US20040192606A1 - Method of promoting angiogenesis - Google Patents

Method of promoting angiogenesis Download PDF

Info

Publication number
US20040192606A1
US20040192606A1 US10/818,859 US81885904A US2004192606A1 US 20040192606 A1 US20040192606 A1 US 20040192606A1 US 81885904 A US81885904 A US 81885904A US 2004192606 A1 US2004192606 A1 US 2004192606A1
Authority
US
United States
Prior art keywords
relaxin
treatment
vegf
administration
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/818,859
Inventor
Elaine Unemori
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Corthera Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/818,859 priority Critical patent/US20040192606A1/en
Publication of US20040192606A1 publication Critical patent/US20040192606A1/en
Assigned to CONNETICS CORPORATION reassignment CONNETICS CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UNEMORI, ELAINE
Assigned to BAS MEDICAL, INC. reassignment BAS MEDICAL, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONNETICS CORPORATION
Assigned to CORHTERA, INC. reassignment CORHTERA, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BAS MEDICAL, INC.
Assigned to CORTHERA, INC. reassignment CORTHERA, INC. CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME PREVIOUSLY RECORDED ON REEL 022018 FRAME 0321. ASSIGNOR(S) HEREBY CONFIRMS THE CHANGE OF NAME. Assignors: BAS MEDICAL, INC.
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/2221Relaxins
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Abstract

Relaxin is useful for promoting angiogenesis and the treatment of infections or ischemic wounds where the injury results from lack of oxygen due to poor circulation.

