CA2659664C - Improved radiation therapy methods - Google Patents

Improved radiation therapy methods Download PDF

Info

Publication number
CA2659664C
CA2659664C CA2659664A CA2659664A CA2659664C CA 2659664 C CA2659664 C CA 2659664C CA 2659664 A CA2659664 A CA 2659664A CA 2659664 A CA2659664 A CA 2659664A CA 2659664 C CA2659664 C CA 2659664C
Authority
CA
Canada
Prior art keywords
seq
artificial sequence
pro
tyr
analogues
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.)
Expired - Fee Related
Application number
CA2659664A
Other languages
French (fr)
Other versions
CA2659664A1 (en
Inventor
Kathleen E. Rodgers
Gere Dizerega
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Southern California USC
Original Assignee
University of Southern California USC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Southern California USC filed Critical University of Southern California USC
Publication of CA2659664A1 publication Critical patent/CA2659664A1/en
Application granted granted Critical
Publication of CA2659664C publication Critical patent/CA2659664C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0819Tripeptides with the first amino acid being acidic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • A61K38/085Angiotensins
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0821Tripeptides with the first amino acid being heterocyclic, e.g. His, Pro, Trp
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1021Tetrapeptides with the first amino acid being acidic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/14Angiotensins: Related peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0644Platelets; Megakaryocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/32Angiotensins [AT], angiotensinogen

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Vascular Medicine (AREA)
  • Immunology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention provides methods and kits for mitigating radiation induced tissue damage, improving the effectiveness of radiation therapy, to support bone marrow transplantation, and promoting megakaryocyte production and mobilization and platelet production, each method comprising the administration of an effective amount of angiotensinogen, angiotensin I(AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof or AII AT2 type 2 receptor agonists.

Description

IMPROVED RADIATION THERAPY METHODS

This is a divisional application of Canadian Patent Application No. 2,323,237 filed March 8, 1999.

Background of the Invention Radiation therapy is currently one of the most useful methods of treating cancerous tumors. However, radiation therapy damages normal tissue surrounding the tumor (U.S. Patent No. 5,599,712).
This damage can include fibrosis, remodeling of the extracellular matrix, vascular damage, aberrant angiogenesis, pneumonitis, atherogenesis, osteonecrosis, mucositis, immunosuppression and functional impairment (U.S. Patent No. 5,616,561).
As a result of these radiation-induced side effects, techniques have been developed to minimize radiation-induced damage to surrounding normal tissues by limiting radiation to the lowest level effective for cancer treatment. Since there is a direct relationship between the amount of radiation and the effectiveness of the treatment, this method compromises the overall effectiveness of the treatment.

For some cancer patients, hematopoietic toxicity frequently limits the opportunity for radiation dose escalation (Watanabe et al., British J
Haematol.

94:619-627 (1996)). Repeated or, high dose cycles of radiation therapy may be responsible for severe stem cell depletion leading to important long-term hematopoietic sequelea and marrow exhaustion (Masse et al., Blood 91:441-449 (1998). Such stem cell depletion leads to depletion of the full range of hematopoietic lineage specific cells, including megakaryocytes, platelets, monocytes, neutrophils, and lymphocytes, and the resulting complications of ,such depletion. For example, in patients suffering from depressed levels of platelets (thrombocytopenia) the inability to form clots is the most immediate and serious consequence, a potentially fatal complication of many therapies for cancer. Such cancer patients are generally treated for this problem with platelet transfusions. Other patients frequently requiring platelet to transfusions are those undergoing bone marrow transplantation or patients with aplastic anemia. Platelets for such procedures are obtained by plateletpheresis from normal donors. Like most human blood products, platelets for transfusion have a relatively short shelf-life and also expose the patients to considerable risk of exposure to dangerous viruses, such as the human immunodeficiency virus (HIV).
The administration of hematopoietic growth factors may reduce short-term side effects induced by radiation, but has been hypothesized to cause long-term hematopoietic damage (Masse et al., 1998; Watanabe et al., 1996). Several studies have suggested that co-administration of negative hematopoietic regulators can minimize radiation therapy-induced. myelotoxicity by reducing the number of progenitor cells that enter the cell cycle. (Watanabe et al., 1996: Dunlop et al., Blood 79:2221-2225 (1992); Paukovits et al., Blood 81:1755-1761; Bogden et A, Annals N.Y. Acad. Sci. 628:126-139 (1991); Deeg et al., Ann. Hematol. 74:117-122 (1997);
Masse et al., 1998). This treatment is based on the premise that hematopoietic stem cells are relatively protected from radiation-related toxicity when quiescent, particularly when the malignant cells are proliferating (Deeg et al., (1997)).
Bone marrow contains pluripotent stem cells that are capable of reconstituting the entire hematopoietic system. Bone marrow transplantation has been used to treat various intractable hematopoietic diseases including leukemia and severe aplastic anemia. (U.S 1 Patent No. 5,186,931).
Typically, a bone marrow transplant patient is subjected to irradiation to reduce the leukocyte count to zero, followed by transplantation of bone marrow cells which function by producing a sufficient number of normal leukocytes. However, various complications, such as death, infectious diseases, graft versus host disease, radiation = nephritis, and interstitial pneumonia frequently occur during the time period between transplantation and the return to normal white blood cell levels after transplantation.
As a result of these frequent side effects, no satisfactory methods are currently available for supporting bone marrow transplantation which are capable of both increasing survival of bone marrow transplant patients and also accelerating the reconstitution of the hematopoietic system of the patient.
Chronic radiation injuries, such as radiation nephropathy, have been viewed as inevitable, progressive and untreatable (Moulder et al., Bone Marrow Transplantation 19:729-735 (1997)). The progressive and untreatable nature of late tissue damage follows from the assumption that the injury is due to delayed mitotic cell death resulting from genetic injury that is produced and irrevocably fixed in place at the time of irradiation (Moulder et at., 1997). Under this view, the only way to decrease the probability of injury is by limiting the radiation dose or shielding the at risk organs.

However, recent results indicate that late-onset radiation-induced tissue injury involves complex and dynamic interactions among parenchymal and vascular cells within a particular organ (Moulder et al., 1997). This model of chronic radiation injury suggests that pharmacological intervention after radiation exposure would be effective.
Thus, despite advances in the field of radiation therapy, prior art methods have proven to be of limited utility in minimizing radiation-induced tissue damage, and improving the efficacy of tumor radiation therapy and bone marrow transplantation.
Thus, there is a need for improved therapeutic methods to mitigate radiation induced to tissue damage and to improve the effectiveness of radiation therapy.
Furthermore, the ability to stimulate endogenous platelet formation in thrombocytopenic patients with a concomitant reduction in their dependence on platelet transfusion would be of great benefit. In addition the ability to correct or prevent thrombocytopenia in patients undergoing radiation therapy or chemotherapy for cancer would make such treatments safer and possibly permit increases in the intensity of the therapy thereby yielding greater anti-cancer effects.

Summary of the Invention In one aspect, the present invention provides methods and kits for mitigating radiation induced tissue damage, improving the effectiveness of radiation therapy, to support bone marrow transplantation, and promoting megakaryocyte production and mobilization and platelet production, each method comprising the administration of angiotensinogen, angiotensin I (Al), Al analogues, Al fragments and analogues thereof, angiotensin II (All), All analogues, All fragments or analogues thereof or All AT2 type 2 receptor agonists to a patient in need thereof.
Specific aspects of the invention include:
- a use of an effective amount of at least one active agent for increasing megakaryocyte production and mobilization, platelet production or a combination thereof, the active agent comprising a peptide with an amino acid sequence selected_from the group consisting of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO:
18, SEQ ID NO: 31, and SEQ ID NO: 38; and - a megakaryocyte production and mobilization kit, a platelet production kit or a combination thereof, comprising:
(a) an amount effective to support increasing megakaryocyte production and mobilization, platelet production or a combination thereof of at least one active agent comprising a peptide with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO: 18, SEQ ID
NO: 31, and SEQ ID NO: 38, and (b) instructions for using the effective amount of active agent for increasing megakaryocyte production and mobilization, platelet production or a combination thereof.

