, and the like. More typically, the subject is a mammal, and in a particular embodiment, human.
  • a active agent such as, for example, by infusion, injection (e.g., intradermal, intramuscular or intraperitoneal), oral delivery, nasal delivery, intrapulmonary delivery, rectal delivery, transdermal delivery, interstitial delivery or subcutaneous delivery.
  • injection e.g., intradermal, intramuscular or intraperitoneal
  • oral delivery e.g., nasal delivery, intrapulmonary delivery, rectal delivery, transdermal delivery, interstitial delivery or subcutaneous delivery.
  • administration can be achieved by, for example, and not by way of limitation, local infusion, topical application, by injection (e.g., intratesticular or intraprostatic), by means of a catheter, or by means of an implant, the implant being for example, a porous, non-porous, gelatinous or polymeric material, including membranes such as silastic membranes or fibers.
  • administration can be by direct injection at the target site.
  • compositions containing the active agent can be formulated according to the desired delivery system.
  • Such phannaceutical compositions typically comprise a therapeutically effective amount of active agent and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in vertebrates, typically animals, and more typically in humans.
  • carrier refers to a diluent, adjuvant, excipient, stabilizer, preservative, viscogen, or vehicle with which the active agent is formulated for administration.
  • Pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. Suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, ethanol, and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Suitable preservatives include, for example, sodium benzoate, quaternary ammonium salts, sodium azide, methyl paraben, propyl paraben, sorbic acid, ascorbylpalmitate, butylated hydroxyanisole, butylated hydroxytoluene, chlorobutanol, dehydroacetic acid, ethylenediamine, potassium benzoate, potassium metabisulfite, potassium sorbate, sodium bisulfite, sulfur dioxide, organic mercurial salts, phenol and ascorbic acid.
  • Suitable viscogens include, for example, carboxymethylcellulose, sorbitol, dextrose, and polyethylene glycols.
  • suitable pharmaceutical carriers are described in, for example, Remington 's Pharmaceutical Sciences (Gennaro (ed.), Mack Publishing Co., Easton, Pennsylvania (1990)).
  • the active agents can also be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, and the like, and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, and the like.
  • the active agent is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • a pharmaceutical composition adapted for intravenous administration to human beings.
  • water is a typical carrier.
  • Saline, aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition can also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water- free concentrate in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • Orally deliverable compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained-release formulations, and the like.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like.
  • the compositions are formulated according to standard pharmaceutical procedures.
  • the composition is formed as a meltable composition, such as a suppository.
  • Suppositories can contain adjuvants which provide the desired consistency to the composition. They can also contain water-soluble carriers, such as polyethylene glycol, polypropylene glycol, glycerogelatine, methylcellulose or carboxymethylcellulose.
  • Wetting agents such as fatty acids, fatty acid glycerides, polyoxyethylene sorbitan fatty acid esters, polyoxyethylene sorbitol fatty acid esters, polyoxyethylene fatty acid esters, as well as higher alcohol esters of polyoxyethylene and esters of lower alkylsulfonic acids.
  • the suppositories can also contain suitable emulsifying and dispersing components as well as components for adjusting the viscosity and coloring substances.
  • Nasal administration is typically performed using a solution as a nasal spray and can be dispensed by a variety of methods known to those skilled in the art. Systems for intranasally dispensing liquids as a spray are well known (see, e.g., U.S. Patent No. 4,511,069, which is inco ⁇ orated by reference herein).
  • Preferred nasal spray solutions comprise the active agent in a liquid carrier that optionally includes a nonionic surfactant for enhancing abso ⁇ tion of the drug and one or more buffers or other additives to minimize nasal irritation.
  • the nasal spray solution further comprises a propellant.
  • the pH of the nasal spray solution is typically between about pH 6.8 and 7.2.
  • ingredients which improve the abso ⁇ tion of nasally administered active agent and reduce nasal irritation, especially when used in a chronically administered treatment protocol are desirable.
  • the utilization of surfactants to enhance abso ⁇ tion of the active agent is prefened.
  • Nasal administration of drugs enhanced by surfactants can cause nasal irritation, including stinging, congestion and rhinorrhea.
  • compositions which enhance abso ⁇ tion through the nasal mucosa with reduced irritation are desirable, such as, for example, nonionic surfactants such as nonoxynol-9, laureth-9, poloxamer-124, octoxynol-9 and lauramide DEA.
  • nonionic surfactants such as nonoxynol-9, laureth-9, poloxamer-124, octoxynol-9 and lauramide DEA.
  • Nonoxynol-9 (N-9) is an ethoxylated alkyl phenol, the polyethyleneoxy condensate of nonylphenol with 9 moles of ethylene oxide.
  • This surfactant has been used in detergent products and is sold under trade names such as SURFONIC ® N-95 (Jefferson), NEUTRONYX ® 600 (Onyx) and IGEPAL ® (CO-630 (GAF).
  • N-9 is considered to be a hard detergent, and has been used as a spermatocide.
  • one or more anti-irritant additives are included in the emulsion.
  • polysorbate-80 has been shown to reduce the irritation caused by intranasally administered drugs where delivery was enhanced by use of a nonionic surfactant (see, e.g., U.S. Patent No. 5,902,789, which is inco ⁇ orated by reference herein).
  • Intrapulmonary dosage forms containing the active agent can be administered to the respiratory tract intranasally or by breathing a spray or aerosol containing the active agent.
  • the active agent is typically delivered directly into the lungs in a small particle aerosol, which is specifically targeted to the smallest air passages and alveoli.
  • Intrapulmonary dosage forms are typically formed as particulate dispersed forms. This can be accomplished by preparing an aqueous aerosol of solid particles which contain the composition. Typically, an aqueous aerosol is made by formulating an aqueous solution or suspension of the composition together with conventional pharmaceutically acceptable carriers and stabilizers.
  • the carriers and stabilizers typically include nonionic surfactants (e.g., Tweens, Pluronics or polyethylene glycol), innocuous proteins such as serum albumin, sorbitan esters, oleic acid, amino acids such as glycine, buffers, salts, sugars or sugar alcohols.
  • the formulations can also include mucolytic agents, such as those described in U.S. Patent No.
  • Aerosols are generally prepared from isotonic solutions.
  • the particles optionally include normal lung surfactant proteins.
  • the aerosol of particles can be formed in aqueous or nonaqueous (e.g., fluorocarbon propellant) suspensions.
  • the aerosols are preferably free of lung irritants (i.e., substances that cause acute bronchoconstriction, coughing, pulmonary edema or tissue destruction).
  • Nonirritating, abso ⁇ tion enhancing agents are also suitable for use herein.
  • Sonic nebulizers can be used to prepare aerosols. Sonic nebulizers minimize exposure of the composition to shear, which can result in degradation.
  • a suitable device is the Bird Micronebulizer.
  • Other suitable atomizing or nebulizing systems or intratracheal delivery systems include, for example, those disclosed in U.S. Patent No. 3,915,165; European Patent No. 0 166 476; the jet nebulizers described by Newman et al. (Thorax 40:671-76 (1985)); metered dose inhalers (see, e.g., Berenberg, J. Asthma-USA 22:87-92 (1985)); the endotracheal catheter assembly of Braunner (U.S. Patent No.
  • the particulate aerosol suspensions are typically fine dry powders containing the active agent.
  • Particulate aerosol suspension are prepared by any number of conventional procedures. The simplest method of preparing such suspensions is to micronize the active agent (e.g., as crystals or lyophilization cakes), and suspend the particles in dry fluorocarbon propellants. In these formulations the active agent is preferably suspended in the fluorocarbon. In an alternate embodiment, the active agent is stored in a compartment separate from the propellant. Discharge of the propellant withdraws a predetermined dose from the storage compartment.