Description

    RELATED APPLICATION
  • This application is a continuation-in-part of U.S. Provisional Application Serial No. 60/002,355, filed Aug. 15, 1995.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to the treatment of ischemic wounds, for example, where the injury results from lack of oxygen due to poor circulation such as in the diseases diabetes, scleroderma, and the like, by the administration of relaxin. The present invention also relates to the promotion of angiogenesis. [0002]
  • BACKGROUND INFORMATION
  • Mature human relaxin is a hormonal peptide of approximately 6000 daltons known to be responsible for remodelling the reproductive tract before parturition, thus facilitating the birth process. This protein appears to modulate the restructuring of connective tissues in target organs to obtain the required changes in organ structure during pregnancy and parturition. See, Hisaw, F. L., Proc. Soc. Exp. Biol. Med., 23: 661-663 (1926); Schwabe, C., et al., Biochem. Biophys. Res. Comm., 75: 503-570 (1977); James, R. et al., Nature, 267: 544-546 (1977). A concise review of relaxin was provided by Sherwood, D. in [0003] The Physiology of Reproduction, Chapter 16, “Relaxin”, Knobil, E. and Neill, J., et al. (eds.), (Raven Press Ltd., New York), pp. 585-673 (1988). Circulating levels of relaxin are elevated for the entire nine months of pregnancy and drop quickly following delivery.
  • While predominantly a hormone of pregnancy, relaxin has also been detected in the non-pregnant female as well as in the male. Bryant-Greenwood, G. D., Endocrine Reviews, 3: 62-90 (1982) and Weiss, G., Ann. Rev. Physiol., 46:43-52 (1984). [0004]
  • Relaxin has been purified from a variety of species including porcine, murine, equine, shark, tiger, rat, dogfish and human, and shows at least primary and secondary structural homology to insulin and the insulin-like growth factor. In the human, relaxin is found in most abundance in the corpora lutea (CL) of pregnancy. However, specific nuclei in the brain have relaxin receptors and other nuclei contain messenger RNA for relaxin. Several nuclei with cells bearing relaxin receptors are found in the area of the hypothalamus. [0005]
  • Two human gene forms have been identified, (H1) and (H2). Hudson, P., et al., Nature, 301: 628-631 (1983); Hudson, P., et al., The EMBO Journal, 3: 2333-2339 (1984); and U.S. Pat. Nos. 4,758,516 and 4,871,670. Only one of the gene forms (H2) has been found to be transcribed in CL. It remains unclear whether the (HI) form is expressed at another tissue site, or whether it represents a pseudo-gene. When synthetic human relaxin (H2) and certain human relaxin analogs were tested for biological activity, the tests revealed a relaxin core necessary for biological activity as well as certain amino acid substitutions for methionine that did not affect biological activity. Johnston, et al., in [0006] Peptides: Structure and Function, Proc. Ninth American Peptide Symposium, Deber, C. M., et al. (eds.) (Pierce Chem. Co. 1985).
  • Methods of making relaxin are also described in U.S. Pat. No. 4,835,251 and in co-pending U.S. Ser. Nos. 07/908,766 (PCT US90/02085) and 08/080,354 (PCT US94/0699). Methods of using relaxin in cardiovascular therapy and in the treatment of neurodegenerative diseases are described in U.S. Pat. No. 5,166,191 and in U.S. Ser. No. 07/902,637 (PCT US92/06927). Certain formulations of human relaxin are described in allowed U.S. Ser. No. 08/050,745. [0007]
  • Recombinant human relaxin (H2) in currently in Phase I human clinical trials in scleroderma patients. Scleroderma is a disease involving an imbalance in tissue reformation giving rise to the overproduction of collagen, and ultimately resulting in swelling and hardening of the skin (and affected organs). [0008]
  • Vascular Endothelial Growth Factor (VEGF) has also been localized in situ in the corpus luteum (CL) of pregnancy, as well as the placenta and the endometrium. See Sharkey et al., J. Reprod. Fert. 99:609-615 (1993); Li et al. Growth Factors 22:277-282 (1994); Phillips et al. Endocrinology 127:965-967 (1990). VEGF, highly conserved glycoprotein secreted by macrophages, exhibits a potent ability to induce new vessel growth in vivo. VEGF is mitogen specific for endothelial cells and can induce both endothelial cell migration and serine and metalloproteinase expression (for review, see Thomas, K. A., J. Biol. Chem. 271:603-606 (1996). The strongest sites of VEGF expression are the fetal and maternal macrophages. Besides its proposed role in promoting new vessel growth during pregnancy, VEGF has also been proposed to be involved in persistent and dysregulated vessel growth in pathological conditions such as tumor metastasis, diabetic retinopathy, and rheumatoid arthritis. [0009]
  • SUMMARY OF THE INVENTION
  • In one aspect, the invention relates to a method of promoting angiogenesis in a mammal in need thereof by administering a therapeutically effective amount of relaxin. In a preferred embodiment, relaxin is administered in an amount sufficient to maintain a serum concentration of at least about 1 ng/ml. In a further preferred embodiment the relaxin is recombinant human relaxin (H2). [0010]
  • In another aspect, the invention relates to the treatment of infections or ischemic wounds by administering a therapeutically effective amount of relaxin. In a particularly preferred embodiment, the infection or ischemic wound is one where injury has resulted from lack of oxygen due to poor circulation. [0011]
  • In yet another aspect of the invention, there is provided a method of using relaxin for the manufacture of a medicant for the treatment of an infection or ischemic wound, or for the manufacture of a medicant for the promotion of angiogenesis. In preferred versions of these embodiments, the relaxin is recombinant human relaxin (H2). [0012]
  • DETAILED DESCRIPTION OF THE INVENTION Definitions and General Parameters
  • As used in the present specification, the following words and phrases are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise. [0013]
  • “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. [0014]
  • The term “treatment” or “treating” means any therapeutic intervention in a mammal, including: [0015]
  • (i) prevention, that is, causing the clinical symptoms not to develop; [0016]
  • (ii) inhibition, that is, arresting the development of clinical symptoms;, and/or [0017]
  • (iii) relief, that is, causing the regression of clinical symptoms. [0018]
  • The term “effective amount” means a dosage sufficient to provide treatment for the disease state being treated. This will vary depending on the patient, the disease and the treatment being effected. [0019]
  • The term “relaxin” means human relaxin, including intact full length relaxin or a portion of the relaxin molecule that retains biological activity [as described in U.S. Pat. No. 5,023,321, preferably recombinant human relaxin (H2)] and other active agents with relaxin-like activity, such as Relaxin Like Factor (as described in co-pending application Ser. No. 08/484,219, relaxin analogs (as described in co-pending application Ser. No. 08/483,476, and agents that competitively displace bound relaxin from a receptor. Relaxin can be made by any method known to those skilled in the art, preferably as described in U.S. Pat. No. 4,835,251 and in co-pending U.S. Ser. Nos. 07/908,766 (PCT US90/02085) and 08/080,354 (PCT US94/0699). [0020]
  • The Role of Relaxin Promoting Angiogenesis
  • The invention is based, in part, on the surprising discovery that relaxin promotes angiogenesis in an in vivo assay, as described more fully below by way of working examples. Specifically, relaxin was found angiogenic in both a rabbit corneal injection protocol and by the matrigel subcutaneous insert vascularization protocol. [0021]
  • Also reported herein is the novel discovery that relaxin induces secretion of a potent angiogenic factor, Vascular Endothelial Growth Factor (“VEGF”), in the monocyte-like cell line THP-1. This finding further broadens the scope of relaxin's known biological activity. Since macrophages are known to play a key role in angiogenesis, both during and outside of pregnancy, relaxin's potential regulatory role straddles both. [0022]
  • In the THP-1 cell line, relaxin stimulates the expression of at least 3 of the 4 isoforms of VEGF: the 121, the 165, and the 189 amino acid isoforms. Although all forms are reportedly bioactive, the 121 and 165 amino acid forms are secreted while the larger molecules remain associated with the extracellular matrix unless enzymatically released. This stimulation of relaxin expression occurred at the transcriptional level, even in the presence of cycloheximide, indicating that no de novo protein synthesis was required. Additionally, results presented herein indicate that the induction of VEGF by relaxin in THP-1 cells may be mediated by cAMP and protein kinase C. This finding is consistent with the known role of cAMP in the stimulation of VEGF expression in other cell types. See, for example, Claffey et al., J. Biol. Chem. 267:16317-16322 (1992), and Garrido et al., Growth Factors 8:109-117 (1993). The rapid increase in VEGF transcripts following relaxin treatment of THP-1 cells is similar to that seen in preadipocytes following forskolin stimulation (Garrido et al., supra). Indeed, the rapidity of the induction and the lack of cycloheximide effect may suggest a pathway common to THP-1 cells and preadipocytes. [0023]
