WO2013158959A1 - Compositions et procédés pour le traitement de la maladie du greffon contre l'hôte - Google Patents

Compositions et procédés pour le traitement de la maladie du greffon contre l'hôte Download PDF

Info

Publication number
WO2013158959A1
WO2013158959A1 PCT/US2013/037308 US2013037308W WO2013158959A1 WO 2013158959 A1 WO2013158959 A1 WO 2013158959A1 US 2013037308 W US2013037308 W US 2013037308W WO 2013158959 A1 WO2013158959 A1 WO 2013158959A1
Authority
WO
WIPO (PCT)
Prior art keywords
angiotensin
peptide
transplantation
gvhd
administered
Prior art date
Application number
PCT/US2013/037308
Other languages
English (en)
Inventor
Richard Franklin
Gere Dizerega
Original Assignee
Tarix Pharmaceuticals Ltd.
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 Tarix Pharmaceuticals Ltd. filed Critical Tarix Pharmaceuticals Ltd.
Publication of WO2013158959A1 publication Critical patent/WO2013158959A1/fr

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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

Definitions

  • GVHD Graft-Versus-Host Disease
  • GVHD can occur after allogeneic cell transplants, such as stem cell transplants and/or bone marrow transplants. GVHD can also occur after blood transfusion. In GVHD, the transplanted immune cells can recognize and attack the cells of the host as foreign.
  • treatment of GVHD typically involves administration of steroids. Steroids are effective for acute GVHD but may cause serious side effects such as infection with bacteria and viruses. Therefore, there is a great need for more effective and safer treatment of GVHD.
  • the present invention provides improved methods for treating or preventing
  • GVHD in particular, based on the use of an angiotensin (1-7) peptide.
  • angiotensin (1-7) peptide As described in the Examples section below, the present invention is, in part, based on the surprising discovery that treatment with an angiotensin (1-7) peptide significantly reduced the occurrence of GVHD in cord blood transplant recipients. Therefore, the present invention represents a more effective and safer treatment for GVHD.
  • the present invention provides methods of treating or reducing risk for Graft- Versus-Host Disease comprising administering to a subject who is suffering from or susceptible to Graft-Versus-Host Disease (GVHD) an angiotensin (1-7) peptide.
  • GVHD Graft-Versus-Host Disease
  • the angiotensin (1-7) peptide is administered at an effective dose periodically at an administration interval such that at least one symptom or feature of Graft-Versus-Host Disease is reduced in intensity, severity, duration, or frequency or has delayed in onset.
  • the at least one symptom or feature of Graft-Versus-Host Disease is selected from liver damage, skin rash, jaundice, intestinal inflammation, sloughing of the mucosal membrane, diarrhea, abdominal pain, nausea, and vomiting.
  • the GVHD is acute GVHD.
  • the GVHD is chronic GVHD.
  • the subject being treated is immune-compromised recipient of transplant.
  • the Graft-Versus-Host Disease is associated with hematopoietic stem cell (HSC) (e.g., bone marrow, peripheral blood (PBSC), and cord blood) transplantation.
  • HSC hematopoietic stem cell
  • PBSC peripheral blood
  • cord blood transplantation is associated with cord blood transplantation.
  • the cord blood transplantation is selected from the group consisting of single cord blood transplantation, double cord blood transplantation, manipulated cord blood transplantation, and combination thereof.
  • the manipulated cord blood transplantation comprises ex vivo expanded cord blood transplantation.
  • the manipulated cord blood transplantation comprises treatment of the cord blood with prostaglandins prior to transplant.
  • the manipulated cord blood transplantation comprises depleting T-cells from the cord blood prior to transplant.
  • the Graft-Versus-Host Disease is associated with bone marrow transplantation. In some embodiments, the Graft-Versus-Host Disease is associated with peripheral blood transplantation. Suitable bone marrow or peripheral blood may be obtained from either children or adults.
  • the Graft-Versus-Host Disease is associated with adult stem cell transplantation.
  • the adult stem cell transplantation is allogeneic adult stem cell transplantation.
  • the Graft-Versus-Host Disease is associated with embryonic stem cell transplantation.