  • the devices used to deliver active agents in this manner are known as metered dose inhalers (MDIs) (see, e.g., Byron, Drug Development and Industrial Pharmacy 12:993 (1986), which is inco ⁇ orated by reference herein).
  • MDIs metered dose inhalers
  • the size of the aerosols or particles generally will range about from 0.5 to about 5 ⁇ m, typically about 2 ⁇ m to 5 ⁇ m, preferably about 2 to about 4 ⁇ m or about 4 to about 5 ⁇ m. In some aspects, smaller particles are less acceptable because they tend not to be deposited, but instead are exhaled.
  • larger particles are not prefened because they are less likely to be deposited in the alveoli, being removed by impaction within the nasopharyngeal or oral cavities (see, e.g., Byron, J. Pharni. Sci. 75:433 (1986)).
  • the aerosol or particulate compositions can be heterogeneous in size distribution, although heterogeneity can be reduced by known methods (e.g., the screening unit described in EP 0 135 390). Heterogeneity is typically not disadvantageous unless the proportion of particles having an average mean diameter in excess of about 4 ⁇ m is so large as to impair the delivery of a therapeutic dose by pulmonary inhalation.
  • Suspensions containing greater than about 15% of particles within the 0.5-5 ⁇ m range can be used, but generally the proportion of particles having an average mean diameter larger than 5 ⁇ m is typically less than about 25%, and preferably not greater than 10%, of the total number of particles.
  • the diameters recited refer to the particle diameters as introduced into the respiratory tract.
  • the amount of the active agent which will be effective in the treatment of a particular subject will depend on the specific abnormality being treated, and can be determined by standard clinical techniques. In addition, in vitro assays can optionally be employed to help identify optimal dosage ranges. The precise dose of the active agent to be employed in the formulation will also depend on the route of administration, and the seriousness of the condition, and should be decided according to the judgment of the practitioner and each subject's circumstances.
  • Suitable dosage ranges for administration are generally about 0.001 mg/kg to about 100 mg/kg of active agent per kilogram body weight. Effective doses can also be extrapolated from dose response curves derived from in vitro or animal model test systems. Suppositories generally contain active ingredient in the range of 0.5% to 10% by weight; oral formulations typically contain 10% to 95% active ingredient.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the active agent can be delivered in a controlled release system.
  • a pump can be used (see, e.g., Langer, supra; Sefton, Crit. Ref Biomed. Eng. 14:201-40 (1987); Buchwald et al, Surgery 88:507-16 (1980); Saudek et al, N Engl J. Med. 321 :574-79 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J Macromol Sci. Rev. Macromol Chem. 23:61 (1983); see also Levy et al, Science 228:190-92 (1985); During et al, Ann. Neurol 25:351-56 (1989); Howard et al, J. Neurosurg. 71:105-12 (1989)) (the disclosures of which are inco ⁇ orated by reference herein).
  • a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, supra, Vol. 2, pp. 115-38 (1984)).
  • Other controlled release systems are discussed in, for example, the review by Langer (Science 249:1527-33 (1990), which is inco ⁇ orated by reference herein).
  • nucleic acids e.g., nucleic acids encoding relaxin agonists or antagonists, such as relaxins, relaxin analogs, soluble relaxin receptor, relaxin binding agents, relaxin receptor binding agents, relaxin antisense nucleic acids and/or relaxin receptor antisense nucleic acids
  • Nucleic acids both sense and antisense, can be used in the process of gene therapy.
  • Gene therapy refers to the process of providing for the expression of nucleic acids of exogenous origin, including antisense nucleic acids or those encoding relaxin agonist or antagonist in a subject for the modulation of apoptosis (e.g. to treat a tissue abnormality) within the subject.
  • nucleic acids encoding a relaxin or a relaxin analog are administered to decrease apoptosis in cells have a relaxin receptor.
  • nucleic acids encoding a relaxin binding agent e.g., a relaxin a soluble relaxin receptor
  • a relaxin receptor binding agent e.g., a relaxin analog
  • Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
  • the nucleic acid comprises a sense nucleic acid (e.g., encoding a relaxin analog, soluble relaxin receptor, and the like) that is part of a vector that expresses the nucleic acid in a suitable host.
  • a nucleic acid has a promoter operably linked to the coding region (e.g., relaxin or relaxin receptor) in a sense orientation, the promoter being inducible or constitutive, and, optionally, tissue- specific.
  • the nucleic acid comprises an antisense nucleic acid (e.g., a relaxin antisense nucleic acid or relaxin receptor antisense nucleic acid) that is part of a vector that expresses the nucleic acid in a suitable host.
  • an antisense nucleic acid e.g., a relaxin antisense nucleic acid or relaxin receptor antisense nucleic acid
  • such a nucleic acid has a promoter operably linked to the coding region (e.g., relaxin or relaxin receptor) in an antisense orientation, the promoter being inducible or constitutive, and, optionally, tissue-specific.
  • a nucleic acid e.g., a sense nucleic acid encoding a relaxin analog, soluble relaxin receptor, and the like, or an antisense nucleic acid encoding a relaxin antisense nucleic acid, a relaxin receptor antisense nucleic acid, and the like
  • the nucleic acid and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the nucleic acid (see, e.g., Roller and Smithies, Proc. Natl. Acad. Sci.
  • delivery of the nucleic acid into a subject can be either direct, in which case the subject is directly exposed to the nucleic acid or nucleic acid-carrying vector, or indirectly, in which case cells are first transformed with the nucleic acid in vitro, and then transplanted into the subject.
  • these two approaches are known, respectively, as in vivo or ex vivo gene therapy.
  • the nucleic acids are directly administered in vivo, where they are expressed to produce the encoded product.
  • This can be accomplished by any of numerous methods known in the art (e.g., by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, for example, by infection using a defective or attenuated retroviral or other viral vector (infra), by direct injection of naked DNA (see, e.g., Asahara et al, Semin. Interv.
  • Nucleic acids can also be inserted into cells by coating naked nucleic acids with lipids or cell- surface receptors or transfection agents, encapsulation in liposomes, derivatized liposomes, microp articles, or microcapsules (see, e.g., De Smedt et al, Pharm. Res. 17:113-26 (2000); Maurer et al, Mol Membr. Biol 16:129-40 (1999); Tarahovsky and Ivanitsky, Biochemistry 63:607-18 (1998); Lasci, Trends Biotechnol. 16: 307-21 (1998); Gao and Huang, Gene Ther.
  • Nucleic acids can also be administered in linkage to a peptide which is known to enter the cell or by administering the nucleic acids in linkage to a ligand subject to receptor-mediated endocytosis, which can be used to target cell types specifically expressing the receptors, and the like.
  • a ligand subject to receptor-mediated endocytosis which can be used to target cell types specifically expressing the receptors, and the like.
  • a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the ligand comprises a fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the antisense nucleic acid can be targeted in vivo for cell specific uptake and expression by targeting a specific receptor (see, e.g., Phillips, Biologicals 23:13-16 (1995); International Patent Publications WO 92/06180; WO 92/22635; WO 92/20316; WO 93/14188, and WO 93/20221; the disclosures of which are inco ⁇ orated by reference herein).
  • a viral vector is used that contains the nucleic acid (e.g., a sense nucleic acid encoding a relaxin analog, soluble relaxin receptor, and the like, or an antisense nucleic acid encoding a relaxin antisense nucleic acid, a relaxin receptor antisense nucleic acid, and the like).
  • a retroviral vector can be used (see, e.g., Palu et al, Rev. Med. Virol. 10:185-202 (2000); Buchschacher and Wong-Staal, Blood 15:2499-504 (2000); Miller et al, Meth.
  • retroviral vectors are typically modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