  • Utility, Testing and Administration
  • Utility [0024]
  • Relaxin is useful for promoting angiogenesis and treating infections and ischemic wounds (e.g., poorly healing ischemic ulcers) characteristic of diseases, such as diabetes and scleroderma, involving poorly vascularized disease sites and macrophage associated inflammation. Macrophages are one of the most important sources of angiogenic factors. It has surprisingly been discovered that certain macrophage lines contain relaxin binding sites. It has also surprisingly been discovered that relaxin promotes angiogenesis in vivo. [0025]
  • Testing [0026]
  • In vitro activity for relaxin binding to macrophages is determined using P[0027] 32 labeled relaxin binding sites.
  • In vivo activity for angiogenesis is determined by the rabbit corneal injection protocol and by the matrigel subcutaneous insert vascularization protocol. [0028]
  • Administration [0029]
  • Relaxin is administered at a therapeutically effective dosage, e.g., a dosage sufficient to promote angiogenesis and/or provide treatment for the above-referenced disease states. [0030]
  • Administration of relaxin can be via any of the accepted modes of administration for agents that serve similar utilities, preferably by systemic administration. [0031]
  • While human dosage levels for treating depression have yet to be optimized for relaxin, generally, a daily dose is from about 0.1 to 500.0 μg/kg of body weight per day, preferably about 6.0 to 200.0 μg/kg, and most preferably about 12.0 to 100.0 μg/kg. Generally it is sought to obtain a serum concentration of relaxin approximating or greater than normal circulating levels in pregnancy, i.e., 1.0 ng/ml, such as 0.5 to 50 ng/ml, preferably 1.0 to 20 ng/ml. In the ongoing clinical trials, dosages of about 6.0 μg/kg, 12.0 μg/kg and 50 μg/kg have respectively resulted in serum concentrations of about 1.8 ng/ml±0.3, 3.6 ng/ml±0.6, and 11.8 ng/ml±1.6. Thus, for administration to a 70 kg person, the dosage range would be about 7.0 μg to 3.5 mg per day, preferably about 42.0 μg to 2.1 mg per day, and most preferably about 84.0 to 700.0 μg per day. The amount of relaxin administered will, of course, be dependent on the subject and the severity of the affliction, the manner and schedule of administration and the judgment of the prescribing physician. [0032]
  • In employing relaxin for treatment of the above conditions, any pharmaceutically acceptable mode of administration can be used. Relaxin can be administered either alone or in combination with other pharmaceutically acceptable excipients, including solid, semi-solid, liquid or aerosol dosage forms, such as, for example, tablets, capsules, powders, liquids, gels, suspensions, suppositories, aerosols or the like. Relaxin can also be administered in sustained or controlled release dosage forms (e.g., employing a slow release bioerodable delivery system), including depot injections, osmotic pumps (such as the Alzet implant made by Alza), pills, transdermal and transcutaneous (including electrotransport) patches, and the like, for prolonged administration at a predetermined rate, preferably in unit dosage forms suitable for single administration of precise dosages. The compositions will typically include a conventional pharmaceutical carrier or excipient and relaxin. In addition, these compositions may include other active agents, carriers, adjuvants, etc. Generally, depending on the intended mode of administration, the pharmaceutically acceptable composition will contain about 0.1% to 90%, preferably about 0.5% to 50%, by weight of relaxin, the remainder being suitable pharmaceutical excipients, carriers, etc. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 15th Edition, 1975. The formulations of human relaxin described in U.S. Ser. No. 08/050,745 are particularly preferred. [0033]
  • Parenteral administration is generally characterized by injection, either subcutaneously, intradermally, intramuscularly or intravenously, preferably subcutaneously. Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like. In addition, if desired, the pharmaceutical compositions to be administered may also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, solubility enhancers, and the like, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, cyclodextrins, and the like. [0034]
  • The percentage of relaxin contained in such parenteral compositions is highly dependent on the specific nature thereof, as well as the needs of the subject. However, percentages of active ingredient of 0.01% to 10% in solution are employable, and will be higher if the composition is a solid which will be subsequently diluted to the above percentages. Preferably the composition will comprise 0.2-2% of the relaxin in solution. [0035]
  • A more recently devised approach for parenteral administration employs the implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained. Various matrices (e.g., polymers, hydrophilic gels, and the like) for controlling the sustained release, and for progressively diminishing the rate of release of active agents such as relaxin are known in the art. See, U.S. Pat. No. 3,845,770 (describing elementary osmotic pumps); U.S. Pat. Nos. 3,995,651, 4,034,756 and 4,111,202 (describing miniature osmotic pumps); U.S. Pat. Nos. 4,320,759 and 4,449,983 (describing multichamber osmotic systems referred to as push-pull and push-melt osmotic pumps); and U.S. Pat. No. 5,023,088 (describing osmotic pumps patterned for the sequentially timed dispensing of various dosage units). [0036]
  • Formulations of relaxin may also be administered to the respiratory tract as a nasal or pulmonary inhalation aerosol or solution for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose, or with other pharmaceutically acceptable excipients. In such a case, the particles of the formulation may advantageously have diameters of less than 50 microns, preferably less than 10 microns. See, e.g., U.S. Pat. No. 5,364,838, which discloses a method of administration for insulin that can be adapted for the administration of relaxin in the present invention.[0037]
  • EXAMPLES
  • The following examples are given to enable those skilled in the art to more clearly understand and to practice the present invention. They should not be considered as limiting the scope of the invention, but merely as being illustrative and representative thereof. [0038]
  • Example 1 Rabbit Corneal Assay
  • Test and control material is mixed with hydron into a 0.1 cc disc (resembling a small contact lens). The disc is implanted into a rabbit corneal stroma micro pocket using appropriate anesthesia for the procedure. After one week, neo-capillary growth toward the disc is measured and scored (0-4) utilizing a standard anatomic grading scale. A score of 0 means no growth. A score of 1 means capillaries (one or two) just entering the corneal stroma from the limbus toward the pocket. A score of 3 means vessels have grown to the base of the implant. A score of 4 means vessels have grown around the implant. [0039]
  • By following the above-described procedure testing sixteen blinded samples, one corneal vitreous was punctured and 15 scores were obtained. Mean placebo score (n=7) was 0.6 with a standard error of 0.3 while the mean relaxin score (n=8) was 1.5 with a standard error of 0.5. P value was 0.08. [0040]
  • In conclusion, relaxin was angiogenic when tested in this assay. [0041]
  • Example 2 Murine Matrigel Assay
  • Matrigel (Collaborative Biomedical, Bedford, Mass.) is a reconstituted basement membrane complex containing primarily laminin and type IV collagen. It is isolated from the Engelbreth-Holm-Swarm (EHS) murine tumor. At room temperature, matrigel is liquid; but when injected subcutaneously into a mouse it reconstitutes as a gel. [0042]
  • Test substance is mixed with matrigel (1%v/v media in 1 cc matrigel) at room temperature and then injected subcutaneously into a mouse dorsum (5 month old female Swiss-Webster). After one week, the mouse is sacrificed and the gel plug(s) harvested. Samples are fixed in buffered formalin, paraffin embedded, sectioned (4 μ) and H&E stained. The samples are scored 1 (positive), 0.5 (indeterminate) or 0 (negative). [0043]
  • Relaxin, when tested as described above, caused endothelial cells (confirmed with factor VIII staining) to migrate into the gel, organize linear structures, and form vessels with blood cells as compared with non-supplemented control gel plugs, which supported no endothelial cell infiltration. Sixteen blinded samples were tested. Sixteen scores were obtained. The mean control score (n=8) was 0.3 with a standard error of 0.1. The mean relaxin score was 0.8 with a standard error of 0.1. P value was 0.02. [0044]