  • the Graft-Versus- Host Disease is associated with organ transplantation.
  • the Graft-Versus- Host Disease is associated with blood transfusion.
  • the angiotensin (1-7) peptide is administered concurrently
  • angiotensin (1-7) peptide is administered periodically subsequent to transplantation.
  • the angiotensin (1-7) peptide is administered for at least 1 day, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 15 weeks, 18 weeks, 21 weeks, or 24 weeks subsequent to the transplantation.
  • the angiotensin (1- 7) peptide is administered for at least 1 month, 2 months, 3 months, 6 months, 1 year, 2 years, or longer subsequent to the transplantation.
  • the angiotensin (1-7) peptide is administered daily, twice a week, weekly, once every two weeks, once every three weeks, monthly, or at a variable interval.
  • the angiotensin (1-7) peptide is administered periodically prior to the transplantation. In some embodiments, the angiotensin (1-7) peptide is administered for at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, or 4 weeks prior to the transplantation. In some embodiments, the angiotensin (1-7) peptide is administered daily, once every two days, twice a week, weekly, or at a variable interval.
  • the angiotensin (1-7) peptide is administered
  • transdermally intravenously, intradermally, orally, by inhalation, transdermally (topical), subcutaneously (e.g., abdomen or thigh), and/or transmucosally.
  • the angiotensin (1-7) peptide is administered at the effective dose of 5-1,500 ⁇ g/kg/day (e.g., 5-1,200 ⁇ g/kg/day, 5-1,000 ⁇ g/kg/day, 50-1,200 ⁇ g/kg/day, 50-1,000 ⁇ g/kg/day, 50-800 ⁇ g/kg/day, 100-1200 ⁇ g/kg/day, 100-800 ⁇ g/kg/day, 100-600 ⁇ g/kg/day, 200-1,500 ⁇ g/kg/day, 200-1,200 ⁇ g/kg/day, 200-1,000 ⁇ g/kg/day, 200-800 ⁇ g/kg/day, 200-600 ⁇ g/kg/day, 300-1,500 ⁇ g/kg/day, 300-1,200 ⁇ g/kg/day, 300-1,000 ⁇ g/kg/day, 300-800 ⁇ g/kg/day, or 300-600 ⁇ g/kg/day).
  • the angiotensin (1-7) peptide is administered at the effective dose of 300-1,000 ⁇ g/kg/day. In some embodiments, the angiotensin (1-7) peptide is administered at the effective dose of 1, 2.5, 5, 10, 20, 25, 50, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, 1,200, 1,300, 1,400, or 1,500 ⁇ g/kg/day.
  • the administration of the angiotensin (1-7) peptide results in substantially free of Stage 4, 3, 2, or 1 acute GVHD in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • the administration of the angiotensin (1-7) peptide results in substantially free of Grade IV, III, II, or I acute GVHD in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • the administration of the angiotensin (1-7) peptide results in substantially free of acute GVHD symptom in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • the administration of the angiotensin (1-7) peptide results in substantially free of chronic GVHD symptoms in the subject at least 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 2.5 years, 3 years, 4 years, 5 years or more following transplantation.
  • the present invention provides methods of transplantation comprising administering an angiotensin (1-7) peptide in conjunction with introducing an allogeneic organ, tissue or cell into a subject.
  • the allogeneic tissue or cell comprises cord blood. In some embodiments, the allogeneic organ, tissue or cell comprises bone marrow. In some embodiments, the allogeneic organ, tissue or cell comprises adult stem cell. In some embodiments, the allogeneic organ, tissue or cell comprises embryonic stem cell. In some embodiments, the allogeneic organ, tissue or cell comprises an organ (i.e., solid organ).
  • the angiotensin (1-7) peptide is administered prior to introducing the allogeneic organ, tissue or cell. In some embodiments, the angiotensin (1-7) peptide is administered concurrently with introducing the allogeneic organ, tissue or cell. In some embodiments, the angiotensin (1-7) peptide is administered subsequent to introducing the allogeneic organ, tissue or cell.