  • the antisense nucleic acid to be used in gene therapy is cloned into the vector, which facilitates delivery of the antisense nucleic acid into the subject.
  • Lentiviral vectors can also be used. (See, e.g., Buchschacher and Wong-Staal, supra; Naldini et al, Science 272:263-67 (1996), the disclosures of which are inco ⁇ orated by reference herein).
  • Adenoviruses can also be used in gene therapy.
  • Adenoviruses are especially attractive vehicles for delivering genes to prostate, liver, the central nervous system, endothelial cells, and muscle.
  • Adenoviruses have the advantage of being capable of infecting non-dividing cells.
  • Kozarsky and Wilson (Curr. Opin. Genet Dev. 3:499-503 (1993), the disclosure of which is inco ⁇ orated by reference herein) present a review of adenovirus-based gene therapy. Herman et al.
  • Adeno-associated virus can also be used in gene therapy (see, e.g., Rabinowitz and Samulski, Curr. Opin. Biotechnol. 9:475-85 (1988); Carter and Samulski, Int. J. Mol. Med. 6:17-27 (2000); Tal, J. Biomed. Sci. 7:279-91 (2000); Ali et al, Gene Therapy 1:367-84 (1994); U.S. Patent Nos. 4,797,368 and 5,139,941; Walsh et al, Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); Grimm et al, Human Gene Therapy 10:2445-50 (1999)) (the disclosures of which are inco ⁇ orated by reference herein).
  • Another approach to gene therapy involves transferring a nucleic acid to cells in tissue culture by methods such as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection.
  • the method of transfer includes the transfer of a selectable marker to the cells.
  • the cells are then placed under selection to isolate those cells that have taken up and are expressing the nucleic acid.
  • the selected cells are then delivered to a subject.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, and the like. Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Muramatsu et al, Int. J. Mol Med. 1:55-62 (1998); Liang et al, Pharmazie 54:559-66 (1999); Loeffler and Behr, Meth.
  • the technique typically provides for the stable transfer of the nucleic acids to the cell, so that the nucleic acids are expressible by the cell and is heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a subject by various methods known in the art. Typically, cells are injected subcutaneously. In another embodiment, recombinant skin cells can be applied as a skin graft onto the subject.
  • Cells into which a nucleic acid can be introduced for pu ⁇ oses of gene therapy encompass any desired, available cell type, and include but are not limited to cells or populations of cells of the male reproductive tract (e.g., prostate cells, cells of the testes, seminiferous cells, or epididymal cells), female reproductive tract (e.g., uterus, cervix, the inte ⁇ ubic ligament, connective tissues within the pelvic girdle, and the like), liver, kidney, spleen, thymus, brain, heart, intestine, skin, lung, and the like.
  • male reproductive tract e.g., prostate cells, cells of the testes, seminiferous cells, or epididymal cells
  • female reproductive tract e.g., uterus, cervix, the inte ⁇ ubic ligament, connective tissues within the pelvic girdle, and the like
  • Suitable cells further include epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, and stem or progenitor cells.
  • the cells used for gene therapy generally are autologous to the subject, but heterologous cells that can be typed for compatibility with the subject can be used.
  • nucleic acids e.g., a sense nucleic acid encoding a relaxin analog, soluble relaxin receptor, and the like, or an antisense nucleic acid encoding a relaxin antisense nucleic acid, a relaxin receptor antisense nucleic acid, and the like
  • nucleic acids e.g., a sense nucleic acid encoding a relaxin analog, soluble relaxin receptor, and the like, or an antisense nucleic acid encoding a relaxin antisense nucleic acid, a relaxin receptor antisense nucleic acid, and the like
  • the nucleic acids are at least six nucleotides and are typically oligonucleotides (ranging from 6 to about 50 nucleotides or more).
  • the oligonucleotide is at least 10 nucleotides, at least 15 nucleotides, at least 100 nucleotides, or can be at least 200 nucleotides.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or analogs thereof, and can be single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide can include other appending groups such as peptides, or agents facilitating transport across the cell membrane (see, e.g., Nielsen, Pharmacol. Toxicol. 86:3-7 (2000); Soomets et al, Front. Biosci. LD782-86 (1999); Galderisi et al, J. Cell Physiol 181:251-57 (1999); Letsinger et al. , Proc. Natl. Acad. Sci. USA 86:6553-56 (1989); Lemaiire et al, Proc. Natl. Acad. Sci.
  • antisense oligonucleotides are provided as single- stranded DNA.
  • the oligonucleotides can be modified at any position on its structure with substituents generally known in the art.
  • the oligonucleotides can comprise at least one modified base moiety, such as, for example, 5-fluorouracil, 5-bromouracil, 5- chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxy- hydroxylmethyl) uracil, 5-carboxymethylaminomethyl -2-thiouridine, 5- carboxymethylamino-methyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1 -methylguamne, 1-methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methyl
  • the oligonucleotides comprise at least one modified sugar moiety, such as, for example, arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the antisense oligonucleotides comprise at least one modified phosphate backbone, such as, for example, a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
  • the antisense oligonucleotide is an ⁇ - anomeric oligonucleotide.
  • An ⁇ -anomeric oligonucleotide forms specific double- stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (see Gautier et al, Nucl Acids Res. 15:6625-41 (1987)).
  • the oligonucleotide can be conjugated to another molecule (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent, and the like).
  • the antisense oligonucleotide comprises catalytic RNA, or a ribozyme (see, e.g., Welch et al, Curr. Opin. Biotechnol. 9:486-96 (1998); Norris et al, Adv. Exp Med. Biol 465:293-301 (2000); International Patent Publication WO 90/11364; Sarver et al, Science 247:1222-25 (1990)).
  • the oligonucleotide is a 2'-0-methylribonucleotide (Inoue et al, Nucl. Acids Res. 15:6131-48 (1987)), or a chimeric RNA-DNA analogue (Inoue et al, FEBS Lett. 215:327-30 (1987)).
  • double-stranded RNA directs the sequence-specific degradation of mRNA by RNA interference.
  • RNA interference See generally Hunter, Curr. Biol. 10.-R137-40 (2000); Bosher and Labouesse, Nat. Cell. Biol. 2:e31-36 (2000); the disclosures of which are inco ⁇ orated by reference herein.
  • double-stranded nucleic acids are introduced into a cell to selectively inhibit gene expression by causing degradation of the mRNA.
  • Zamore et al Cell 101:25-33 (2000), the disclosure of which is inco ⁇ orated by reference herein.
  • Nucleic acids according to the present invention can be synthesized by standard methods known in the art. Enzymatic methods for the synthesis of nucleic acids frequently employ Klenow, T7, T4, Taq or Escherichia coll DNA polymerases, as described in Sambrook et al (supra). Enzymatic methods of RNA nucleic acids frequently employ SP6, T3 or T7 RNA polymerases, as described in Sambrook et al. (supra). Reverse transcriptase can also be used to synthesize DNA from RNA (Sambrook et al, supra).
  • Nucleic acids are typically prepared enzymatically using a template nucleic acid which can either be synthesized chemically, or be obtained as mRNA, genomic DNA, cloned genomic DNA, cloned cDNA or other nucleic acid. Some enzymatic methods of DNA nucleic acid synthesis can require an additional primer which can be synthesized chemically. Finally linear nucleic acids can be prepared by polymerase chain reaction (PCR) techniques as described, for example, by Saiki et al. (Science 239:487 (1988)). Chemical methods can also be used to synthesize nucleic acids (e.g., antisense oligonucleotides), such as by use of a commercially available automated DNA synthesizer).
  • PCR polymerase chain reaction
  • phosphorothioate nucleic acids can be synthesized by the method of Stein et al. (Nucl. Acids Res. 16:3209-21 (1988)), methylphosphonate nucleic acids can be prepared by use of controlled pore glass polymer supports (see, e.g., Sarin et al, Proc. Natl. Acad. Sci. USA 85:7448-51
  • Relaxin nucleic acids, polypeptides, analogs and fragments, and relaxin receptor nucleic acids, polypeptides, analogs and fragments and analogs also have uses in screening assays to detect candidate compounds that specifically bind to relaxin polypeptides, or to relaxin receptor, and modulate apoptosis.