  • In conclusion, relaxin was angiogenic when tested in this assay. [0045]
  • Example 3 Effect of Relaxin on VEGF Production in THP-1 Cells
  • A cell line derived from the peripheral blood cells of a 1 year old male with leukemia which has been described as monoblast or immature monocytes, THP-1, was chosen for studying the effect of relaxin on cultured cells. THP-1 cells (ATCC# TIB202) were grown in Iscove's medium supplemented with 10% fetal bovine serum and 2 mM L-glutamine. For experiments, THP-1 cells were cultured at 5×10[0046] 5 cells/ml in 24 well plates and incubated at 37° C. with 20% O2 and 5% CO2. Cells were treated with recombinant human relaxin (H2) in 10 mM citrate, pH 5.0 at doses ranging from 0.04-50 ng/ml, or with diluent alone, for 8 hours. Conditioned media and cells were then collected, and cells removed by centrifugation at 500 g for 5 minutes. VEGF protein secretion was quantified in an ELISA kit (R & D Systems, Minneapolis, Minn.).
  • Relaxin caused no observable morphological changes, such as adherence to plastic or clumping, in THP-1 cells, nor did it influence thymidine uptake, metalloproteinase expression patterns, or nitric oxide production. However, relaxin was shown to induce VEGF protein secretion is a dose dependent manner, peaking at 1 ng/ml relaxin. [0047]
  • A time-course study of expression following treatment with 1 ng/ml and 100 ng/ml relaxin demonstrated that VEGF increased in the conditioned media in roughly linear fashion up to at least 72 hours. [0048]
  • This same assay was used to compare the effect of other members of the relaxin family, specifically insulin, IGF-I, and IGF-II (Promega, Madison, Wis.), on VEGF induction at equivalent molar concentrations (17 nM). Neither IGF-I and IGF-II induced VEGF production. However, insulin stimulated a small but significant amount of VEGF expression, roughly 30% of that induced by relaxin. [0049]
  • Because estrogen treatment appears to upregulate binding of relaxin to some cell types, THP-1 cells were treated with 1 μM 17-β-estradiol simultaneously with relaxin, or pretreated for 48 hours prior to relaxin addition. Neither treatment protocol resulted in a difference in VEGF stimulation when compared to relaxin treatment alone. [0050]
  • Thus, the results showed that relaxin induces VEGF secretion in THP-1 cells in a dose dependent fashion which is independent of the effects of estrogen. [0051]
  • Example 4 Relaxin Regulation of VEGF Transcription
  • In order to determine whether relaxin-induced increases in VEGF protein were reflected at the transcript level and to characterize VEGF isoform regulation, PCR analysis of steady state VEGF mRNA was performed. Total RNA was extracted from 10[0052] 6 THP-1 cells following a 2 hour treatment with recombinant human relaxin (H2) (10 ng/ml) using RNAzol (Tel-Test, Tex.), according to the manufacturer's instructions. Primers designed to amplify and distinguish among all four VEGF isoforms were used: 5′-CCA TGA ACT TTC TGC CCT-3′ (sense) (SEQ ID NO: 1) and 5′-TGC ATC GTT CTG TAT CAG TCT-3′ (antisense) (SEQ ID NO: 2). These primers spanned the gene from exon 1 to exon 8 and generated 4 different product sizes based on the number of exons spliced out during mRNA processing. A product size of 520 bp indicates the presence of the VEGF121 transcript, a size of 650 bp corresponds to VEGF165, a 730 bp band to VEGF189, and a 780 bp band to VEGF206. 0.7 μM of each primer was used in a reaction mixture with 2.5 U Taq polymerase. Samples were denatured at 95° C. for 30 s, annealed at 60° C. for 30 s, and extension performed at 72° C. for 30 s. Amplification occurred over 30 cycles.
  • PCR products were size-fractionated on a 1.5% agarose gel and stained with ethidium bromide. The gel results showed that recombinant human relaxin (H2) increased the levels of expression of three bands corresponding to VEGF121, 165, and 189. The increase was detectable at 30 minutes, and was roughly 2-fold by 2 hours. The addition of cycloheximide did not interfere with or amplify this increase, indicating that de novo protein synthesis is not involved in this effect. [0053]
  • Southern blot analysis of the PCR products was then performed using probes that distinguish among the transcripts. The gel-fractionated PCR products were transferred to Genescreen and probed with [0054] 32P-end labelled probes specific to sequences within exons 4, 6, and 7. Probes were designed as follows: Ex4: 5′-TTC CTA CAG CAC AAC AAA TGT GAA TGC-3′ (SEQ ID NO: 3); Ex6: 5′-AAA TCA GTT CGA GGA AAG GGA AAG-3′ (SEQ ID NO: 4); Ex7: 5′-AAG CAT TTG TTT GTA CAA GAT G-3′ (SEQ ID NO: 5). The Ex4 probe, designed according to the sequence within exon 4 of the VEGF gene, recognized transcripts of all 4 isoforms on Southern blotting. The Ex6 probe recognized transcripts corresponding to VEGF189 and 206, and the Ex7 probe hybridized to transcripts of the VEGF165, 189, and 206 isoforms. Hybridizations were carried out in 6×SSC/0.01% SDS and 10×Denhardts for 2 hours at 56° C. Blots were washed in 2×SSC/0.1% SDS for 2 hours, then exposed to Kodak X-Omat X ray film.
  • The Ex4 probe recognized three distinct bands, the Ex6 probe one band, and the Ex7 probe two bands. These results indicated that transcripts of VEGF121, 165, and 189 were present in THP-1 cells. A second larger band, presumably that corresponding to VEGF206, is occasionally recognized by the Ex6 probe. Importantly, the intensity of all bands is increased following addition of relaxin (100 ng/ml), indicating that relaxin stimulates VEGF production at the transcriptional level. [0055]
  • Example 5 Second Messenger Pathways in VEGF Stimulation
  • Since relaxin causes an increase in cAMP in THP-1 cells, we investigated the potential role for this mediator in relaxin-induced VEGF secretion. Therefore, several different agents known to alter second messenger pathways were tested for their effect on VEGF secretion in THP-1 cells. For these experiments, forskolin, dbcAMP, dbcGMP and isobutylmethylxanthing (IBMX) were obtained from Sigma (St. Louis, Mo.). Cholera toxin, pertussis toxin, H-89, and bisindolylmalemide were purchased from Calbiochem (La Jolla, Calif.), and Ro, staurosporin, KT5720, and SQ22536 were obtained from Biomol (Plymouth Meeting, Pa.). The results of these experiments are shown below in Table I. [0056]
    TABLE I
    VEGF Regulation in THP-1 Cells
    Net VEGF
    Exp # Agent Dose Expr pg/ml)* (n)#
    1 IBMX  50 × 10−6M 154 ± 46 (3)
    2 IBMX  50 × 10−6M 209 ± 20 (3)
    RLX 10 ng/ml 161 ± 19 (3)
    IBMX + RLX 422 ± 37 (3)
    3 Ro 500 × 10−6M 201.5 ± 9.7  (3)
    3 dbcAMP 10−6M -0- (3)
    10−5M 142 ± 26 (3)
    10−4M 488 ± 27 (3)
    dbcGMP 10−6M-10−4M -0- (3 each dose)
    4 Cholera toxin  1 μg/ml 1278 ± 106 (3)
    Pertussis toxin  1 μg/ml -0- (3)
    5 PMA  1 ng/ml 342 ± 18 (3)
  • THP-1 cells were treated with agents for 8 h, except in Experiment 1, where the cells were treated for 24 h. Media were collected, and assayed for VEGF content by ELISA. Results are expressed as net VEGF expression calculated by subtracting the VEGF level expressed by the appropriate placebo-treated control group from the experimental group. [0057]
  • * Means±SEM, p<0.01 vs placebo-treated group [0058]
  • # Numbers in parentheses indicate number of paired wells used to calculate mean differences. [0059]
  • Treatment of THP-1 cells with IBMX, a phosphodiesterase inhibitor, significantly increased their secretion of VEGF. THP-1 cells treated with recombinant human relaxin (H2) (1 ng/ml) in the presence of IBMX secreted more VEGF than cells treated with either relaxin or IBMX alone. A more specific cAMP phosphodiesterase, Ro, also increased VEGF expression. Forskolin, a diterpene that stimulates adenyl cyclase, induced VEGF production a dose-dependent bi-phasic manner. Dibutyryl cAMP (at 10[0060] −6 M and 10−4 M), but not dibutyryl cGMP, also stimulated VEGF expression in a dose-dependant fashion. Cholera toxin, which elevates cAMP levels in these cells, stimulated VEGF expression while pertussis toxin did not.
  • Because previous experiments have suggested a role for Protein Kinase C in the induction of VEGF gene expression, we tested the ability of PMA to modulate VEGF production in THP-1 cells. PMA (1 ng/ml) significantly increased VEGF protein expression. However, at this same dose, PMA does not increase cAMP levels in these cells (data not shown). [0061]
  • Selective inhibitors of PKA and PKC were used to block VEGF induction by relaxin. PKA inhibitors (H-89, SQ22536, KT5720) used at or above their K[0062] i doses failed to completely inhibit VEGF stimulation, as measured by ELISA (data not shown). Furthermore, combining PKA and PKC inhibitors, KT5720 and bisindolylmaleimide respectively, did not inhibit the response over that of KF5720 alone.
  • While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto. All patents and publications cited above are hereby incorporated by reference. [0063]