  • the angiotensin (1-7) peptide is administered in
  • the immunosuppressant is selected from the group consisting of antithymocyte globulin, anti-TNF agents, azathioprine or other inosine 5 '-monophosphate dehydrogenase inhibitors, azodiacarbonide, bisindolyl maleimide VIII, brequinar, chlorambucil, CTLA4-Ig, corticosteroids, cyclophosphamide, cyclosporine A, deoxyspergualin, dexamethasone, FK506, glucocorticoids, IL-2 antagonists, leflunomide, mercaptopurine, 6-mercaptopurine, methotrexate, methylprednisolone, mizoribine, mizoribine monophosphate, muromonab CD3, mycophenolate mofetil, OKT3, prednisone, sirolimus, rapamycin, rho (D) immune globin
  • the administration of the angiotensin (1-7) peptide results in reduced intensity, severity, duration, or frequency or delayed onset of at least one symptom or feature of GVHD (e.g., acute or chronic GVHD). In some embodiments, the administration of the angiotensin (1-7) peptide results in reduced intensity, severity, duration, or frequency or delayed onset of at least one symptom or feature of mucositis.
  • an angiotensin (1-7) peptide suitable for the present invention comprises the naturally-occurring Angiotensin (1-7) amino acid sequence of Asp 1 - Arg 2 -Val 3 -Tyr 4 -Ile 5 -His 6 -Pro 7 (SEQ ID NO:l).
  • the functional equivalent is a linear peptide.
  • the linear peptide comprises a sequence that includes at least four amino acids from the seven amino acids that appear in the naturally-occurring Angiotensin (1-7), wherein the at least four amino acids maintain their relative positions as they appear in the naturally- occurring Angiotensin (1-7).
  • the linear peptide comprises a sequence that includes at least five amino acids from the seven amino acids that appear in the naturally- occurring Angiotensin (1-7), wherein the at least five amino acids maintain their relative positions as they appear in the naturally-occurring Angiotensin (1-7).
  • the linear peptide comprises a sequence that includes at least six amino acids from the seven amino acids that appear in the naturally-occurring Angiotensin (1-7), wherein the at least six amino acids maintain their relative positions as they appear in the naturally-occurring Angiotensin (1- 7).
  • the linear peptide contains 4-25 amino acids.
  • the linear peptide is a fragment of the naturally-occurring Angiotensin (1-7).
  • the linear peptide contains amino acid substitutions, deletions and/or insertions in the naturally-occurring Angiotensin (1-7).
  • the linear peptide has an amino acid sequence of Asp -Arg -Nle -Tyr -He -His -Pro (SEQ ID NO:2). In some embodiments, the
  • linear peptide has an amino acid sequence of Asp -Arg -Val -Ser -lie -His -Cys' (SEQ ID NO:3).
  • the functional equivalent is a cyclic peptide.
  • the cyclic peptide comprises a linkage between amino acids.
  • the linkage is located at residues corresponding to positions Tyr 4 and Pro 7 in naturally-occurring Angiotensin (1-7).
  • the linkage is a thioether bridge.
  • the cyclic peptide comprises an amino acid sequence otherwise identical to the naturally-occurring Angiotensin (1-7) amino acid sequence of Asp 1 -Arg 2 -Val 3 -Tyr 4 -Ile 5 -His 6 - Pro 7 (SEQ ID NO: l).
  • the cyclic peptide comprises a norleucine (Nle) replacing position Val 3 in naturally-occurring Angiotensin (1-7).
  • the cyclic peptide is a 4,7-cyclized angiotensin (1-7) with the following formula:
  • the angiotensin (1-7) peptide comprises one or more chemical modifications to increase protease resistance, serum stability and/or bioavailability.
  • the one or more chemical modifications comprise pegylation.
  • the present invention provides methods of treating GVHD including administering to a subject who is suffering from or susceptible to GVHD an angiotensin (1-7) receptor agonist.
  • angiotensin (1-7) receptor agonist is a non-peptidic agonist.
  • the non-peptidic agonist is a compound with the following structure:
  • Acute when used in connection with tissue damage and related diseases, disorders, or conditions, has the meaning understood by any one skilled in the medical art.