In another aspect of the present invention, an improved cell culture medium and kits are provided for the production of megalcaryocy-tes and platelets wherein the improvement comprises addition to the cell culture medium of an effective amount of angiotensinogen, Al. Al analogues, Al fragments and analogues thereof; All, All analogues, All fragments or analogues thereof or All AT, type 2 receptor 4gonists.
These aspects and other aspects of the invention become apparent in light of the following detailed description.

Brief Description of the Drawings Figure 1 is a graph showing the effect of All treatment two days prior to exposure on post-irradiation mouse mortality.
Figure 2 is a graph showing the effect of All treatment on the day of exposure on post-irradiation mouse mortality.
is Figure 3 is a graph showing the effect of All treatment two days following exposure on post-irradiation mouse mortality.
Figure 4 is a graph showing the effect of All treatment two days prior to exposure on white blood cell number after irradiation.
Figure 5 is a graph showing the effect of All treatment on the day of exposure on white blood cell number after irradiation.
Figure 6 is a graph showing the effect of Al! treatment two days following exposure on white blood cell number after irradiation.

5a Figure 7 is a graph showing the effect of All treatment two days prior to exposure on megakaryocyte number after irradiation.
Figure 8 is a graph showing the effect of All treatment on the day of exposure on megakaryocyte number after irradiation.
Figure 9 is a graph showing the effect of All treatment two days following exposure on megakaryocyte percentage after irradiation.
Figure 10 is a graph showing the effect of All treatment two days prior to exposure on monocyte number after irradiation.
Figure 11 is a graph showing the effect of All treatment on the day of exposure on to monocyte number after irradiation.
Figure 12 is a graph showing the cffect of All treatment two days following exposure on monocyte number after irradiation.
Figure 13 is a graph showing the effect of All treatment two days prior to exposure on neutrophil number after irradiation.
Figure 14 is a graph showing the effect of All treatment on the day of exposure on neutrophil number after irradiation.
Figure 15 is a graph showing the effect of All treatment two days following exposure on neutrophil number after irradiation.
Figure 16 is a graph showing the effect of All treatment two days prior to exposure on lymphocyte number after irradiation.
Figure 17 is a graph showing the effect of All treatment on the day of exposure on lymphocyte number after irradiation.
Figure 18 is a graph showing the effect of All treatment two days following exposure on lymphocyte number after irradiation. =

76909-151E) Figure 19 is a graph showing is a graph showing the effect of AU analogues and fragments treatment on white blood cell number after irradiation.
Figure 20 is a graph showing is a graph showing the effect of All analogues and fragments treatment on platelet number after irradiation.
Figure 21 is a graph showing the effect of All on mouse survival receiving bone marrow transplantation after lethal irradiation. =
Figure 22 is a graph showing the effect of All treatment on mouse survival receiving bone marrow transplantation after lethal irradiation.
Figure 23 is a graph showing the effect of All on white blood cell number in the blood of mice receiving bone marrow transplantation after lethal irradiation.
Figure 24 is a graph showing the effect of All on white blood cell number in the blood of mice receiving bone marrow transplantation after lethal irradiation..

Detailed Description of the Preferred Embodiments The present invention fulfills the needs for improved therapeutic methods to mitigate radiation induced tissue damage, to improve the effectiveness of radiation therapy, to support bone marrow transplantation, and to promote megalcaryocyte .20 production and mobilization and platelet production.
As defined herein the phrase "mitigation of tissue damage" refers not only to reduction of damage, but also encompasses recovery of tissue from damage. As used herein "tissue" refers to any tissue type, and also includes hematopoietic stem and progenitor cells, white blood cells and platelets.

As defined herein the term "megakaryocyte mobilization" refers to the movement of a megakaryocyte precursor cell from the bone marrow into the periphery.
As defined herein, the phrase "improved platelet production" or "improved megakaryocyte production," means that the number of platelets or megakaryocytes is signifiCantly elevated above the normal range of platelets or megakaryocytes in the particular mammal involved. The elevation of platelet or megakaryocyte counts may occur in a time-dependent manner, and may be cyclical, increasing and then constant or decreasing, or constant, etc.
tO Unless otherwise indicated, the term "active agents" as used herein refers to the group of compounds comprising angiotensinogen, angiotensin. I (A1), Al analogues, Al fragments and analogues thereof, angiotensin II (All), All analogues, All fragments or analogues thereof and All AT, type 2 receptor agonists.
Within this application, unless otherwise stated, the techniques utilized may be found in any of several well-known references such as: Molecular Cloning: A
Laboratory Manual (Sambrook, et al., 1989, Cold Spring Harbor Laboratory Press), Gene Expression Technology (Methods in Errzymology, Vol. 185, edited by D.
Goeddel, 1991, Academic Press, San Diego, CA), "Guide to Protein Purification"
in Methods in Enzymology (M.P. Deutshcer, ed., (1990) Academic Press, Inc.); PCR
Protocols: A Guide to Methods and Applications (Innis, et al. 1990. Academic Press, San Diego, CA), Culture of Animal Cells: A Manual of Basic Technique, ri Ed.
(R.I.
Freshney. 1987. Liss, Inc. New York, NY), Gene Transfer and Expression Protocols, pp. 109-128, ed. E.J. Murray, The Humana Press Inc., Clifton, NJ), and the Ambion 1998 Catalog (Ambion, Austin, TX).

U.S. Patent No. 5,015429 to DiZerega describes a method for increasing the rate of healing of wound tissue, comprising the application to such tissue of angiotensin II
(AII) in an amount which is sufficient for said increase. The application of All to wound tissue significantly increases the rate of wound healing, leading to a more rapid re-enithelialization and tissue repair. The term AL! refers .to an octapeptide present in humans and other species having the sequence Asp-Arg-Val-Tyr-Ile-His-= Pro-Phe [SEQ ID NO:1]., The biological formation of angiotensin is initiated by the action of renin on the plasma substrate angiotensinogen. The substance so formed is a to decapeptide called angiotensin I (Al) which is converted to All by the converting enzyme angiotensinase which removes the C-terminal His-Leu residues from Al (Asp-Arg-Val-Tyr-Ile-His-Pro-Phe-His-Leu [SEQ ID NO:37]). All is a known pressor agent and is commercially available. The use of All analogues and fragments, AT2 agonists, as well as AIII and AIII analogues and fragments in wound healing has also. been described. (U.S. Patent No. 5,629,292; U.S. Patent No. 5,716,935;
WO
96.139164).
Studies have shown that All increases mitogenesis and chemotaxis in cultured cells that are involved in wound repair, and also increases their release of growth factors and extracellular matrices (diZerega, U.S. Patent No. 5,015,629; Dzau et. al., = J. Alol. Cell. CardioL 21:S7 (Supp 111) 1989; Berk et. al., Hypertension 13:305-14 (1989); Kawahara, et al., BBRC 150:52-9 (1988); Nafiilan, et al., .1. Clin.
Invest.
83:1419-23 (1989); Taubman et at.. J. BioL Chem 264:526-530 (1989); Nakahara, et al., BBRC 184:811-8 (1992); Stouffer and Owens, Circ. Res. 70:820 (1992);
Wolf, et al., Am. J. Pathol. 140:95-107 (1992); Bell and Madri, Am. J. PathoL 137:7-12 (1990). In addition, MI was shown to be angiogenic in rabbit corneal eye and chick chorioallantoic membrane models (Fernandez, et al., J. Lab. Clin. Med. 105:141 (1985); LeNoble, et al., Eur. J. Pharmacol. 195:305-6 (1991). Therefore, All may accelerate wound repair through increased neovascularization, growth factor release, reepithelialization and/or production of extracellular matrix.
AII has also been implicated in both cell growth and differentiation (Meffert et al., MoL and Cellul. ,Endocrin. 122:59 (1996)). Two main classes of All receptors, ATI and AT, have been identified (Meffert, 1996). The growth-promoting effects of All have been attributed to mediation by the ATI receptor, while some evidence suggests that the AT2 receptor may bc involved in mediation of the cell differentiation effects of All (Bedecs et al., Biochern. J. 325:449 (1997)).
The effects of All receptor and All receptor antagonists have been examined in two experimental models of vascular injury and repair which suggest that both All receptor subtypes (AT! and AT2) play a role in wound healing (Janiak et at., Hypertension 20:737-45 (1992); Prescott, etal., Ain. I PathoL 139:1291-1296 (1991);
Kauffman. et al., Life Sci. 49:223-228 (1991); Viswanathan, et al., Peptides 13:783-786 (1992); Kimura, et at., BBRC 187:1083-1090(1992).
Many studies have focused upon A11(1-7) (All residues 1-7) or other fragments of All to evaluate their activity. AI1(177) elicits some, but not the full range of effects elicited by All. Pfeilschifter, et al., Eur. J. Pharniacol. 225:57-62 (1992);
Jaiswal, et al., Hypertension 19(Supp. II):II-49-II-55 (1992); Edwards and Stack, J.
Pharrnacol. Exper. Ther. 266:506-510 (1993); Jaiswal, et al., J. Pharmacol.
Exper.
Ther. 265:664-673 (1991); Jaiswal, et at., Hypertension 17:1115-1120 (1991);
Portsi, eta., Br. J. Pharmacol. 111:652-654 (1994).