Claims (22)

What is claimed is:
1. A method for treating an infection or ischemic wound in a mammal in need thereof comprising administering a therapeutically effective amount of relaxin.
2. The method of treatment of claim 1 comprising the administration of relaxin in an amount sufficient to maintain a serum concentration of at least about 1 ng/ml.
3. The method of treatment of claim 1 comprising the administration of recombinant human relaxin (H2).
4. The method of treatment of claim 2 comprising the administration of recombinant human relaxin (H2).
5. The method of treatment of claim 3 wherein said relaxin is administered parenterally.
6. The method of treatment of claim 5 wherein said relaxin is administered by subcutaneous injection.
7. The method of treatment of claim 3 wherein said relaxin is administered by inhalation to the patient's respiratory tract.
8. The method of treatment of claim 3 wherein said infection or ischemic wound is one where the injury is the result from lack of oxygen due to poor circulation.
9. The method of treatment of claim 8 wherein said lack of oxygen due to poor circulation is the result of a disease.
10. The method of treatment of claim 9 wherein the disease is scleroderma or diabetes.
11. The method of treatment of claim 1 wherein the wound is an ischemic ulcer.
12. A method for promoting angiogenesis in a mammal in need thereof comprising administering a therapeutically effective amount of relaxin.
13. The method of claim 12 comprising the administration of relaxin in an amount sufficient to maintain a serum concentration of at least about 1 ng/ml.
14. The method of claim 12 comprising the administration of recombinant human relaxin (H2).
15. The method of claim 13 comprising the administration of recombinant human relaxin (H2).
16. The method of claim 14 wherein said relaxin is administered parenterally.
17. The method of claim 16 wherein said relaxin is administered by subcutaneous injection.
18. The method of claim 14 wherein said relaxin is administered by inhalation to the patient's respiratory tract.
19. A method of using relaxin for the manufacture of a medicant for the treatment of an infection or ischemic wound.
20. The method of claim 19 wherein the relaxin is recombinant human relaxin (H2).
21. A method of using relaxin for the manufacture of a medicant for the promotion of angiogenesis.
22. The method of claim 21 wherein the relaxin is recombinant human relaxin (H2).
US10/818,859 1995-08-15 2004-04-05 Method of promoting angiogenesis Abandoned US20040192606A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/818,859 US20040192606A1 (en) 1995-08-15 2004-04-05 Method of promoting angiogenesis