  • the term typically refers to a disease, disorder, or condition in which there is sudden or severe onset of symptoms.
  • acute damage is due to an ischemic or traumatic event.
  • the term "acute” is used in contrast to the term “chronic.”
  • Allogeneic means from a different organism of the same species. In the context of transplantation, the term is used to mean that the cells, tissues and/or organs referred to as “allogeneic" are from a different individual than a recipient into which said cells, tissues and/or organs are being transplanted. Typically, allogeneic cells, tissues or organs have different genotype than the recipient.
  • animal refers to any member of the animal kingdom. In some embodiments, “animal” refers to humans, at any stage of development. In some embodiments, “animal” refers to non-human animals, at any stage of development. In certain embodiments, the non-human animal is a mammal ⁇ e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, and/or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, insects, and/or worms. In some embodiments, an animal may be a transgenic animal, genetically-engineered animal, and/or a clone.
  • Autologous means from the same organism. In the context of transplantation, the term is used to mean that the cells, tissues and/or organs referred to as “autologous” are derived from the recipient itself. Typically, autologous cells, tissues and/or organs do not contain any substantial amount of material which could be regarded as allogeneic or xenogeneic, that is to say derived from a “foreign” cellular source.
  • Biologically active refers to a characteristic of any agent that has activity in a biological system, and particularly in an organism.
  • an agent that, when administered to an organism, has a biological effect on that organism is considered to be biologically active.
  • a peptide is biologically active
  • a portion of that peptide that shares at least one biological activity of the peptide is typically referred to as a "biologically active" portion.
  • a peptide has no intrinsic biological activity but that inhibits the effects of one or more naturally- occurring angiotensin compounds is considered to be biologically active.
  • Carrier or diluent refers to a pharmaceutically acceptable (e.g., safe and non-toxic for administration to a human) carrier or diluting substance useful for the preparation of a pharmaceutical formulation.
  • exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • Chronic As used herein, the term “chronic,” when used in connection with tissue damage or related diseases, disorders, or conditions has the meaning as understood by any one skilled in the medical art. Typically, the term “chronic” refers to diseases, disorders, or conditions that involve persisting and/or recurring symptoms. Chronic diseases, disorders, or conditions typically develop over a long period of time. The term “chronic” is used in contrast to the term “acute.” In some embodiments, a chronic disease, disorder, or condition results from cell degeneration. In some embodiments, a chronic disease, disorder, or condition results from age-related cell degeneration.
  • Dosage form As used herein, the terms “dosage form” and “unit dosage form” refer to a physically discrete unit of a therapeutic agent for the patient to be treated. Each unit contains a predetermined quantity of active material calculated to produce the desired therapeutic effect. It will be understood, however, that the total dosage of the composition will be decided by the attending physician within the scope of sound medical judgment.
  • Dosing regimen is a set of unit doses (typically more than one) that are administered individually to a subject, typically separated by periods of time.
  • a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses.
  • a dosing regimen comprises a plurality of doses each of which are separated from one another by a time period of the same length; in some embodiments, a dosing regime comprises a plurality of doses and at least two different time periods separating individual doses.
  • the therapeutic agent is administered continuously over a predetermined period. In some embodiments, the therapeutic agent is administered once a day (QD) or twice a day (BID).
  • Functional equivalent or derivative denotes, in the context of a functional derivative of an amino acid sequence, a molecule that retains a biological activity (either function or structural) that is substantially similar to that of the original sequence.
  • a functional derivative or equivalent may be a natural derivative or is prepared synthetically.
  • Exemplary functional derivatives include amino acid sequences having substitutions, deletions, or additions of one or more amino acids, provided that the biological activity of the protein is conserved.
  • the substituting amino acid desirably has chemico-physical properties which are similar to that of the substituted amino acid. Desirable similar chemico-physical properties include, similarities in charge, bulkiness, hydrophobicity, hydrophilicity, and the like.
  • “reduce,” or grammatical equivalents indicate values that are relative to a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of the treatment described herein.