While a single pilot study has suggested that All-induced hypertension might be effective in combination with radiation therapy in the treatment of lung cancer patients (Kato et al., Radiation Medicine 11:86-90 (1993)), many studies have demonstrated that antagonists of angiotensin converting enzyme (ACE), which mediate the production of All, are effective in reducing radiation nephropathy, bone mart;ow transplantation nephropathy, and acute radiation injury (Moulder et nl., Int. I.
Radiation Onc. Biol. Phys. 27:93-99 (1993); Moulder et al., Bone Marrow TranspL
19:729-735 (1997); Moulder et al., Radiation Res. 146:106-110 (1996); Cohen et al., J. Lab. Clin. Med. 129:536-547 (1997); Moulder et al., Radiation Res. 136:404-(1993); Yoon et al., Int. J. Radial. Oncol. Biol. Phys. 30:873-878 (1994);
Ward:et al., -Radiation Res. 135:81-87 (1993); Cohen et at, Lab. Invest. 75:349-360 (1996);
Cohen et al., J. Lab. Clin. Med. 124:371-380 (1994); Gerarci et al., Radiation Res.
143:58-68 (1995)). The effect of the ACE inhibitors has been demonstrated, in at least one case, to be directly caused by the reduction of activation of the ATI receptor by All (Moulder et al.. Radiation Res. 146:106-110 (1996)). These results have led to the suggestion that, in the case of radiation nephropathy, the most effective treatment is the use of ACE inhibitors (Moulder et al., Bone Marrow Transplantation 19:729-(1997)).
Furthermore, it has recently been demonstrated that angiotensinogen, angiotensin I (Al). Al analogues, Al fragments and analogues thereof, All, All analogues, All fragments or analogues thereof or AU AT-, type 2 receptor agonists are potent stimulators of hematopoietic stem cell proliferation (j.s. Patent No.
6,335,195).
Therefore, it would be expected that the use of these compounds might cause long-term hematopoietic damage if used in conjunction with radiation therapy (Masse et al., 1998; Watanabe et al., 1996).
Based on all of the above, it would be unexpected that the use of angiotensinogen, angiotensin I (Al), Al analogues, Al fragments and analogues thereof, All, All analogues, All fragments or analogues thereof or All AT, type 2 receptor agonists would be effective in reducing radiation-induced human tissue damage or in treating patients in need of radiation therapy.
None of these studies teach or suggest the use of angiotensinogen, angiotensin I (Al), Al analogues, Al fragments and analogues thereof, angiotensin II
(All), All analogues, All fragments or analogues thereof or All AT2 type. 2 receptor agonists to stimulate the production and mobilization of megakaryocytes, or to stimulate the production of platelets.
A peptide agonist selective for the AT2 receptor (MI has 100 times higher affinity for AT2 than ATI) is p-aminophenylalanine6-AII ["(p-N1-12-Phe)6-AII)"], Asp-Arg-Val-Tyr-Ile-Xaa-Pro-Phe [SEQ ID NO.36] wherein Xaa is p-NH2-Phe (Speth and Kim, BBRC 169:997-1006 (1990). This peptide gave binding characteristics comparable to AT2 antagonists in the experimental models tested (Catalioto, et al., Eur. I. Phartnacol. 256:93-97 (1994); Bryson, et al., Eur.

Pharmacol. 225119-127 (1992).
The active Al, Al analogues, Al fragments and analogues thereof, All analogues, fragments of All and analogues thereof of particular interest in accordance with the present invention are characterized as comprising a sequence consisting of at least three contiguous amino acids-of groups RI-R8 in the sequence of general formula I

RI -R2-R3-R4-R5-R6_R7-R8 in which RI and R2 together form a group of formula X-RA-RB-, wherein X is H or a one to three peptide group, RA is - suitably selected from Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc, RB is suitably selected from Arg, Lys, Ala, Om, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile.
Gly, Pro, Aib, Acpc, Lys, and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(P03)2, Thr, Ser, homoSer, Ala, and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-1\1112-Phe;
R7 is Pro or Ala; and R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
Compounds falling within the category of AT2 agonists useful in the practice of the invention include the All analogues set forth above subject to the restriction that R6 is p-NH2-Phe. In addition to peptide agents, various nonpeptidic agents (e.g., peptidomimetics) having the requisite AT2 agonist activity are further contemplated for use in accordance with the present invention.

Particularly preferred combinations for RA and RB are Asp-Arg, Asp-Lys, Glu-Arg and Glu-Lys. Particularly preferred embodiments of this class include the following: All, AHI or AII(2-8), Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:21;
A1I(3-8), also known as desl-AIII or MV, Val-Tyr-Ile-His-Pro-Phe [SEQ ID
NO:31;
AII(1-7), Asp-Arg-Val-Tyr-Ile-His-Pro [SEQ ID NO:4]; Arg-Val-Tyr-Ile-His-Pro [SEQ ID NO:5]; MI(3-7), Val-Tyr-Ile-His-Pro [SEQ ID NO:6]; AII(5-8), Ile-His-Pro-Phe [SEQ ID NO:7]; AII(I-6), Asp-Arg-Val-Tyr-Ile-His [SEQ ID NO:8];
A1I(1-5), Asp-Arg-Val-Tyr-Ile [SEQ ID NO:9]; AH(1-4), Asp-Arg-Val-Tyr [SEQ ID
NO:10]; and MI(1-3), Asp-Arg-Val [SEQ ID NO:11]. Other preferred embodiments to include: Arg-norLeu-Tyr-Ile-His-Pro-Phe [SEQ ID NO:12] and Arg-Val-Tyr-norLeu--His-Pro-Phe [SEQ ID NO:13]. Still another preferred embodiment encompassed within the scope of the invention is a peptide having the sequence Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe [SEQ ID NO:31]. AII(6-8), His-Pro-Phe [SEQ ID NO:14] and AII(4-8), Tyr-Ile-His-Pro-Phe [SEQ ID NO:15] were also tested and found not to be effective.
A class of particularly preferred compounds in accordance with the present invention consists of those with the following general structure:
RI -Arg-R2-R3-R4-His-Pro-R5 wherein RI is selected from the group consisting of H and Asp;
R2 is selected from the group consisting of Val and Pro;
R3 is selected from the group consisting of Tyr and Tyr(P03)2;
R4 is selected from the group consisting of Ala, Ile, Leu, and norLeu; and R5 is Phe, Ile, or is absent.