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US235595P 1995-08-15 1995-08-15
US08/698,359 US6211147B1 (en) 1995-08-15 1996-08-15 Method of promoting angiogenesis using relaxin
US09/780,758 US6780836B2 (en) 1995-08-15 2001-02-08 Method of promoting angiogenesis using relaxin
US10/818,859 US20040192606A1 (en) 1995-08-15 2004-04-05 Method of promoting angiogenesis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/780,758 Continuation US6780836B2 (en) 1995-08-15 2001-02-08 Method of promoting angiogenesis using relaxin

Publications (1)

Publication Number Publication Date
US20040192606A1 true US20040192606A1 (en) 2004-09-30

Family

ID=21700396

Family Applications (3)

Application Number Title Priority Date Filing Date
US08/698,359 Expired - Lifetime US6211147B1 (en) 1995-08-15 1996-08-15 Method of promoting angiogenesis using relaxin
US09/780,758 Expired - Lifetime US6780836B2 (en) 1995-08-15 2001-02-08 Method of promoting angiogenesis using relaxin
US10/818,859 Abandoned US20040192606A1 (en) 1995-08-15 2004-04-05 Method of promoting angiogenesis

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/698,359 Expired - Lifetime US6211147B1 (en) 1995-08-15 1996-08-15 Method of promoting angiogenesis using relaxin
US09/780,758 Expired - Lifetime US6780836B2 (en) 1995-08-15 2001-02-08 Method of promoting angiogenesis using relaxin

Country Status (9)

Country Link
US (3) US6211147B1 (en)
EP (1) EP0845992B1 (en)
JP (1) JP4568383B2 (en)
AT (1) ATE228004T1 (en)
AU (1) AU6777196A (en)
CA (1) CA2229479C (en)
DE (1) DE69624925T2 (en)
ES (1) ES2187667T3 (en)
WO (1) WO1997006814A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050238639A1 (en) * 2004-03-18 2005-10-27 Bas Medical, Inc. Use of relaxin to increase arterial compliance
US20060247172A1 (en) * 2004-04-30 2006-11-02 Bas Medical, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US20090232670A1 (en) * 2005-08-29 2009-09-17 Heng-Yi Lai Compressor muffler
US20100048475A1 (en) * 2008-05-16 2010-02-25 Corthera, Inc. Method of Treating Dyspnea Associated with Acute Heart Failure
US20110172159A1 (en) * 2008-09-05 2011-07-14 Coelho De Sampaio Tatiana Lobo Proteic acid polymer, production processes, use of proteic acid polymer, pharmaceutical composition and method of treatment
US11161891B2 (en) 2015-12-09 2021-11-02 The Scripps Research Institute Relaxin immunoglobulin fusion proteins and methods of use