  • a “control individual” is an individual afflicted with the same form of disease as the individual being treated, who is about the same age as the individual being treated (to ensure that the stages of the disease in the treated individual and the control individual(s) are comparable).
  • in vitro refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, etc., rather than within a multi-cellular organism.
  • in vivo refers to events that occur within a multi-cellular organism, such as a human and a non-human animal. In the context of cell-based systems, the term may be used to refer to events that occur within a living cell (as opposed to, for example, in vitro systems).
  • Isolated refers to a substance and/or entity that has been (1) separated from at least some of the components with which it was associated when initially produced (whether in nature and/or in an experimental setting), and/or (2) produced, prepared, and/or manufactured by the hand of man. Isolated substances and/or entities may be separated from at least about 10%, about 20%>, about 30%>, about 40%>, about 50%>, about 60%, about 70%, about 80%, about 90%, about 95%, about 98%, about 99%, substantially 100%, or 100% of the other components with which they were initially associated.
  • isolated agents are more than about 80%>, about 85%, about 90%>, about 91%>, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, substantially 100%), or 100%) pure.
  • a substance is "pure” if it is substantially free of other components.
  • isolated cell refers to a cell not contained in a multi-cellular organism.
  • Prevent As used herein, the term “prevent” or “prevention”, when used in connection with the occurrence of a disease, disorder, and/or condition, refers to reducing the risk of developing the disease, disorder and/or condition. See the definition of "risk.”
  • Polypeptide refers a sequential chain of amino acids linked together via peptide bonds. The term is used to refer to an amino acid chain of any length, but one of ordinary skill in the art will understand that the term is not limited to lengthy chains and can refer to a minimal chain comprising two amino acids linked together via a peptide bond. As is known to those skilled in the art, polypeptides may be processed and/or modified.
  • Protein refers to one or more polypeptides that function as a discrete unit. If a single polypeptide is the discrete functioning unit and does not require permanent or temporary physical association with other polypeptides in order to form the discrete functioning unit, the terms “polypeptide” and “protein” may be used interchangeably. If the discrete functional unit is comprised of more than one polypeptide that physically associate with one another, the term “protein” refers to the multiple polypeptides that are physically coupled and function together as the discrete unit.
  • a "risk" of a disease, disorder, and/or condition comprises a likelihood that a particular individual will develop a disease, disorder, and/or condition ⁇ e.g., GVHD).
  • risk is expressed as a percentage.
  • risk is from 0,1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90 up to 100%.
  • risk is expressed as a risk relative to a risk associated with a reference sample or group of reference samples.
  • a reference sample or group of reference samples have a known risk of a disease, disorder, condition and/or event (e.g., GVHD).
  • a reference sample or group of reference samples are from individuals comparable to a particular individual.
  • relative risk is 0,1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, or more.
  • Stability refers to the ability of the therapeutic agent to maintain its therapeutic efficacy (e.g., all or the majority of its intended biological activity and/or physiochemical integrity) over extended periods of time.
  • the stability of a therapeutic agent, and the capability of the pharmaceutical composition to maintain stability of such therapeutic agent may be assessed over extended periods of time (e.g., for at least 1, 3, 6, 12, 18, 24, 30, 36 months or more).
  • pharmaceutical compositions described herein have been formulated such that they are capable of stabilizing, or alternatively slowing or preventing the degradation, of one or more therapeutic agents formulated therewith.
  • a stable formulation is one in which the therapeutic agent therein essentially retains its physical and/or chemical integrity and biological activity upon storage and during processes (such as freeze/thaw, mechanical mixing and lyophilization).
  • Subject refers to a human or any non-human animal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate).
  • a human includes pre and post natal forms.
  • a subject is a human being.
  • a subject can be a patient, which refers to a human presenting to a medical provider for diagnosis or treatment of a disease.
  • the term "subject” is used herein interchangeably with “individual” or "patient.”
  • a subject can be afflicted with or is susceptible to a disease or disorder but may or may not display symptoms of the disease or disorder.
  • the term “substantially” refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest.
  • One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result.
  • the term “substantially” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
  • Susceptible to An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition.
  • an individual who is susceptible to a disease, disorder, condition, or event may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, condition, and/or event (5) having undergone, planning to undergo, or requiring a transplant.