Particularly preferred embodiment of this class are selected from the group consisting of SEQ ID NO:1, SEQ ID NO:4, SEQ ID NO:18, SEQ ID NO:26, SEQ liD
NO:31, SEQ ID NO:32, SEQ ID NO:34, and SEQ ID NO:38 Another class of compounds of particular interest in accordance with the present invention are those of the general formula II

in which R2 is selected from the group consisting of H, Arg, Lys, Ma, Om, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Cily, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(P03)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of lie, Ala. Leu, norLeu, Val and Gly;
R6 is His, Aru or 6-NH2-Phe;
R7 is Pro or Ala; and R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
A particularly preferred subclass of the compounds of general formula II has the formula R2-R3-Tyr-R5-His-Pro-Phe [SEQ ID NO:161 wherein R2, R3 and R5 are as previously defined. Particularly preferred is angiotensin III of the formula Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:2].
Other preferred compounds include peptides having the structures Arg-Val-Tyr-Gly-His-Pro-Phe [SEQ ID NO:17] and Arg-Val-Tyr-Ala-His-Pro-Phe [SEQ ID NO:18]. The fragment A11(4-8) was ineffective in repeated tests; this is believed to be due to the exposed tyrosine on the N-terminus.

In the above formulas, the standard three-letter abbreviations for amino acid residues are employed.. In the absence of an indication to the contrary, the L-form of the amino acid is intended. Other residues are abbreviated as follows:

Abbreviation for Amino Acids Me2Gly N,N-dimethylglycyl Bet 1-carboxy-N,N,N-trimethylmethanaminium hydroxide inner salt (betaine) Suc Succinyl : Phe(Br) p-bromo-L-phenylalanyl azaTyr aza-a ' -homo-L-tyrosyl Acpc 1-aminocyclopentane carboglic acid Alb 2-aminoisobutyric acid Sar N-methylglycyl (sarcosine) =

It has been suggested that All and its analogues adopt either a gamma or a beta turn (Regoli, et al., Pharmacological Reviews 26:69 (1974). In general, it is believed that neutral side chains in position R3, R5 and R7 may be involved in maintaining the appropriate distance between active groups in positions R4, R6 and R8 primarily responsible for binding to receptors and/or intrinsic activity.
Hydrophobic side chains in positions R3, R5 and R8 may also play an important role in the whole conformation of the peptide and/or contribute to the formation of a hypothetical hydrophobic pocket.
Appropriate side chains on the amino acid in position R2 may contribute to affinity of the compounds for target receptors and/or play an important role in the conformation of the peptide. For. this reason, Arg and Lys are particularly preferred as R2.
For purposes of the present invention, it is believed that R3 may be involved in the formation of linear or nonlinear hydrogen bonds with R5 (in the gamma turn to model) or R6 (in the beta turn model). R3 would also participate in the first tarn in a beta antiparallel structure (which has also been proposed as a possible structure). In contrast to other positions in general formula I. it appears that beta and gamma branching are equally effective in this position. Moreover, a single hydrogen bond may be sufficient to maintain a relatively stable conformation. Accordingly, R3 may suitably be selected from Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr. In another preferred embodiment, R3 is Lys.
With respect to R4, conformational analyses have suggested that the side chain in this position (as well as in R3 and R5) contribute to a hydrophobic cluster believed to be essential for occupation and stimulation of receptors. Thus, R4 is preferably selected from Tyr, Thr, Tyr (P03)2, homoSer, Ser and azaTyr. In this position, Tyr is particularly preferred as it may form a hydrogen bond with the receptor site capable of accepting a hydrogen from the phenolic hydroxyl (Regoli, et al. (1974), supra). In a further preferred embodiment, R4 is Ala.

In position R5, an amino acid with a 13 aliphatic or alicyclic chain is particularly desirable. Therefore, while Gly is suitable in position Rs, it is preferred that the amino acid in this position be selected from Ile, Ala, Leu, norLeu, Gly and Val.

In the Al, Al analogues, Al fragments and analogues thereof, All, All analogues,; fragments and analogues of fragments of particular interest in accordance with the present invention, R6 is His, Arg or 6-NH2-Phe. The unique properties of the imidazole ring of histidine (e.g., ionization at physiological pH, ability to act as proton donor or acceptor, aromatic character) are believed to contribute to its particular utility as R6. For example, conformational models suggest that His may participate in hydrogen bond formation (in the beta model) or in the second turn of the antiparallel structure by influencing the orientation of R7. Similarly, it is presently considered that R7 should be Pro in order to provide the most desirable orientation of R8. In position R8, both a hydrophobic ring and an anionic carboxyl terminal appear to be particularly useful in binding of the analogues of interest to receptors; therefore, Tyr and especially Phe are preferred for purposes of the present invention.

Analogues of particular interest include the following:

Angiotensin II Analogues All Amino Acid Sequence Sequence Analogue Identifier Name Analogue 1 Asp-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 19 Analogue 2 Asn-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 20 Analogue 3 Ala-Pro-Gly-Asp-Arg-Ile-Tyr-Val-His-Pro-Phe SEQ ID NO: 21 Analogue 4 Glu-Arg-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 22 Analogue 5 Asp-Lys-Val-Tyr-Ile-His-No-Phe SEQ ID NO: 23 Analogue 6 Asp-Arg-Ala-Tyr-Ile-His-Pro-Phe = SEQ ID NO: 24 Analogue 7 Asp-Arg-Val-Thr-Ile-His-Pro-Phe SEQ ID NO: 25 'Analogue 8 _ Asp-Arg-Val-Tyr-Leu-His-Pro-Phe SEQ ID NO: 26 ¨
Analogue 9 Asp-Arg-Val-Tyr-Ile-Arg-Pro-Phe SEQ ID NO: 27 , Analogue 10 Asp-Arg-Val-Tyr-Ile-His-Ala-Phe SEQ ID NO: 28 Analogue 11 Asp-Arg-Val-Tyr-Ile-His-Pro-Tyr SEQ ID NO: 29 Analogue 12 Pro-Arg-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 30 Analogue 13 _ Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe SEQ ID NO: 31 Analogue 14 Asp-Arg-Val-Ty003)2-11e-His-Pro-Phe SEQ ID NO: 32 Analogue 15 Asp-Arg-norLeu-Tyr-Ile-His-Pro-Phe SEQ ID NO: 33 , Analogue 16 _ Asp-Arg-Val-Tyr-norLeu-His-Pro-Phe SEQ ID NO: 34 Analogue 17 Asp-Arg-Val-homoSer-Tyr-Ile-His-Pro-PheSEQID NO: 35 The polypeptides of the instant invention may be synthesized by methods such as those set forth in J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, 2nd ed., Pierce Chemical Co., Rockford, 111. (1984) and J. Meienhofer, Hormonal 3 Proteins and Peptides. Vol. 2, Academic Press, New York, (1973) for solid phase synthesis and E. Schroder and K. Lubke, The Peptides, Vol. 1, Academic Press, New York, (1965) for solution synthesis.

In general, these methods involve the sequential addition of protected amino to acids to a growing peptide chain (U.S. Patent No. 5,693,616).
Normally, either the amino or carboxyl group of the first amino acid and any reactive side chain group are protected. This protected amino acid is then either attached to an inert solid support, or utilized in solution, and the next amino acid in the sequence,- also suitably protected, is added under conditions 15 amenable to formation of the amide linkage. After all the desired amino acids have been linked in the proper sequence, protecting groups and any solid support are removed to afford the crude poTypeptide. The polypeptide is desalted and purified, preferably chromatographically, to yield the final product.