  • candidate compounds are typically small molecule effectors (agonists) or antagonists, and can be identified by in vitro and/or in vivo assays.
  • Such assays can be used to identify small molecule effectors or antagonists that are therapeutically effective as relaxin agonists or antagonists or as lead compounds for drug development.
  • the invention thus provides assays to detect compounds that specifically affect the activity or expression of relaxin nucleic acids, relaxin polypeptides, relaxin receptor nucleic acids, relaxin receptor, and the like.
  • recombinant cells expressing relaxin or relaxin receptor nucleic acids can be used to screen candidate compounds for those that affect relaxin or relaxin receptor nucleic acid expression.
  • Agonistic or antagonistic effects on relaxin or relaxin receptor expression can include stimulation or inhibition (e.g., up or down regulation) of transcription of mRNA, a increase or decrease in mRNA stability, translation of the mRNA, synthesis of relaxin or relaxin receptor polypeptides, relaxin or relaxin receptor polypeptide function (e.g., binding to relaxin receptor), and/or effects on relaxin or relaxin receptor polypeptide stability or localization.
  • Such effects on expression can be identified as physiological changes, such as, for example, changes in relaxin-responsive tissue growth rate, division, viability, collagen deposition, apoptosis, and the like.
  • candidate compounds are administered to recombinant cells expressing a relaxin or relaxin receptor polypeptide to identify those compounds that produce a physiological change (e.g., stimulate or inhibit relaxin or relaxin receptor polypeptide function).
  • a physiological change e.g., stimulate or inhibit relaxin or relaxin receptor polypeptide function.
  • candidate compounds can also be identified by in vitro screening methods.
  • recombinant cells expressing a relaxin or a relaxin receptor nucleic acid can be used to recombinantly produce relaxin or relaxin receptor polypeptide for in vitro assays to identify candidate compounds that bind to relaxin or relaxin receptor polypeptide.
  • Candidate compounds are contacted with the polypeptide (or a fragment or analog thereof) under conditions conducive to binding, and then candidate compounds that specifically bind to the polypeptide are identified.
  • Methods that can be used to carry out the foregoing are commonly known in the art, and include diversity libraries, such as random or combinatorial peptide or non-peptide libraries that can be screened for candidate compounds that specifically bind to relaxin or relaxin receptor polypeptide.
  • diversity libraries such as random or combinatorial peptide or non-peptide libraries that can be screened for candidate compounds that specifically bind to relaxin or relaxin receptor polypeptide.
  • Many libraries are known in the art that can be used, for example, include chemically synthesized libraries, recombinant phage display libraries, and in vitro translation- based libraries.
  • phage display libraries are described in Scott and Smith (Science 249:386-90 (1990)), Devlin et al. (Science 249:404-06 (1990)), Christian et al. (J. Mol Biol 227:711-18 (1992)), Lenstra (J. Immunol. Meth. 152:149-57 (1992)), Kay et al. (Gene 128:59-65 (1993)), and International Patent Publication WO 94/18318, the disclosures of which are inco ⁇ orated by reference herein.
  • In vitro translation-based libraries include, but are not limited to, those described in International Patent Publication WO 91/05058, and Mattheakis et al. (Proc. Natl Acad. Sci. USA 91:9022-26 (1994)).
  • a benzodiazepine library see, e.g., Bunin et al, Proc. Natl. Acad. Sci USA 91 :4708-12 (1994)
  • Peptide libraries see, e.g., Simon et al, Proc. Natl. Acad. Sci. USA 89:9367-71(1992) can also be used.
  • Screening the libraries can be accomplished by any of a variety of commonly known methods. See, for example, the following references, which disclose screening of peptide libraries: Parmley and Smith (Adv. Exp. Med. Biol. 251:215-18 (1989)); Scott and Smith (1990, supra); Fowlkes et al (BioTechniques 13:422-28 (1992)); Oldenburg et al. (Proc. Natl. Acad. Sci. USA 89:5393-97 (1992)); Yu et al. (Cell 76:933-45 (1994)); Staudt et al (Science 241 :577-80 (1988)); Bock et al.
  • screening can be carried out by contacting the library members with relaxin, a relaxin analog , or a relaxin receptor immobilized on a solid phase and harvesting those library members that bind to the relaxin, relaxin analog or relaxin analog.
  • relaxin a relaxin analog
  • relaxin receptor immobilized on a solid phase
  • Relaxin nucleic acids both sense and antisense
  • fragments and analogs thereof, and anti-relaxin antibodies also have utility to identify subjects with a relaxin-associated abnormality.
  • Such molecules can be used in assays, such as hybridization or immunoassays, to detect, prognose, diagnose, or monitor various abnormalities, to determine whether relaxin expression, or the response to relaxin or a relaxin analog is affected. Similarly, such molecules have utility to monitor the treatment of the cell or tissue abnormalities.
  • methods such as an immunoassay, can be carried out by steps comprising contacting a sample derived from a subject with an anti-relaxin antibody under conditions conducive to immunospecific binding, and detecting or measuring the amount of any immunospecific binding of protein by the antibody.
  • Binding of antibody to relaxin or relaxin receptor polypeptide, in tissue sections or from seminal fluid, can be used to detect abenant (e.g., low, absent or elevated) levels of relaxin and/or relaxin receptor polypeptide.
  • antibody to relaxin or relaxin receptor polypeptide can be used to assay a subject's tissue or seminal fluid for the presence of relaxin or relaxin receptor polypeptide, where an aberrant level of relaxin is an indication of a relaxin-associated abnormality.
  • aberrant levels is meant increased or decreased levels relative to that present, or to a standard level representing that present, in an analogous sample from a portion of the body or from a subject not having the abnormality.
  • the immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as Western blot, radioimmunoassay, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassay, immunoprecipitation assay, precipitin reaction, gel diffusion precipitin reaction, immunodiffusion assay, agglutination assay, complement-fixation assay, immunoradiometric assay, fluorescent immunoassay, protein A immunoassay, and the like.
  • Relaxin and relaxin receptor nucleic acids both sense and antisense
  • hybridization assays can be used to detect, prognose, diagnose, or monitor diseases or conditions associated with aberrant relaxin or relaxin receptor expression and/or activity, as described supra.
  • a hybridization assay can be carried out by a method comprising contacting a sample containing polynucleotides with a nucleic acid probe capable of hybridizing to relaxin or relaxin receptor DNA or RNA, under conditions such that hybridization can occur, and detecting or measuring any resulting hybridization.
  • abnormalities associated with over- or under- expression of relaxin can be diagnosed, or their suspected presence can be screened for, or a predisposition to develop such abnormalities can be identified by detecting decreased or increased levels of relaxin polypeptide, relaxin RNA, or relaxin functional activity.
  • over-expression of relaxin or increased relaxin functional activity can be diagnosed by detecting mutations in relaxin RNA or DNA or relaxin polypeptide (e.g., translocations in relaxin nucleic acids, truncations of the relaxin gene or relaxin polypeptide, changes in the nucleotide or amino acid sequence relative to wild-type relaxin, respectively) that cause increased expression or activity of relaxin polypeptide.
  • levels of relaxin polypeptide in a biopsy or from seminal fluid can be detected by immunoassay of tissues; levels of relaxin RNA can be detected by hybridization assays (e.g., Northern blot or dot blot). Translocations and point mutations in relaxin or relaxin receptor nucleic acids can be detected by Southern blot, RFLP analysis, PCR using primers that detect point mutations, deletions or insertions, sequencing of the relaxin genomic DNA or cDNA obtained from the sample, and the like.
  • levels of relaxin or relaxin receptor mRNA or polypeptide in a sample of tissue isolated from a subject are detected or measured, in which increased levels indicate that the subject has, or has a predisposition to a relaxin-associated tissue abnormality.
  • abnormal relaxin receptor activity in a tissue is detected or measured using any of the functional assays described above.