  • a genetic mutation associated with development of the disease, disorder, and/or condition for example, GVHD
  • a genetic polymorphism associated with development of the disease, disorder, and/or condition
  • increased and/or decreased expression and/or activity of a protein associated with the disease, disorder, and/or condition (4) habits and/or lifestyles associated with development of the disease, disorder, condition, and/or event (5) having undergone, planning to undergo, or requiring a transplant.
  • an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
  • therapeutically effective amount As used herein, the term "therapeutically effective amount" of a therapeutic agent means an amount that is sufficient, when administered to a subject suffering from or susceptible to a disease, disorder, and/or condition, to treat, diagnose, prevent, and/or delay the onset of the symptom(s) of the disease, disorder, and/or condition. It will be appreciated by those of ordinary skill in the art that a therapeutically effective amount is typically administered via a dosing regimen comprising at least one unit dose.
  • Transplant refers to the transfer of cells, tissues and/or organ(s) to an individual.
  • the transferred cells, tissues, and/or organ(s) are allogeneic cells, tissues and/or organ(s) from another individual (e.g., an individual of the same species as the recipient).
  • the transferred cells, tissues and/or organ(s) are autologous cells, tissues and/or organ(s) taken from the same individual.
  • the autologous cells, tissues and/or organ(s) from the individual are taken at an earlier time point.
  • the transferred cells are blood cells (e.g., cord blood cells).
  • the transferred cells are manipulated or expanded blood cells (e.g., cord blood cells).
  • the transferred cells are bone marrow cells.
  • the transferred cells are adult stem cells.
  • the transferred cells are embryonic stem cells.
  • the transferred cells are induced pluripotent stem cells.
  • Treating refers to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a particular disease, disorder, and/or condition. Treatment may be administered to a subject who does not exhibit signs of a disease and/or exhibits only early signs of the disease for the purpose of decreasing the risk of developing pathology associated with the disease.
  • the present invention provides, among other things, improved compositions and methods for treating or reducing risk of GVHD based on the use of angiotensin-(l-7) peptides, angiotensin (1-7) receptor agonists, and/or functional equivalents, analogs or derivatives thereof.
  • angiotensin-(l-7) peptides angiotensin (1-7) receptor agonists
  • functional equivalents analogs or derivatives thereof.
  • An angiotensin (1-7) peptide and/or angiotensin (1-7) receptor agonist as described herein can be used for treating or reducing risk of GVHD.
  • GVHD can occur when an immune response is mounted in response to allogeneic transplantation including, but not limited to, hematopoietic stem cell (HSC) transplantation such as bone marrow, peripheral blood (PBSC), or cord blood transplantation, other types of stem cell transplantation, and/or solid organ transplantation.
  • HSC hematopoietic stem cell
  • PBSC peripheral blood
  • cord blood transplantation other types of stem cell transplantation
  • solid organ transplantation other types of stem cell transplantation
  • GVHD may also occurs with lower frequency following syngeneic and autologous transplant.
  • GVHD is well known in the art (see, for example, Ferrara, J. et al, "Graft- versus- host disease” Lancet. 2009;373(9674): 1550-61; MacMillan, M. et al., "Acute graft-versus-host disease after unrelated donor umbilical cord blood transplantation: analysis of risk factors," Blood, 2009, 113(11): 2410-2415; Matsumura, T. .et al., "Allogeneic cord blood transplantation for adult acute lymphoblastic leukemia: retrospective survey involving 256 patients in Japan,” Leukemia. 2012 Jan 17 epub; Kobayashi, K.
  • MHC proteins major histocompatibility complex proteins
  • Additional minor histocompatibility proteins exist which can also contribute to immunological recognition events.
  • the individual mammal's immune system can recognize its own MHC proteins, or those of its identical twin, as self and thus does not destroy its own cells or those of its identical twin.
  • HLA histocompatibility proteins
  • tissue such as bone marrow, blood cells, or solid organs
  • the recipient may recognize the donor's cells as non-self and the recipient's immune system may destroy the donor's cells. For this reason, in a tissue
  • transplantation the recipient can be subjected to immunosuppressive drugs and/or irradiation.