=

In one aspect, the present invention provides methods and kits for the mitigation of tissue damage due to radiation exposure comprising the administration of angiotensinogen, angiotensin I (Al), Al analogues, Al fragments and analogues thereof, angiotensin H (MI), All analogues, All fragments or analogues thereof or All AT2 type 2 receptor agonists (the "active agents").
In another aspect, the present invention provides improved methods and kits for treating a patient .afflicted with a neoplastic disease state that is being treated with ionizing or nonionizing radiation, the improvement comprising conjunctive therapy whereby an effective radioprotective amount of the active agents is provided.
to In another aspcct, the present invention provides improved methods and kits for treating a patient in need of radiation therapy, the improvement comprising the administration of the active agents in conjunction with the radiation therapy.
The invention is appropriate for use with any type of ionizing radiation exposure such as therapeutic or accidental X-ray, gamma ray, or beta particle exposure. Examples of ionizing radiation exposure suitable for treatment with the methods and kits of the present invention include, but are not limited to, clinical radiation therapy, medical diagnostics using radioactive tracers, exposure to naturally occurring ionizing radiation sources such as uranium and radon, wartime exposure, and accidental exposures including occupational exposure at nuclear power facilities, and medical and research institutions. Examples of nonionizing radiation exposure suitable for treatment with the methods and kits of the present invention include, but are not limited to, ultraviolet light, X-rays, microwaves, radio-frequency waves, and electromagnetic radiation.

Virtually any tissue susceptible to radiation-induced tissue damage can gain protection by use of the active agents of the invention. For example, breast tissue is an excellent candidate for receiving the benefit of the subject invention.
Radiation-induced tissue damage can be a fatal side effect of over-exposure to radiation therapy.
Typically, the fibrotic reaction common in normal breast tissue surrounding the cancerous tumor being treated with radiation therapy undermines the cosmetic advantages of radiation therapy over surgical treatment. This disadvantage will lead many patients to elect a less effective or more dangerous treatment after radiation therapy.The present invention is also particularly suitable for those patients in need of repeated or high doses of radiation therapy. For some cancer patients, hematopoietic toxicity frequently limits the opportunity for radiation dose escalation (Watanabe et al., British J. Haentatol. 94:619-627 (1996)). Repeated or high dose cycles of radiation therapy may be responsible for severe stem cell depletion leading to important long-term hematopoietic sequelea and marrow exhaustion. The methods of the present invention provide for improved mortality and blood cell count when used in conjunction with radiation therapy.
Skin exposure is particularly common in accidental radiation exposure. It is an excellent candidate for the inventive therapy, especially as the compounds of the .20 invention can be administered topically. Other tissues that are susceptible to radiation-induced damage following accidental or therapeutic ionizing or nonionizing radiation exposure include, but are not limited to: liver, lung, gastrointestinal tract, kidneys, testes, salivary gland, mucosa and brain.

In another aspect, the present invention provides improved methods and kits for supporting bone marrow transplantation comprising the administration of the active agents to a patient in need thereof. These compounds may be administered in combination with auxiliary agents including, but not limited to interleukin (IL)-3, IL-1, IL-4,11-5, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), anticancer agents, antiviral agents, and antibiotics.
In a further aspect, the present invention provides kits for mitigating radiation induced tissue damage and improving the efficacy of radiation therapy, wherein the to kits comprise an effective amount of the active agents of the invention for mitigating radiation induced tissue damage or improving the efficacy of radiation therapy, and instructions for using the amount effective of active agent as a therapeutic.
In a preferred embodiment, the kit further comprises a pharmaceutically acceptable carrier, such as those adjuvants described above. In another preferred embodiment, the kit further comprises a means for delivery of the active agent to a patient. Such devices include, but are not limited to syringes, matrical or micellar solutions.
bandages, wound dressings, aerosol sprays, lipid- foams, transderrnal patches, topical administrative agents, polyethylene glycol polymers, carboxymethyl cellulose preparations, crystalloid preparations. (e.g., saline, Ringer's lactate solution, phosphate-buffered saline, etc.), viscoelastics, polyethylene glycols. and polypropylene glycols. The means for delivery may either contain the effective amount of angiotensinogen, Al, Al analogues, Al fragments and analogues thereof, All, All analogues, All fragments or analogues thereof or All AT7 type 2 receptor agonists, or may be separate from the compounds, which are then applied to the means for delivery at the time of use.
The methods and kits of the present invention, by mitigating radiation induced tissue damage and improving the efficacy of radiation therapy and bone marrow transplantation, significantly enhance the utility of presently available treatments both for radiation-induced tissue damage and for clinical radiation therapy.
In a further aspect of the present invention, a method of increasing megakaryocyte production and mobilization and platelet production by .exposure to the active agents of the inventions is disclosed. In one embodiment, megakaryocytes are isolated from bone marrow as described in U.S.. Patent No. 5,178,856).

Briefly, marrow is flushed from a subject's femur with Iscove's modification of Dulbecco's medium (IMDM) supplemented with Nutridom1SP (Boehringer Mannheim, Indianapolis, hid.), a serum-free medium supplement. For culture studies, a single cell suspension is made by repetitive expulsion through progressively smaller needles. For flow cytornetry controls, a monocellular suspension is made by gentle filtration through a 100 micron nylon mesh. Preferably, adherent cells are -removed to enrich the numbers of megakaryocytes or their progenitor cells. Up to 2 x 106 cells/ml are placed in growth medium at 37 C in a humidified atmosphere in the presence of, preferably, between about 0.1 ng/ml and about 10 mg/m1 of the active agents. The cells are expanded for a period of between 2 and 21 days and cellular proliferation is assessed at various time points during this time period. Subsequent medium changes are performed as needed. In a preferred embodiment, megakaryocyte production and mobilization and platelet production are assessed by the extent of megakaryocyte ploidization by flow *Trade mark 23 '6909-1519 cytometry as described in U.S. Patent No. 5,155,211).
Briefly, the appearance of granules and the extensive surface-connected open canalicular membrane system as well as a substantial decrease in the nucleus:cytoplasm volume distribution, indicates that the megalcaryocyte population has completed the process of polyploidization but has not yet generated, a major portion of their final complement of platelet-specific cytoplasmic components.
In another embodiment, subjects are irradiated as above and active agent is =
injected subcutaneously before, at the time of, and after irradiation. Blood samples =
are taken at various times after administration of the active agent to monitor the number of white blood cells, megalcaryocytes and platelets. In a preferred embodiment, subjects are treated with total body irradiation and active agent is administered subcutaneously (10 jig/kg/day or 100 jig/kg/day) at various times before = and after irradiation. The number of white blood cells, megalcaryocytes and platelets is preferably determined by counting with a hemacytometer followed by differential morphologic analysis.
= In another embodiment of this aspect of the invention; hematopoietic = precursor cells are isolated from bone marrow, peripheral blood or umbilical cord blood and cultured under appropriate growth conditions, in the presence of the active agents. Megalcaryocyte production is assessed at various time points during culture 2Q by differential morphologic analysis.
In a preferred embodiment, hematopoietic precursor cells are isolated from bone marrow aspirates from the posterior iliac crest (Caplan and Haynesworth, U.S.
Patent No. 5.486,359). CD34+ hematopoietic precursor cells- are isolated from the aspirate by attaching a biotinylated monoclonal antibody specific for CD34 (available from Becton Dickinson, Sunnyvale, CA, USA) to a streptavidin affinity column (Ceprate* SC; CellPio, Bothell, WA, USA) and passing the aspirate through the column, folldwed by appropriate column washing and stripping, according to standard techniques in the art. The isolated cells are suspended in culture medium and incubated in the presence of, preferably, between about 0.1 ng/ml and about 10 mg/m1 of th'e active agents of the invention. The cells are expanded for a period of between 8 and 21 days and me_gakaryocyte production is assessed via phase microscopy to detect increased size and polyploidization at various points during this time period.
In a further embodiment of the present invention, a method of increasing megakaryocyte production and mobilization and platelet production by exposure to the active agents is disclosed, either in the presence or absence of other growth factors and cytoldnes. Examples of such growth factors and cytokines include, but are not limited to thrombopoietin, lympholdnes, interleukins - 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, granulocyte colony-stimulating factor, granulocyte/macrophage colony stimulating factor, macrophage colony-stimulating factor, tumor necrosis factor, epidermal growth factor, fibroblast growth factor, platelet derived growth factor, transforming growth factor beta, and stem cell factor.
In a further preferred embodiment, megakaryocytes and/or platelets that have been cultured in the presence of the active agents are used for autologous transplantation, to reconstitute a depleted hematopoietic system. Prior to transplantation, the cells are rinsed to remove all traces of culture fluid, resuspended in an appropriate medium and then pelleted and rinsed several times. After the final rinse, the cells are resuspended at between 0.7 x 106 and 50 x 106 cells per ml in an appropriate medium and reinfused into a subject through intravenous infusions.
*Trade mark 25 ,6909-151D