  • Abnormal relaxin receptor activity can include, for example, an increased or decreased number of a relaxin receptors on relaxin-responsive cells, the presence of relaxin receptors on cells that are not normally responsive to relaxin, increased or decreased relaxin receptor response time, an increased or decreased binding affinity for relaxin or relaxin receptor, or a change in the dissociation constant of relaxin from a relaxin receptor , and the like.
  • Kits for diagnostic and/or prognostic use comprise, in one or more containers, a relaxin agonist or antagonist and, optionally, a labeled binding partner to an antibody.
  • an antibody can be labeled with a detectable marker (e.g., a chemiluminescent, enzymatic, fluorescent, a radioactive moiety, and the like).
  • a kit is also provided that comprises, in one or more containers, a nucleic acid probe capable of hybridizing to relaxin or relaxin receptor mRNA or DNA.
  • the kit can comprise in one or more containers a pair of primers (e.g., each in the size range of 6-30 nucleotides or more) that are capable of priming amplification (e.g., by polymerase chain reaction (see, e.g., Innis et al, PCR Protocols, Academic Press, Inc., San Diego, CA (1989)), ligase chain reaction (see, e.g., EP 0 320 308), use of Q ⁇ replicase, cyclic 5' probe reaction, or other methods known in the art) under appropriate reaction conditions such that at least a portion of a relaxin nucleic acid is amplified.
  • a kit can optionally further comprise in a container a predetermined amount of a purified relaxin or relaxin receptor nucleic acid, for example, for use as a standard or control.
  • EXAMPLE 1 The effect of inactivation of one or both alleles of the mouse RLX genes was examined. The following methods and materials were followed.
  • Wild-type, heterozygous and homozygous relaxin knockout mice were obtained from the Howard Florey Institute of Experimental Physiology and Medicine (Parkville, Victoria, 3052, Australia) and used to establish a breeding program. Subsequent generations of RLX +/+ (wild-type), RLX +/- (heterozygous) and rlx -/- (null mutant) mice were generated from RLX +/- parents. All animals were housed in a controlled environment and maintained on a 14 hours light, 10 hours dark schedule with access to Labdiet rodent lab chow (Deans Animal Feed, San Bruno, CA) and water. These experiments were approved by the Institute's Animal Experimental Ethics Committee, which adheres to the NIH Code of Practice for the care and use of laboratory animals.
  • Mouse DNA was isolated by lysing tail tissue (5-7 millimeters) in 400 ⁇ l of PCR lysis buffer, containing 50 mM Tris-HCl, pH 8.0, 0.5% SDS, 0.1 M EDTA and 1 mg/ml proteinase K (Gibco BRL, Gaithersburg, MD) at 50-55°C overnight. Digested samples were then mixed with 3M sodium acetate (40 ⁇ l), buffer saturated phenol (200 ⁇ l) and chloroform (200 ⁇ l) in serum vaccutainer tubes, before samples were centrifuged at 3000 ⁇ in for 10 minutes.
  • PCR lysis buffer containing 50 mM Tris-HCl, pH 8.0, 0.5% SDS, 0.1 M EDTA and 1 mg/ml proteinase K (Gibco BRL, Gaithersburg, MD) at 50-55°C overnight. Digested samples were then mixed with 3M sodium acetate (40 ⁇ l), buffer saturated phenol (200 ⁇ l) and
  • the DNA (contained in the upper aqueous phase) was decanted into separate microcentrifuge tubes containing isopropyl alcohol (240 ⁇ l) to precipitate the DNA before samples were vortexed, spun and the supernatant discarded. The remaining DNA pellet was dissolved in 40-50 ⁇ l of sterile water.
  • each DNA template (1 ⁇ l) was used in a 30 ⁇ l reaction mixture containing PCR buffer (10 mM Tris-HCl, pH 8.3, 50 mM KCl, 1.5 mM MgCl 2 ), 2.5 mM dNTPs, 2.5 U Taq Polymerase (PGC Scientific, Gaithersburg, MD) and 150 ng of each of the RLX +/+ and rlx -/- primers, designed by Zhao and colleagues (Zhao Endocrinology 140:445-53 (1999)).
  • the amplification protocol consisted of an initial denaturation step at 94°C (3 minutes) followed by 35 sequential cycles of 94°C (60 seconds), 55°C (60 seconds) and 72°C (90 seconds) and concluded by an additional 10 minute extension at 72°C. 15 ⁇ l of each sample were then analyzed by electrophoresis on a 2% (w/v) agarose gel, and stained with ethidium bromide. A 235 bp product was generated by primers designed from the wild-type allele, while a 170 bp product was generated from the mutant allele primers (Zhao (1999), supra).
  • RLX +/+ and rlx -/- male mice were then sacrificed under anesthesia with carbon dioxide for tissue collection.
  • the collected tissues were processed (sequentially dehydrated) from 70% alcohol to paraffin before being embedded and cut (4 ⁇ m sections), using an AO Spencer 820 microtome and placed on poly-L-lysine coated glass slides.
  • Consecutive sections from each tissue were stained with H & E (hematoxylin and eosin) and for collagen, with Masson trichrome (staining kit, Richard-Allan Scientific, Kalamazoo, MI) as described by the manufacturer.
  • the stained slides were viewed using a Zeiss Axioplan-2 microscope, the images captured by digital camera (Hamamatsu) and stored for retrieval and analysis.
  • the images were digitally enhanced for maximum contrast and brightness using Adobe Photoshop (Adobe Systems Inc, Mountain View, CA).
  • the tissues from the reproductive tract of one and three month old male mice were mounted on precoated slides and deparaffmized by heating at 58° C (about 30 minutes), then washed three times in xylene, twice in absolute ethanol and twice in 95%o ethanol, before being briefly soaked in water.
  • the samples were then stained using an Immunocruz staining system utilizing a horseradish peroxidase (HRP)- streptavidin complex (Santa Cruz Biotechnology Inc, Santa Cruz, CA) in a humidified atmosphere.
  • Tissue sections were initially treated with a peroxidase blocker (to quench endogenous peroxidase activity) (5 minutes) before being preb locked in goat serum (20 minutes).
  • Samples were washed in PBS (2 minutes), subjected to the appropriate secondary antibody (goat anti-mouse IgG or goat anti-rabbit IgG) (30 minutes), washed as above (2 minutes), treated with a HRP- streptavidin complex (30-45 minutes) and incubated with a diaminobenzidine chromagen substrate (2-10 minutes) that was prepared in accordance with the manufacturer's instructions.
  • the slides were then washed in distilled water (2 minutes) before being dehydrated from 95% alcohol to xylene and mounted, then photographed as described above.
  • mice were 1 month of age, mean body weights of both rlx -/- males (17.05 ⁇ 0.65g) and females (14.77 ⁇ 0.42g) were significantly less (p ⁇ 0.05) than their respective RLX wildtype counte ⁇ arts (RLX +/+ M: 18.92 ⁇ 0.61g; RLX +/+ F: 16.34 ⁇ 0.51g).
  • mice Male and female null mutant mice continued to be significantly smaller ( ⁇ .05) than RLX wildtype animals at 2 months of age (RLX +/+ M: 25.42 ⁇ 0.41g; rlx -/- M: 24.23 ⁇ 0.42g; RLX +/+ F: 20.96 ⁇ 0.32g; rlx -/- F: 19.94 ⁇ 0.22g); however, the differences in size between the two groups were less than that observed at 1 month of age.
  • mice had reached adulthood (3 months of age) a significant (p ⁇ 0.05) difference in the overall weight of the male reproductive tract as well as in the size of individual organs was observed (Table 1).
  • the weight of the reproductive tract in normal mice increased 248.6% from 1 month to adulthood and represented 3.37% of the total body weight.
  • the weight of the reproductive tract from male RLX null mutant mice only increased by 187.9% and represented 2.33% of the total body. This latter finding implied that the reproductive tract of male rlx -/- mice represented a 31% decrease in % body weight.
  • the size of the testis, epididymis, prostate and seminal vesicle from rlx -/- mice were all smaller (p ⁇ 0.05) than their respective counte ⁇ arts, derived from RLX +/+ mice.
  • Table 1 The weight and size of the male reproductive tract from 1 week of age to adulthood (6.5 months).
  • each 'testis and prostate was measured by its area, multiplying the tissue length by width.
  • the size (area) of the epididymis and seminal vesicle could not be measured accurately, so a close approximation of each tissue was calculated as follows: epididymis (by measuring the length of the tissue by the width of the epididymis head 2 ); seminal vesicle (by measuring the length of each organ by the average width of the tissue 4 ), denotes p ⁇ 0.05.