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1253929B1 (en) * 2000-02-09 2007-05-23 BAS Medical, Inc. Use of relaxin to treat diseases related to vasoconstriction
ITRM20020119A1 (en) * 2002-03-05 2003-09-05 Geymonat Spa COMPOSITIONS CONTAINING PLGF FOR PHARMACEUTICAL AND COSMETIC USE.
CA2559745C (en) * 2004-03-19 2010-11-02 Bas Medical, Inc. Use of relaxin to increase arterial compliance
US7893244B2 (en) * 2005-04-12 2011-02-22 Intradigm Corporation Composition and methods of RNAi therapeutics for treatment of cancer and other neovascularization diseases
CA2604441A1 (en) * 2005-04-12 2006-10-19 Intradigm Corporation Composition and methods of rnai therapeutics for treatment of cancer and other neovascularization diseases
US20060281669A1 (en) * 2005-06-13 2006-12-14 Yue Samuel K Method and compositions for the treatment of diabetes and related complications
US8986253B2 (en) 2008-01-25 2015-03-24 Tandem Diabetes Care, Inc. Two chamber pumps and related methods
EP2288367B1 (en) * 2008-05-16 2015-10-07 Corthera, Inc. H2 relaxin for use in the treatment of premature cervical dilation
EP2278993A4 (en) * 2008-05-16 2012-09-12 Corthera Inc Method of promoting wound healing
US8408421B2 (en) 2008-09-16 2013-04-02 Tandem Diabetes Care, Inc. Flow regulating stopcocks and related methods
US8650937B2 (en) 2008-09-19 2014-02-18 Tandem Diabetes Care, Inc. Solute concentration measurement device and related methods
EP2724739B1 (en) 2009-07-30 2015-07-01 Tandem Diabetes Care, Inc. Portable infusion pump system
HUE045845T2 (en) 2010-08-17 2021-12-28 Ambrx Inc Modified relaxin polypeptides and their uses
WO2013033324A2 (en) * 2011-08-31 2013-03-07 University Of Florida Research Foundation, Inc. Materials and methods for modulating activity of bone marrow derived cells
US9180242B2 (en) 2012-05-17 2015-11-10 Tandem Diabetes Care, Inc. Methods and devices for multiple fluid transfer
US9555186B2 (en) 2012-06-05 2017-01-31 Tandem Diabetes Care, Inc. Infusion pump system with disposable cartridge having pressure venting and pressure feedback
US9173998B2 (en) 2013-03-14 2015-11-03 Tandem Diabetes Care, Inc. System and method for detecting occlusions in an infusion pump
US9526762B1 (en) * 2013-08-09 2016-12-27 William Marsh Rice University Multidomain peptides for promoting angiogenesis

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023321A (en) * 1982-12-13 1991-06-11 Howard Florey Institute Of Experimental Physiology & Medicine Molecular cloning and characterization of a further gene sequence coding for human relaxin
US5023088A (en) * 1987-06-25 1991-06-11 Alza Corporation Multi-unit delivery system
US5166191A (en) * 1991-08-19 1992-11-24 Genentech, Inc. Use of relaxin in cardiovascular therapy
US5451572A (en) * 1988-02-26 1995-09-19 Genentech, Inc. Human relaxin formulation
US5464756A (en) * 1989-05-04 1995-11-07 Genentech Processes and compositions for the isolation of human relaxin
US5478807A (en) * 1991-08-19 1995-12-26 Genentech, Inc. Use of relaxin in the treatment of bradycardia
US5753623A (en) * 1995-06-07 1998-05-19 Connetics Corporation Method of treatment for depression
US5811395A (en) * 1995-06-07 1998-09-22 Medical University Of South Carolina Relaxin analogs and derivatives methods and uses thereof
US5911997A (en) * 1995-06-07 1999-06-15 Connetics Corporation Relaxin-like factor and methods and uses thereof
US5952296A (en) * 1993-07-27 1999-09-14 Bigazzi; Mario Method of using relaxin as therapeutic or preventing agent

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8907799D0 (en) * 1989-04-06 1989-05-17 Erba Carlo Spa Heteroaryl-3-oxo-propanenitrile derivatives useful in the treatment of rheumatoid arthritis and other autoimmune diseases
US5194596A (en) * 1989-07-27 1993-03-16 California Biotechnology Inc. Production of vascular endothelial cell growth factor

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023321A (en) * 1982-12-13 1991-06-11 Howard Florey Institute Of Experimental Physiology & Medicine Molecular cloning and characterization of a further gene sequence coding for human relaxin
US5023088A (en) * 1987-06-25 1991-06-11 Alza Corporation Multi-unit delivery system
US5451572A (en) * 1988-02-26 1995-09-19 Genentech, Inc. Human relaxin formulation
US5464756A (en) * 1989-05-04 1995-11-07 Genentech Processes and compositions for the isolation of human relaxin
US5166191A (en) * 1991-08-19 1992-11-24 Genentech, Inc. Use of relaxin in cardiovascular therapy
US5478807A (en) * 1991-08-19 1995-12-26 Genentech, Inc. Use of relaxin in the treatment of bradycardia
US5952296A (en) * 1993-07-27 1999-09-14 Bigazzi; Mario Method of using relaxin as therapeutic or preventing agent
US5753623A (en) * 1995-06-07 1998-05-19 Connetics Corporation Method of treatment for depression
US5811395A (en) * 1995-06-07 1998-09-22 Medical University Of South Carolina Relaxin analogs and derivatives methods and uses thereof
US5911997A (en) * 1995-06-07 1999-06-15 Connetics Corporation Relaxin-like factor and methods and uses thereof

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8602998B2 (en) 2004-03-18 2013-12-10 University of Pittsburgh—of the Commonwealth System of Higher Education Use of relaxin to increase arterial compliance
US7878978B2 (en) 2004-03-18 2011-02-01 University Of Pittsburgh- Of The Commonwealth System Of Higher Education Use of relaxin to increase arterial compliance
US20050238639A1 (en) * 2004-03-18 2005-10-27 Bas Medical, Inc. Use of relaxin to increase arterial compliance
US8026215B2 (en) 2004-04-30 2011-09-27 Corthera, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US20060247172A1 (en) * 2004-04-30 2006-11-02 Bas Medical, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US20060247163A1 (en) * 2004-04-30 2006-11-02 Bas Medical, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US7553813B2 (en) 2004-04-30 2009-06-30 Corthera, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US20090318356A1 (en) * 2004-04-30 2009-12-24 Corthera, Inc. Methods and compositions for control of fetal growth via modulation of relaxin
US20090232670A1 (en) * 2005-08-29 2009-09-17 Heng-Yi Lai Compressor muffler
US8415301B2 (en) 2008-05-16 2013-04-09 Corthera, Inc. Method of treating dyspnea associated with acute heart failure by administering H2 relaxin
US8053411B2 (en) 2008-05-16 2011-11-08 Corthera, Inc. Method of treating dyspnea associated with acute heart failure
US8372809B2 (en) 2008-05-16 2013-02-12 Corthera, Inc. Method of treating dyspnea associated with acute heart failure
US20100048475A1 (en) * 2008-05-16 2010-02-25 Corthera, Inc. Method of Treating Dyspnea Associated with Acute Heart Failure
US9066916B2 (en) 2008-05-16 2015-06-30 Corthera, Inc. Method of treating neurohormonal imbalance
US9205132B2 (en) 2008-05-16 2015-12-08 Corthera, Inc. Method of lowering mortality
US9579363B2 (en) 2008-05-16 2017-02-28 Corthera Inc. Method of lowering mortality
US20110172159A1 (en) * 2008-09-05 2011-07-14 Coelho De Sampaio Tatiana Lobo Proteic acid polymer, production processes, use of proteic acid polymer, pharmaceutical composition and method of treatment
US11161891B2 (en) 2015-12-09 2021-11-02 The Scripps Research Institute Relaxin immunoglobulin fusion proteins and methods of use