Following transplantation, subject peripheral blood samples are evaluated for increased megakaryocyte ploidy and platelet number by standard flow cytometry and cell sorting techniques. (Talmadge, et al., supra).
In another aspect of the present invention the active agents are used to increase in vivo megakaryocyte production and mobilization and platelet production. For use in increasing megakaryocyte production and mobilization and platelet production, the active agents may be administered by any suitable route, including orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.
The term parenteral as used herein includes, subcutaneous, intravenous, intramuscular, intrasternal, intratendinous, intraspinal, intraeranial, intrathoracic, infusion techniques or intraperitoneally. The active agents of all aspects of the present invention may be administered by any suitable route, including orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. The term parenteral as used herein includes, subcutaneous, intravenous, intraarterial, intramuscular, intrastemal, intratendinous, intraspinal, intracranial, intrathoracic, infusion techniques or intraperitoneally.

=
The active agents of the invention may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions). The compounds of the invention may be applied in a variety of solutions.
Suitable solutions for use in accordance with the invention are sterile, dissolve sufficient amounts of the peptide, and are not harmful for the proposed application. In this regard, the compounds of the present invention are very stable but are hydrolyzed by strong acids and bases. The compounds of the present invention are soluble in organic solvents and in aqueous solutions at pH 5-8.
The active agents may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabipzers, wetting agents, emulsifiers, buffers etc. For administration, the active agents are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration. The compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, acacia, gelatin, sodium alginate, polyvinylpyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration.
Alternatively, the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, carboxymethyl cellulose colloidal solutions, ethanol, corn oil, peanut oil, cottonseed oil, sesame oil, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well known in the pharmaceutical art. The carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose.

The dosage regimen for mitigating radiation-induced tissue damage and improving the efficacy of radiation therapy with the active agents is based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely by a physician using standard methods. Dosage levels of the order of between 0.1 ng/kg and 10 mg/kg body weight of the active agents are useful for all methods of use disclosed herein.
The treatment regime will also vary depending, on the disease being treated, to based on a variety of factors, including the type of injury, thc agc, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. For example, the active agents are administered to an oncology patient for up to 30 days prior to a course of radiation therapy and for up to 60 days post-radiation exposure. The therapy is administered for 1 to 6 times per day at dosages as described above.
In all of these embodiments, the compounds of the invention can be administered either prior to, simultaneously with; or subsequent to radiation exposure.
In a preferred embodiment, the active agent is administered subcutaneously.
A suitable subcutaneous dose of active ingredient of active agent is preferably between about 0.1 ng/kg and about 10 mg/kg administered twice daily for a time sufficient to mitigate radiation-induced tissue damage, to provide a radioprotective effect for a radiation therapy patient afflicted with a neoplastic disease, to effectively treat a patient in need of radiation therapy, to support bone marrow transplantation and to promote megakaryocyte production and mobilization and platelet production.

In a more preferred embodiment, the concentration of active agent is between about 100 ng,/kg body weight and about 10.0 mg/kg body weight. In a most preferred embodiment, the concentration of active agent is between about 10 jig/kg body weight and about 10.0 mg/kg body weight. This dosage regimen maximizes the therapeutic benefits of the subject invention while minimizing the amount of agonist or peptide needed. Such an application minimizes costs as well as possible deleterious side effects.
For subcutaneous administration, the active ingredient may comprise from 0.0001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it to may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
In another preferred embodiment of the present invention, the active agent is administered topically. Suitable topical doses and active ingredient concentration in the formulation are as described for subcutaneous administration.
In a preferred embodiment of all of the aspects of the invention, the active agent is selected from the group consisting of SEQ ID NO. 1, SEQ ID NO:2, SEQ
ID
NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO: 32, SEQ ID NO:33, SEQ ID NO: 34; SEQ ID NO:35, SEQ ID NO:36; and SEQ ID NO:37.

In a further preferred embodiment of the above aspects of the invention, administration of the active agent is localized to the area affected by the tissue-damaging radiation.
In another aspect of the present invention, an improved cell culture medium is provided for megakaryocyte and platelet production, wherein the improvement comprises addition to the cell culture medium of an effective amount of between about 0.1 ng and 10 mg/ml of the active agents of the invention. Any cell culture media that can support megakaryocyte and platelet production can be used with the present invention. Such cell culture media include, but are not limited to Basal Media Eagle, Dulbecco's Modified Eagle Medium, 'wove's Modified Dulbecco's Medium, McCoy's Medium, Minimum Essential Medium, F-10 Nutrient Mixtures, Opti-MEM Reduced-Serum Medium, RPM! Medium, and Macrophage-SFM Medium or combinations thereof.
The improved cell culture medium can be supplied in either a concentrated (ie:
10X) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophilizate. The cell culture may be either chemically defined, or may contain a serum supplement. Culture media is commercially available from many sources, such as GIBCO BRL (Gaithersburg, MD) and Sigma (St. Louis, MO) In a further aspect, the present invention provides kits for megakaryocyte and platelet production, wherein the kits comprise an amount effective for megakaryocyte and platelet production of the active agents of the invention, and instructions for its use as a cell culture media supplement.
In a preferred embodiment, the kits further comprise cell culture growth medium. Any cell culture media that can support megakaryocyte and platelet production can be used with the present invention. Examples of such cell culture media are described above. The cell culture medium can be supplied in either a concentrated (ie: 10X) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophilizate. The cell culture may be either chemically defined, or may contain a serum supplement.
In a further preferred embodiment, the kit further comprises a sterile container, which can comprise either a sealed container, such as a cell culture flask, a roller bottle, or a centrifuge tube, or a non-sealed container, such as a cell culture plate or microtiter plate (Nunc; Naperville, IL).
to In another preferred embodiment, the kit further comprises an antibiotic supplement for inclusion in the reconstituted cell growth medium. Examples of appropriate antibiotic supplements include, but are not limited to actimonycin D, Fungizone , kanamycin, neomycin, nystatin, penicillin, streptomycin, or combinations thereof (GIBCO).The present invention may be better understood with reference to the accompanying example that is intended for purposes of illustration only and should not be construed to limit the scope of the invention, as defined by thc claims appended hereto.
=
Example I Effect of All on rat mortality and white blood cell recovery after irradiation Female C57B1/6 mice (Jackson Labs, Bar Harbor, Maine) were irradiated with 600 cGy total body irradiation. Subcutaneous injection with either All (10 ug/kg/day or 100 ug/kg/day) or saline (placebo) was initiated two days before (-day 2), on the day of (day 0) or 2 days after (+ day 2) irradiation and continued until the animals succumbed to the irradiation or were necropsicd. At various times after irradiation, the mice were anaesthetized with Metofane* (Pittman-Moore Animal Health, NZ) and bled via the retro-orbital sinus. Red blood cells were lysed with 0.3% acetic acid and. the number of white blood cells was determined by counting with a hemacytometer. The data in Figures 1-3 show that administration of All starting at two days prior to irradiation did not protect against mortality resulting from irradiation (Figure 1), but that All administration on the day of irradiation (Figure 2) or two days after irradiation (Figure 3) substantially increased survivaL
Furthermore, MI administration at all time periods tested increased the number of circulating white blood cells (Figures 4-6). Further experiments demonstrated that All administration increased the number of megakaryocytes (Figures 7-9), monocytes (Figures 10-12), neutrophils (Figures 13-15), and lymphocytes (Figures 16-18). These. data demonstrate that in vivo administration of All can improve hematopoietic recovery after irradiation.