  • H&E & Masson Trichrome Staining Male reproductive tissue sections from RLX +/+ and rlx -/- mice, were first observed for differences in sperm maturation, tubule size/compactness (testis, epididymis) and collagen. Testis: At 1 week of age, the seminiferous tubules (the compartments containing germ cells/spermatocytes, Sertoli cells and where sperm maturation occurs) of relaxin wildtype animals were smaller, more cylindrical in shape (Table 2) and mainly supported by a thin layer of collagen sunounding the tunica albuginea (the membrane that covers the oval body) of each organ.
  • tubules derived from rlx -/- mouse testes were much larger and elongated, but were completely surrounded and supported by collagen within the testis. This immediately suggested a difference in the internal organization of the testis in the absence of relaxin, even just after birth. No immature sperm though were detected in either group of tissue sections at one week of age. By one month of age, testis tubules derived from RLX +/+ mice were larger in size (compared to 1 week tubule size), were slightly less compact and contained mainly immature sperm.
  • tubules derived from 1 month old RLX homozygous mice were no different to tubule sizes measured from 1 week old knockout mice and contained less immature sperm compared to samples derived from RLX +/+ mice.
  • relaxin null mutant mice were undergoing a process of delayed sperm maturation which was further confirmed when comparing three month tissue sections; sections derived from RLX +/+ mouse tissues contained larger tubules (compared to 1 month tubule size) with mainly mature sperm.
  • sections derived from rlx -/- mice contained slightly smaller tubules (which were indifferent to tubule sizes derived from 1 week/1 month rlx -/- mice) with less mature sperm (compared to 3 month RLX +/+ tissue sections) and still some immature sperm.
  • the level of sperm maturity in testes derived from RLX +/+ mice was shown to be significantly (p ⁇ 0.05) greater than the level of sperm maturation observed in the testes of rlx -/- mice (see Table 2 for grading scale used). It was also noted that the level of sperm maturation in adult RLX knockout mouse testes resembled that observed in immature (1 month) RLX wildtype mouse tissues.
  • testis tubules derived from rlx -/- mice appeared to contain an increased number of dead cells, compared to tubular cells derived from RLX wildtype animals.
  • the increased number of dead cells seen in immature tissues were variable between animals but were consistently detected as the mice matured with age.
  • Table 2 The effects of relaxin gene knockout on tissue compartment and spenn maturation, during murine growth and development
  • the ducts of tissues derived from RLX knockout mice were more compact and completely supported by connective tissue, while the ducts obtained from RLX wildtype mice were more spread out (loose) and still supported by little collagen. Furthermore, the ducts of the adult prostate obtained from rlx -/- mice appeared to have smaller epithelium, containing less cells, while the ducts of RLX +/+ prostate samples contained larger epithelial layers/glandular tissue.
  • Bax Antibody Staining Overexpression of Bax accelerates apoptotic death induced by cytokine deprivation and also counters the death repressor activity of Bcl-2 (Krajewski et al, Am. J. Pathol. 145:1323-36 (1994)). Using a Bax monoclonal antibody, the in vivo distribution of the Bax protein was evaluated in the male reproductive tract. Testis: Seminiferous tubules derived from immature (1 month) wildtype animals showed weak immunostaining for Bax (dark brown cell staining), which was primarily observed in the germinal cells near the basement membrane. These findings are consistent with those of previous reports (Ben-Hur et al, Calcif Tiss.
  • Epididymis A more intense Bax staining was present in the intracellular membrane of epithelial cells of epididymis tubules derived from 1 month RLX +/+ mouse tissues.
  • the same tubules derived from 1 month rlx -/- mouse tissues contained a relatively stronger level of Bax staining, which was further maintained, if not increased in 3 month tissue sections.
  • the epididymis of adult wildtype mice contained a weaker expression of Bax staining, compared to tissues derived from 1 month normal mice, as the tissue matured with age.
  • Prostate The epithelial cells of the prostate of normal 1 month and 3 month animals showed weak positive staining for Bax.
  • Caspase-9 a central death protease, belongs to a unique family of cysteine proteases that differ in sequence, structure and substrate specificity to other described protease families.
  • the caspase family members (which are usually involved in a cascade of proteolytic cleavage events) function as key components of the apoptotic machinery by acting to destroy specific target proteins which are critical to cellular activity.
  • Testis tubules derived from rlx -/- mice contained slightly fewer apoptotic cells at 3 months (61.6 ⁇ 9.8 cells / testis (n 5)), compared to the density of caspase-9 stained cells in 1 months tissues.
  • Epididymis No staining for caspase-9 was detected from 1 month RLX +/+ epididymis tubules, while trace amounts of positive cells were observed in rlx -/- tissue sections.
  • PCNA staining The proliferating cell nuclear antigen (PCNA) antibody was used to detect cell proliferation in the male reproductive tract based on its ability to associate with nuclear regions where DNA synthesis is occurring.
  • Testis PCNA was detected in immature and mature seminiferous tubules, although no significant differences in PCNA staining were detected in testis tubules derived from RLX +/+ and rlx -/- mice at 1 month and 3 months of age. In immature tissues, PCNA staining is usually highly expressed in mitotically active spermatogonia and occasionally in some Sertoli cells, which conesponds to proliferative activity.
  • RLX wildtype (RLX +/+) and RLX gene knockout (rlx -/-) male and female mice were generated from RLX heterozygous (RLX +/-) parents, as described above. Mice were weighed and sacrificed at 1 week, 1 month and 3 months of age for tissue collection. The following tissues, including the brain, heart, liver, kidneys, thymus, spleen and male reproductive tract were collected, weighed and placed into 10% formalin for detailed histological analysis. A summary of the weights of these tissues are shown in Tables 4-8.
  • mice At 1 week of age, male rlx -/- mice contained significantly (p ⁇ 0.05) smaller livers, kidneys and spleens compared to RLX +/+ animals, but these weight differences were no longer apparent at 1 month of age. Instead, the thymus of 1 month old rlx -/- male mice was smaller (p ⁇ 0.05) than their RLX +/+ counte ⁇ arts. This weight difference was no longer apparent at 3 months of age.
  • RLX +/+ mice obtained from the corresponding set of parents and were sacrificed at 1 week, 1 month and 3 months of age.
  • the brain, heart, liver, kidneys, thymus, spleen, male reproductive tract, intestine and lung were collected and weighed. The weights of these organs are shown in Tables 4-8.
  • organ (tissue) weight to body weight ratio indicates a change in underlying organ or tissue development or cellular architecture.
  • a small liver that is infiltrated (altered) by fibrosis will weigh the same or more than a larger normal liver or one filled with fat.
  • tissues from brain, heart, liver, kidneys, thymus, spleen, intestine and lung of male and female RLX-/- mice show evidence of increased apoptosis and extracellular matrix (collagen) accumulation.
  • Table 8 Tissue Comparison of 3 month old mice (from RLX+/+ and RLX-/- parents, respectively), represented as Mean ⁇ SE (n):
  • rlx-null mice rlx-null mice
  • rlx-/- mice were the progeny of rlx-/- male and female parents.
  • Sequential skin samples from the ear and dorsum of the back of at least 5 male and 5 female mice were stained with H&E and Masson's trichrome stain and examined at each time point. Similar samples at the same time points were obtained from Rlx+/+ mice of the same strain.
  • the dermis was found to have thickened progressively with time and had increased fibrosis throughout the dermis.
  • relaxin may be produced and circulate at biologically relevant concentrations below curcent levels of detection in males and females. Relaxin participates in the ordered maintenance of matrix turnover in concert with other matrix-regulating molecules. Relaxin can influence tissue remodeling, promotion of blood vessel formation and stimulation of vasodilatation. These results also indicate that the relaxin synthetic pathway, and/or relaxin receptor, is associated with fibrotic conditions, such as scleroderma.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Gynecology & Obstetrics (AREA)
  • Diabetes (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Neurosurgery (AREA)
  • Toxicology (AREA)
  • Neurology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Obesity (AREA)