Also Published As

Publication number Publication date
JPH11511174A (en) 1999-09-28
WO1997006814A1 (en) 1997-02-27
US6211147B1 (en) 2001-04-03
JP4568383B2 (en) 2010-10-27
US6780836B2 (en) 2004-08-24
EP0845992A4 (en) 2000-04-19
EP0845992A1 (en) 1998-06-10
EP0845992B1 (en) 2002-11-20
ATE228004T1 (en) 2002-12-15
DE69624925T2 (en) 2003-09-04
ES2187667T3 (en) 2003-06-16
DE69624925D1 (en) 2003-01-02
CA2229479A1 (en) 1997-02-27
CA2229479C (en) 2010-03-30
AU6777196A (en) 1997-03-12
US20010018418A1 (en) 2001-08-30

Similar Documents

Publication Publication Date Title
US6211147B1 (en) Method of promoting angiogenesis using relaxin
KR101058467B1 (en) Cortical Stimulating Hormone Analogs and Related Methods
Holsboer et al. Effects of intravenous corticotropin-releasing hormone upon sleep-related growth hormone surge and sleep EEG in man
Greb et al. Vascular endothelial growth factor in primate endometrium is regulated by oestrogen-receptor and progesterone-receptor ligands in vivo.
FERRARA et al. The 16K fragment of prolactin specifically inhibits basal or fibroblast growth factor stimulated growth of capillary endothelial cells
EP0309100B1 (en) Use of amylin or CGRP for the treatment of diabetes mellitus
Rehman et al. Use of intralesional verapamil to dissolve Peyronie’s disease plaque: a long-term single-blind study
AU781084B2 (en) Nicotine in therapeutic angiogenesis and vasculogenesis
Tomas et al. Effect of glucocorticoid administration on the rate of muscle protein breakdown in vivo in rats, as measured by urinary excretion of N τ-methylhistidine
Fine et al. Loss of central nervous system component of dopaminergic inhibition of prolactin secretion in patients with prolactin-secreting pituitary tumors
CA2397200C (en) Use of relaxin to treat diseases related to vasoconstriction
Koulu et al. The effect of methysergide, pimozide, and sodium valproate on the diazepam-stimulated growth hormone secretion in man
Leung et al. Mechanism (s) by which adrenalectomy and corticosterone influence prolactin release in the rat
Jeejeebhoy et al. Effects of hormones on the synthesis of α1 (acute-phase) glycoprotein in isolated rat hepatocytes
Lehtinen et al. Modifying effects of epidural analgesia or general anesthesia on the stress hormone response to laparoscopy for in vitro fertilization
Papachristou et al. Glucocorticoids regulate somatostatin peptide and steady state messenger ribonucleic acid levels in normal rat tissues and in a somatostatin-producing islet tumor cell line (1027B2)
US5753623A (en) Method of treatment for depression
EP1522311A1 (en) Use of somatostatin or analogs thereof for the preparation of a medicament for the regulation of ovarian follicles reserve by non menopausaled women
MXPA04002125A (en) Use of lh in controlled ovarian hyperstimulation.
AU757803B2 (en) Gene therapy for regulating smooth muscle cell tone
Crosignani et al. Metergoline treatment of hyperprolactinemic states
GLUCKMAN et al. Hormone Ontogeny in the Ovine Fetus, XV. Studies of Adenohypophysial Hormones after Fetal Pituitary Stalk Section: Evidence for an Extrahypothalamic Dopaminergic Influence on Fetal Prolactin Secretion
EP1854476A2 (en) Use of relaxin to treat diseases related to vasoconstriction
Cavagnini et al. Evidence against a self-inhibition of ACTH secretion in man
Stoll Mechanisms of Regression and Revised Approaches to Endocrine Therapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: CONNETICS CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UNEMORI, ELAINE;REEL/FRAME:015612/0097

Effective date: 19980223

AS Assignment

Owner name: BAS MEDICAL, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CONNETICS CORPORATION;REEL/FRAME:018879/0488

Effective date: 20030715

AS Assignment

Owner name: CORHTERA, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:BAS MEDICAL, INC.;REEL/FRAME:022018/0321

Effective date: 20080211

AS Assignment

Owner name: CORTHERA, INC., CALIFORNIA

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE ASSIGNEE NAME PREVIOUSLY RECORDED ON REEL 022018 FRAME 0321;ASSIGNOR:BAS MEDICAL, INC.;REEL/FRAME:022535/0632

Effective date: 20080211

STCB Information on status: application discontinuation

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