Example 2. Effect of All and All Analogs/Fragments on WBC and platelet numbers After Irradiation The animals were irradiated and treated as in Example 1, however, treatment started on day 0 only with one subcutaneous injection of either 10. g/lcg or 100 fig/kg daily until the study was terminated. Analogues and fragments of All (see Table 3) were assessed for their effect on WBC recovery and platelet number after irradiation.

The data are shown in Figures 19 and 20. and show that the peptides increase the production of both of these blood ele?nents.

*Trade mark 32 Table 3: Designation for Analogues/Fragments Name Abbreviation Sequence SEQ ID NO:
GSD 28 I1e8-AII DRVY11HPI SEQ ID NO: 38 GSD 2413 Pro3-AII DRPYIHPF SEQ ID NO:31 GSD 22A A1a4-AIII RVYAHPF SEQ ID NO:18 AII(1-7) DRVYIHP SEQ ID NO:4 All DRVYIHPF SEQ ID NO. 1 Example 3. EffeCt of All on survival of mice receiving bone marrow transplantation after lethal irradiation Donor C57B1/6 mice (female, 6-8 weeks old) were irradiated with 600 cGy total body irradiation. Starting on the day of irradiation, the mice received either saline (0.1 ml) or 20 pg/m1 angiotensin 11 (0.1 ml, 100 g/kg) subcutaneously for fourteen days. At the end of this period, the bone marrow was harvested from the femur by flushing and the number of viable nucleated cells determined by counting under a light microscope on a hemacytometer in the presence of trypan blue.
These donor bone marrow cells were then injected intravenously into recipient mice (female C57B1/6, 6-8 weeks old) that had been lethally irradiated (900 cGy total body irradiation) at two. concentrations: 1 x 1,06 or I x 105 cells per mouse.
After injection, the recipient mice received either saline or 100 pg/kg All subcutaneously until death or termination. The study design in its entirety is as follows:
Donor Recipient Cell Number Saline Saline 1 x 106 Saline Saline 1 x 10) Saline All I X 106 Saline All lx 105 All Saline 1 x 106 All Saline 1 x 105 All All 1 x 106 All All 1 x 105 The survival of the mice and the number of circulating white blood cells were measured as a function of time post-bone marrow transplantation. The data are presented , in Figures 21-24, and demonstrate that All treatment increased both survival and white blood cell number in mice receiving bone marrow transplantation after irradiation. The greatest benefit was conferred by treatment of both the donor bone marrow cells and the recipient mice with All.
The methods and kits of the present invention, by mitigating radiation induced tissue damage and improving the efficacy of radiation therapy, significantly enhance to the utility of presently available treatments both for radiation-induced tissue damage and for clinical radiation therapy, as well as bone marrow transplantation by increasing the survival rate of patients and accelerating the reconstitution of the patient's hematopoietic system. Similarly, by providing a method for megakaryocyte and platelet production, the present invention will greatly augment clinical cancer treatments and bone marrow transplantation and other conditions that lead to decreased megakaryocyte production and mobilization and platelet production.
The method of the present invention also increases the potential utility of megakaryocytes as vehicles for gene therapy in hematopoietic disorders, by providing a more efficient means to rapidly expand transfected megakaryocytes.
It is to be understood that the invention is not to be limited to the exact details of operation, or to the exact compounds, compositions, methods, procedures or embodiments shown and described, as obvious modifications and equivalents will be apparent to one skilled in the art, and the invention is therefore to be limited only by the full scope of the appended claims.

SEQUENCE LISTING

<110> Rodgers, Kathleen diZerega, Gere <120> Radiation Therapy Methods <130> 97017K2B
<140> PCT/US99/05194 <141> 1999-03-08 <160> 38 <170> PatentIn Ver. 2.0 <210> 1 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:ATI

<400> 1 Asp Arg Val Tyr Ile His Pro Phe <210> 2 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:ATI (2-8) <400> 2 Arg Val Tyr Ile His Pro Phe <210> 3 <211> 6 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (3-8) <400> 3 Val Tyr Ile His Pro Phe <210> 4 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:ATI (1-7) <400> 4 Asp Arg Val Tyr Ile His Pro <210> 5 <211> 6 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (2-7) <400> 5 Arg Val Tyr Ile His Pro <210> 6 <211> 5 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (3-7) <400> 6 Val Tyr Ile His Pro <210> 7 <211> 4 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (5-8) <400> 7 Ile His Pro Phe <210> 8 <211> 6 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-6) <400> 8 Asp Arg Val Tyr Ile His <210> 9 <211> 5 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-5) <400> 9 Asp Arg Val Tyr Ile <210> 10 <211> 4 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-4) <400> 10 Asp Arg Val Tyr <210> 11 <211> 3 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (1-3) <400> 11 Asp Arg Val <210> 12 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <220>
<221> MOD RES
<222> (2) <223> Nle <400> 12 Arg Xaa Tyr Ile His Pro Phe <210> 13 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <220>
<221> MOD_RES
<222> (4) <223> Nle <400> 13 Arg Val Tyr Xaa His Pro Phe <210> 14 <211> 3 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (6-8) <400> 14 His Pro Phe <210> 15 <211> 5 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII (4-8) <400> 15 Tyr Ile His Pro Phe <210> 16 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue class <220>
<221> UNSURE
<222> (1) <223> Xaa at position 1 can be Arg, Lys, Ala, Orn, Ser, MeGly, D-Arg, or D-Lys <220>
<221> UNSURE
<222> (2) <223> Xaa at position 2 can be Val, Ala, lieu, Nle, Ile, Gly, Pro, Aib, Acp, or Tyr <220>
<221> UNSURE
<222> (4) <223> Xaa at position 4 can be Ile, Ala, lieu, Nle, Val, or Gly <400> 16 Xaa Xaa Tyr Xaa His Pro Phe <210> 17 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <400> 17 Arg Val Tyr Gly His Pro Phe <210> 18 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue <400> 18 Arg Val Tyr Ala His Pro Phe <210> 19 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 1 <400> 19 Asp Arg Val Tyr Val His Pro Phe <210> 20 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 2 <400> 20 Asn Arg Val Tyr Val His Pro Phe <210> 21 <211> 11 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 3 <400> 21 Ala Pro Gly Asp Arg Ile Tyr Val His Pro Phe <210> 22 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 4 <400> 22 Glu Arg Val Tyr Ile His Pro Phe <210> 23 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 5 <400> 23 Asp Lys Val Tyr Ile His Pro Phe <210> 24 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 6 <400> 24 Asp Arg Ala Tyr Ile His Pro Phe <210> 25 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 7 <400> 25 Asp Arg Val Thr Ile His Pro Phe <210> 26 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 8 <400> 26 Asp Arg Val Tyr Leu His Pro Phe <210> 27 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 9 <400> 27 Asp Arg Val Tyr Ile Arg Pro Phe <210> 28 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 10 <400> 28 Asp Arg Val Tyr Ile His Ala Phe <210> 29 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 11 <400> 29 Asp Arg Val Tyr Ile His Pro Tyr <210> 30 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 12 <400> 30 Pro Arg Val Tyr Ile His Pro Phe <210> 31 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 13 <400> 31 Asp Arg Pro Tyr Ile His Pro Phe <210> 32 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 14 <220>
<221> MOD_RES
<222> (4) <223> PHOSPHORYLATION

<400> 32 Asp Arg Val Tyr Ile His Pro Phe <210> 33 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 15 <220>
<221> MOD RES
<222> (3) <223> Nle <400> 33 Asp Arg Xaa Tyr Ile His Pro Phe <210> 34 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AII analogue 16 <220>
<221> MOD RES
<222> (5) <223> Nle <400> 34 Asp Arg Val Tyr Xaa His Pro Phe <210> 35 <211> 9 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:ATI analogue 17 <220>
<221> MOD_RES
<222> (4) <223> homo Ser <400> 35 Asp Arg Val Ser Tyr Ile His Pro Phe <210> 36 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:p-aminophenylalanine 6 All <220>
<221> MOD_RES
<222> (6) <223> p-aminophenylalanine <400> 36 Asp Arg Val Tyr Ile Xaa Pro Phe <210> 37 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:angiotensin I
<400> 37 Asp Arg Val Tyr Ile His Pro Phe His Leu <210> 38 <211> 8 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:1GD: I1e8-AII