  • immunosuppressive drugs and/or irradiation the recipient can also be subject to immunologic recognition in the opposite direction, that is, the donor tissue may contain immunologically competent cells which proceed to destroy the recipient's cells.
  • GVHD can develop when any allogeneic cells, for example cord blood, bone marrow, peripheral blood, adult stem cells, embryonic stem cells, blood products, and/or solid organs containing immunocompetent cells are transferred from a donor to a recipient.
  • any allogeneic cells for example cord blood, bone marrow, peripheral blood, adult stem cells, embryonic stem cells, blood products, and/or solid organs containing immunocompetent cells are transferred from a donor to a recipient.
  • T-lymphocytes from the donor recognize the differences based HLA antigens and attack the new body, i.e., the recipient's body.
  • GVHD results when immunocompetent T cells in the donor graft are infused into an immuno-compromised recipient.
  • GVHD may also develop when there are antigenic differences between donor and recipient for the minor histocompatibility antigens. Thus, GVHD can also develop between MHC-matched persons.
  • surgery patients who receive directed blood transfusion for example, transfusion of blood from an HLA homozygous child to a heterozygous parent, may also develop GVHD.
  • GVHD occurs when the blood is transfused into an immuno-compromised patient (e.g. organ transplant patients on high dose immunosuppressives, children with primary immunodeficiencies, or (years ago) into HIV infected patients with AIDS.
  • Acute GVHD can occur within the first 100 days following a transplant. Without wishing to be held to a particular theory, it is thought that T-cells present in the donor's tissue and/or cells at the time of transplant can attack the patient's skin, liver, stomach, and/or intestines. The earliest signs of acute GVHD can be a skin rash that appears on the hand, feet and face. Other than blistering skin, patients with severe GVHD can also develop large amounts of watery or bloody diarrhea with cramping from donor T-cells attacking the stomach and intestines. Jaundice (yellowing of the skin and eyes) is a usual indication that GVHD disease involves the liver. The severity of acute GVHD disease can be assessed by the number of organs involved and the degree of symptoms.
  • Cases of acute GVHD can be categorized into stage depending on clinical severity
  • Stage 1 comprises a skin rash over less than 25% of the body.
  • Stage 2 comprises a skin rash over more than 25% of the body accompanied by mild liver or stomach and intestinal disorders.
  • Stage 3 comprises redness of the skin, similar to a severe sunburn, and moderate liver, stomach and intestinal problems.
  • Stage 4 comprises blistering, peeling skin, and severe liver, stomach, and intestinal problems.
  • acute GVHD can also be characterized into five Clinical Grades 0, 1, II, III, and IV.
  • Grade 0 is substantially symptom free with respect to skin, liver, gut or functional impairment.
  • Grade I is considered mild with skin stage of 1 to 2.
  • Grade II is considered moderate and characterized with skin stage of 1 to 3, liver and gut stage of 1 and functional impairment stage of 1.
  • Grade III is considered severe and characterized with skin stage of 2 to 3, liver and gut stage of 2-3, and functional impairment stage of 2.
  • Grade IV is considered life-threatening and characterized with skin stage of 2 to 4, liver and gut stage of 2 to 4, and functional impairment stage of 3. Exemplary detailed staging and grading are further described in the Examples section.
  • Chronic GVHD can occur after the first 100 days following a transplant. Without wishing to be held to a particular theory, it is thought that Chronic GVHD can be caused by T- cells produced by engrafted tissue and/or cells. The same organs and systems can be attacked as in acute GVHD and additionally chronic GVHD can be associated with damage to connective tissue. Patients with chronic GVHD can experience skin problems that may include a dry itching rash, a change in skin color, and tautness or tightening of the skin. Partial hair loss or premature graying may also occur. Similarly to patients with acute GVHD, patients with chronic GVHD may show jaundice as a sign of liver involvement.