Abstract

Cette invention concerne la découverte selon laquelle la relaxine est associée au développement ou à la maturation des tissus de l'organisme. La rupture du gène codant la relaxine entraîne de nombreux dysfonctionnements dans le développement de divers tissus. L'invention concerne des méthodes de modulation de l'apoptose par l'administration d'un agoniste ou d'un antagoniste de la relaxine à un sujet.
PCT/US2001/042484 2000-10-04 2001-10-04 Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine WO2002028418A1 (fr)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CA2425712A CA2425712C (fr) 2000-10-04 2001-10-04 Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine
AU1185502A AU1185502A (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
JP2002532242A JP2004510743A (ja) 2000-10-04 2001-10-04 レラキシンのアゴニストまたはアンタゴニストの投与によるアポトーシスを調節する方法
AU2002211855A AU2002211855B2 (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
IL15525001A IL155250A0 (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
US10/398,553 US20050032683A1 (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
NZ525389A NZ525389A (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
EP01979945A EP1326627A4 (fr) 2000-10-04 2001-10-04 Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine
US11/781,872 US7833526B2 (en) 2000-10-04 2007-07-23 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
US12/945,685 US8119136B2 (en) 2000-10-04 2010-11-12 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
US13/399,620 US9534034B2 (en) 2000-10-04 2012-02-17 Methods of modulating apoptosis by administration of relaxin agonists or antagonists

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US23823200P 2000-10-04 2000-10-04
US60/238,232 2000-10-04
US24203700P 2000-10-20 2000-10-20
US24199100P 2000-10-20 2000-10-20
US60/242,037 2000-10-20
US60/241,991 2000-10-20

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US10398553 A-371-Of-International 2001-10-04
US10/398,553 A-371-Of-International US20050032683A1 (en) 2000-10-04 2001-10-04 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
US11/781,872 Division US7833526B2 (en) 2000-10-04 2007-07-23 Methods of modulating apoptosis by administration of relaxin agonists or antagonists
US11/781,872 Continuation-In-Part US7833526B2 (en) 2000-10-04 2007-07-23 Methods of modulating apoptosis by administration of relaxin agonists or antagonists

Publications (2)

Publication Number Publication Date
WO2002028418A1 true WO2002028418A1 (fr) 2002-04-11
WO2002028418B1 WO2002028418B1 (fr) 2002-07-18

Family

ID=27399064

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/042484 WO2002028418A1 (fr) 2000-10-04 2001-10-04 Methodes de modulation de l'apoptose par l'administration d'agonistes ou d'antagonistes de relaxine

Country Status (7)

Country Link
EP (1) EP1326627A4 (fr)
JP (3) JP2004510743A (fr)
AU (2) AU1185502A (fr)
CA (2) CA2967607A1 (fr)
IL (1) IL155250A0 (fr)
NZ (1) NZ525389A (fr)
WO (1) WO2002028418A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1399537A2 (fr) * 2001-04-03 2004-03-24 Curagen Corporation Polypeptides therapeutiques, acides nucleiques les codant, et procedes d'utilisation
EP2377891A1 (fr) * 2008-12-26 2011-10-19 The University of Tokyo Diagnostic et traitement du cancer à l'aide d'un anticorps anti-lgr7
US20140213520A1 (en) * 2013-01-25 2014-07-31 Cardiorentis Ltd. Methods of treating cardiovascular indications
US9358271B2 (en) 2005-04-07 2016-06-07 Cardiorentis Ag Use of natriuretic peptide for treating heart failure

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8445635B2 (en) 2006-04-11 2013-05-21 Armour Therapeutics Inc. Modified H2 relaxin for tumor suppression
CN113147632B (zh) * 2021-05-17 2023-03-07 中国第一汽车股份有限公司 一种车辆控制方法、装置、车辆及存储介质