<400> 38 Asp Arg Val Tyr Ile His Pro Ile

Claims (10)

1. A use of an effective amount of at least one active agent for increasing megakaryocyte production and mobilization, platelet production or a combination thereof, the active agent comprising a peptide with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO: 18, SEQ ID

NO: 31, and SEQ ID NO: 38.
2. A use of an effective amount of at least one active agent in the manufacture of a medicament for increasing megakaryocyte production and mobilization, platelet production or a combination thereof, the active agent comprising a peptide with an amino acid sequence selected from the group consisting of SEQ ID
NO: 1, SEQ ID NO: 4, SEQ ID NO: 18, SEQ ID NO: 31, and SEQ ID NO: 38.
3. The use of claim 1 wherein the active agent comprises a peptide with the amino acid sequence of SEQ ID NO: 4.
4. The use of claim 2 wherein the active agent comprises a peptide with the amino acid sequence of SEQ ID NO: 4.
5. The use of claim 1 wherein the active agent consists of a peptide with the amino acid sequence of SEQ ID NO: 4.
6. The use of claim 2 wherein the active agent consists of a peptide with the amino acid sequence of SEQ ID NO: 4.
7. A megakaryocyte production and mobilization kit, a platelet production kit or a combination thereof, comprising:
(a) an amount effective to support increasing megakaryocyte production and mobilization, platelet production or a combination thereof of at least one active agent comprising a peptide with an amino acid sequence selected from the group consisting of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO: 18, SEQ ID NO: 31, and SEQ ID NO: 38, and (b) instructions for using the effective amount of active agent for increasing megakaryocyte production and mobilization, platelet production or a combination thereof.
8. The kit of claim 7 wherein the active agent comprises a peptide with the amino acid sequence of SEQ ID NO:4.
9. The kit of claim 7 wherein the active agent consists of a peptide with the amino acid sequence of SEQ ID NO:4.
10. The kit of any one of claims 7 to 9 further comprising a means for delivery of the active agent.
CA2659664A 1998-03-10 1999-03-08 Improved radiation therapy methods Expired - Fee Related CA2659664C (en)

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
US7738298P 1998-03-10 1998-03-10
US60/077,382 1998-03-10
US8126298P 1998-04-09 1998-04-09
US60/081,262 1998-04-09
US8367098P 1998-04-30 1998-04-30
US60/083,670 1998-04-30
US9009698P 1998-06-19 1998-06-19
US60/090,096 1998-06-19
US9021698P 1998-06-22 1998-06-22
US60/090,216 1998-06-22
US9995798P 1998-09-11 1998-09-11
US60/099,957 1998-09-11
CA002323237A CA2323237C (en) 1998-03-10 1999-03-08 Improved radiation therapy methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CA002323237A Division CA2323237C (en) 1998-03-10 1999-03-08 Improved radiation therapy methods

Publications (2)

Publication Number Publication Date
CA2659664A1 CA2659664A1 (en) 1999-09-16
CA2659664C true CA2659664C (en) 2013-05-07

Family

ID=27557165

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002323237A Expired - Fee Related CA2323237C (en) 1998-03-10 1999-03-08 Improved radiation therapy methods
CA2659664A Expired - Fee Related CA2659664C (en) 1998-03-10 1999-03-08 Improved radiation therapy methods

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CA002323237A Expired - Fee Related CA2323237C (en) 1998-03-10 1999-03-08 Improved radiation therapy methods

Country Status (5)

Country Link
EP (1) EP1061937A1 (en)
JP (1) JP3728400B2 (en)
AU (1) AU744799B2 (en)
CA (2) CA2323237C (en)
WO (1) WO1999045945A1 (en)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6730775B1 (en) 1999-03-23 2004-05-04 University Of Southern California Methods for limiting scar and adhesion formation
US7338938B2 (en) 1999-05-10 2008-03-04 University Of Southern California Methods for treating a patient undergoing chemotherapy
US7122523B2 (en) * 2001-05-01 2006-10-17 University Of Southern California Methods for inhibiting tumor cell proliferation
DK1395566T3 (en) 2001-05-31 2008-01-07 Vicore Pharma Ab Tricyclic compounds useful as angiotensin II agonists
MX2012011036A (en) 2010-03-26 2013-03-18 Univ Southern California Methods for treating combined radiation and thermal injury.
US9439925B2 (en) 2010-04-06 2016-09-13 Synedgen, Inc. Methods and compositions for treating wounds utilizing chitosan compounds
EP2603228A1 (en) * 2010-08-10 2013-06-19 The University Of Southern California Use of angiotensin- ii (1-7) in cell transplantation and as an agent for preventing/treating norovirus infection
EP2455388A1 (en) 2010-11-23 2012-05-23 LanthioPep B.V. Novel angiotensin type 2 (AT2) receptor agonists and uses thereof.
US20150050250A1 (en) * 2012-04-05 2015-02-19 University Of Southern California Cell therapy technology to deliver radio-protective peptides
US8557958B1 (en) 2012-06-18 2013-10-15 Tarix Pharmaceuticals Ltd. Compositions and methods for treatment of diabetes
WO2014047506A1 (en) * 2012-09-20 2014-03-27 Synedgen, Inc. Methods for treatment or prevention of damage resulting from radiation, trauma or shock
US8633158B1 (en) 2012-10-02 2014-01-21 Tarix Pharmaceuticals Ltd. Angiotensin in treating brain conditions
US9333233B2 (en) 2014-02-25 2016-05-10 Tarix Pharmaceuticals Ltd. Methods and compositions for the delayed treatment of stroke
WO2021023698A1 (en) 2019-08-02 2021-02-11 Lanthiopep B.V Angiotensin type 2 (at2) receptor agonists for use in the treatment of cancer

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE457055B (en) * 1986-08-18 1988-11-28 Ferring Ab TOPIC RADIATION GEL FOR MUKOSA CONTAINING VARIETY CONDUCTIVE SUBSTANCES
US5015629A (en) * 1989-06-26 1991-05-14 University Of Southern California Tissue repair
DE69435301D1 (en) * 1993-09-24 2010-08-26 Univ Southern California Use of angiotensin II analogues for tissue repair
WO1997034627A2 (en) * 1996-03-19 1997-09-25 Societe De Conseils De Recherches Et D'applications Scientifiques S.A. Protection of hemopoietic cells during chemotherapy or radiotherapy

Also Published As

Publication number Publication date
CA2659664A1 (en) 1999-09-16
AU2995799A (en) 1999-09-27
AU744799B2 (en) 2002-03-07
CA2323237A1 (en) 1999-09-16
EP1061937A1 (en) 2000-12-27
JP3728400B2 (en) 2005-12-21
CA2323237C (en) 2009-05-19
JP2002506042A (en) 2002-02-26
WO1999045945A1 (en) 1999-09-16

Similar Documents

Publication Publication Date Title
US7776828B2 (en) Radiation therapy methods
US6455500B1 (en) Radiation therapy methods
US6239109B1 (en) Method of promoting erythropoiesis
AU737058B2 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6248587B1 (en) Method for promoting mesenchymal stem and lineage-specific cell proliferation
AU759285B2 (en) Methods to increase white blood cell survival after chemotherapy
CA2659664C (en) Improved radiation therapy methods
WO1999026644A9 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
MXPA00008843A (en) Improved radiation therapy methods
EP1410801B1 (en) Method for promoting hematopoietic cell proliferation and differentiation
MXPA00007509A (en) Method of promoting erythropoiesis
MXPA00011112A (en) Methods to increase white blood cell survival after chemotherapy

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20160308