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811395A (en) * 1995-06-07 1998-09-22 Medical University Of South Carolina Relaxin analogs and derivatives methods and uses thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH661662A5 (en) * 1984-04-16 1987-08-14 Georg L Prof Dr Floersheim Product for the treatment of the skin and of the connective tissue
WO1994029452A2 (fr) * 1993-06-09 1994-12-22 Ribozyme Pharmaceuticals, Inc. Molecule d'arn enzymatique pour le traitement de la fibrose et de la maladie du tissu fibreux
JPH09502726A (ja) * 1993-09-14 1997-03-18 マリンクロット・メディカル・インコーポレイテッド 血管障害を防ぐポリペプチドを用いる治療的処置
WO1999040929A1 (fr) * 1998-02-12 1999-08-19 The General Hospital Corporation Procedes de potentialisation des cancerotherapies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5811395A (en) * 1995-06-07 1998-09-22 Medical University Of South Carolina Relaxin analogs and derivatives methods and uses thereof

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
HAGEMANN ET AL.: "Relaxin enhances in vitro invasiveness of breast cancer cell lines and expression of matrix metalloproteases", ONCOLOGY, vol. 23, no. 7, 25 October 2000 (2000-10-25), pages 156, ABSTRACT 0594, XP002908330 *
HANSELL ET AL.: "The role of relaxin in promoting prostate cancer cellular motility and wound healing", BREAST AND PROSTATE CANCER II, no. SUPPL. 0(18D), March 1994 (1994-03-01), pages 230, ABSTRACT Y110, XP002908332 *
PARK ET AL.: "Effects of relaxin on the fertilization capacity of human spermatozoa", AMERICAN JOURNAL OF OBSTETRICS AND GYNECOLOGY, vol. 158, no. 4, April 1988 (1988-04-01), pages 974 - 979, XP002908335 *
See also references of EP1326627A4 *
SOKOL ET AL.: "Immunohistochemical localization of relaxin in human prostate", THE JOURNAL OF HISTOCHEMISTRY AND CYTOCHEMISTRY, vol. 37, no. 8, 1989, pages 1253 - 1255, XP002908334 *
TASHIMA ET AL.: "Human relaxins in normal, benign and neoplastic breast tissue", J. OF ENDOCRINOLOGY, vol. 12, 1994, pages 351 - 364, XP002908331 *
UNEMORI ET AL.: "Human relaxin decreases collagen accumulation in vivo in two rodent models of fibrosis", THE JOURNAL OF INVESTIGATIVE DERMATOLOGY, vol. 101, no. 3, September 1993 (1993-09-01), pages 280 - 285, XP002908333 *
WANG-LEE ET AL.: "Regulation of urokinase- and tissue-type plasminogen activator by relaxin in the uterus and cervix of the prepubertal gilt", JOURNAL OF REPRODUCTION AND FERTILITY, vol. 114, 1998, pages 119 - 125, XP002908336 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1399537A2 (fr) * 2001-04-03 2004-03-24 Curagen Corporation Polypeptides therapeutiques, acides nucleiques les codant, et procedes d'utilisation
EP1399537A4 (fr) * 2001-04-03 2006-09-06 Curagen Corp Polypeptides therapeutiques, acides nucleiques les codant, et procedes d'utilisation
US9358271B2 (en) 2005-04-07 2016-06-07 Cardiorentis Ag Use of natriuretic peptide for treating heart failure
EP2377891A1 (fr) * 2008-12-26 2011-10-19 The University of Tokyo Diagnostic et traitement du cancer à l'aide d'un anticorps anti-lgr7
EP2377891A4 (fr) * 2008-12-26 2012-08-29 Univ Tokyo Diagnostic et traitement du cancer à l'aide d'un anticorps anti-lgr7
US20140213520A1 (en) * 2013-01-25 2014-07-31 Cardiorentis Ltd. Methods of treating cardiovascular indications

Also Published As

Publication number Publication date
IL155250A0 (en) 2003-11-23
EP1326627A4 (fr) 2004-06-16
AU1185502A (en) 2002-04-15
WO2002028418B1 (fr) 2002-07-18
JP2004510743A (ja) 2004-04-08
EP1326627A1 (fr) 2003-07-16
JP2008297313A (ja) 2008-12-11
JP2012184273A (ja) 2012-09-27
CA2425712A1 (fr) 2002-04-11
AU2002211855B2 (en) 2007-06-07
NZ525389A (en) 2009-04-30
CA2425712C (fr) 2017-06-06
CA2967607A1 (fr) 2002-04-11

Similar Documents

Publication Publication Date Title
KR101870246B1 (ko) 신질환의 예방 또는 치료제
Topilko et al. Multiple pituitary and ovarian defects in Krox-24 (NGFI-A, Egr-1)-targeted mice
JP6143015B2 (ja) 肝疾患の予防または治療剤
US9534034B2 (en) Methods of modulating apoptosis by administration of relaxin agonists or antagonists
KR20050096212A (ko) 당뇨병 치료제
BG63332B1 (bg) Метод за промяна на една или повече характеристики на клетки на репродуктивния тракт на бозайник
JP2012184273A (ja) レラキシンのアゴニストまたはアンタゴニストの投与によるアポトーシスを調節する方法
Stahl et al. Thyroid hormone is essential for pituitary somatotropes and lactotropes
US20060148001A1 (en) Compositions and methods for treating female fertility
EP1411971B1 (fr) Utilisation de longue pentraxine ptx3 aux fins de traitement de l'infertilite feminine
EP1074265A1 (fr) Utilisation de l'AMH et/ou d'un agoniste de l'AMH et/ou d'un antagoniste de l'AMH pour contoler la fertilité féminine à long terme
AU2002211855A1 (en) Methods of modulating apoptosis by administration of relaxin agonists or antagonists
Johnston et al. Expression of v-Ha-ras driven by the calcitonin/calcitonin gene-related peptide promoter: a novel transgenic murine model for medullary thyroid carcinoma
Kedzia et al. Medullary thyroid carcinoma arises in the absence of prolactin signaling
JP4451158B2 (ja) 転写制御シスエレメント及びそれに特異的に結合する転写調節因子並びにそれらの用途
Giorgi et al. Gene expression and in vitro release of galanin in rat hypothalamus during development
KR20230173105A (ko) 사춘기 이전의 비인간 동물 및 인간에서 사춘기를 예방 또는 지연시키기 위한 조성물 및 방법
Okada et al. Corticotroph Localization and Thyroid-Stimulating Hormone Receptor Binding Properties of Corticotroph-derived Glycoprotein Hormone
Qiu Kiss1 Neurons and Metabolic Sensing
Van Nguyen Roles of colony stimulating factor-1 in mammary gland development and transforming growth factor-β3 in mammary gland involution
Monts Peptides of the growth hormone-releasing hormone family: Expression, localization, and functions in testis
Rinkenberger et al. From Normalcy to Neoplasia. The Role of Epithelial-Stromal Interactions in Regulating Mammary Growth and Differentiation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

AK Designated states

Kind code of ref document: B1

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

AL Designated countries for regional patents

Kind code of ref document: B1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 155250

Country of ref document: IL

Ref document number: 2002532242

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2425712

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002211855

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 525389

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2001979945

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001979945

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 10398553

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 2002211855

Country of ref document: AU