  • Chronic GVHD can also attack glands in the body that secrete mucous, saliva or other lubricants. Patients with chronic GVHD can experience dryness or stinging in their eyes due to impairment of the lacrimal gland. Glands that secrete saliva in the mouth can also be affected by chronic GVHD and, less often, those that lubricate the esophagus, making swallowing and eating difficult. Patients with chronic GVHD can experience a burning sensation in their mouths when using toothpaste or eating acidic foods. Chronic GVHD can attack glands that lubricate the stomach lining and intestines, interfering with the body's ability to properly absorb nutrients. Symptoms can include heartburn, stomach pain and/or weight loss. Occasionally patients with chronic GVHD can experience
  • GVHD can occur after a recipient receives a transplant of allogeneic cells, tissues or organs (for example cord blood, bone marrow, adult stem cells, embryonic stem cells, blood products, and/or solid organs).
  • allogeneic cells for example cord blood, bone marrow, adult stem cells, embryonic stem cells, blood products, and/or solid organs.
  • One type of transplant that can result in GVHD is a cord blood transplant.
  • Cord blood comprises red blood cells, white blood cells, plasma, platelets, and hematopoietic stem cells.
  • Cord blood is of particular interest in treating bone marrow
  • Cord blood can be obtained from the placenta or umbilical cord after childbirth.
  • cord blood can be obtained by sterilely puncturing the vein of the severed umbilical cord and collecting the blood.
  • the retrieved blood can be cryopreserved.
  • Methods of obtaining and storing cord blood are well known in the art (see, for example, Sirchia, G. et al., "Placental/umbilical cord blood transplantation,” Haematologica. 1999 Aug;84(8):738-47).
  • Cord blood is transplanted intraveneously.
  • Cord blood transplantation can involve the transplantation of cord blood cells into patients with naturally and/or artificially reduced numbers of hematopoietic stem cells to increase their number of hematopoietic stem cells and/or into patients whose naturally occurring hematopoietic stem cells are suffering from a disease, disorder or condition to replace their hematopoietic stem cells with healthy stem cells.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a cord blood
  • Hematologic malignancies are a closely related group of cancers that affect blood, bone marrow, and/or lymph nodes. In some
  • hematologic malignancies can include leukemias, including but not limited to acute lymphoblastic leukemia (also called acute lymphocytic leukemia); acute myeloid leukemia (also called acute myelogenous leukemia) such as but not limited to myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia leukemias and myelodysplastic syndrome; chronic lymphocytic leukemia (also called chronic lymphocytic leukemia); chronic myelogenous leukemia (also called chronic myeloid leukemia) and/ or hairy cell leukemia.
  • acute lymphoblastic leukemia also called acute lymphocytic leukemia
  • acute myeloid leukemia also called acute myelogenous leukemia
  • myelogenous leukemia such as but not limited to myeloblastic, promyelocytic, myelomonocytic, monocytic, and ery
  • hematologic malignancies can include lymphomas, including but not limited to Hodgkin's lymphoma and non-Hodgkin's lymphoma. In some embodiments, hematologic malignancies can include myelomas.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a cord blood
  • Diseases that affect bone marrow production can include, but are not limited to sickle cell anemia, aplastic anemia, thalassemia, congenital neutropenia, and/or severe immunodeficiency syndromes.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a cord blood
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a cord blood
  • myeloablative therapy substantially all of a patient's hematopoietic stem cells can be eliminated before transplantation, through cell killing or cell inactivation. This can be done through high doses of chemotherapy or radiation therapy.
  • a combination of cyclophosphamide with busulfan can be used.
  • total body irradiation can be used.
  • myeloablation can also suppress a subject's immune response, reducing the chance of rejection by the host. Methods of myeloablation are well known in the art as described, for example, in Sirchia et al. ("Placental/umbilical cord blood transplantation," Haematologica. 1999 Aug;84(8):738-47).
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a cord blood
  • myeloablative therapy In non-myeloablative therapy,
  • chemotherapy and radiation can be given before transplantation at dosages sufficient to transiently injure but not eliminate a patient's hematopoietic stem cells.
  • Methods for non- myeloablative therapy are well known in the art, as described for example, in Spitzer, T.
  • a chimeric hematopoietic stem cell population can exist, consisting of the patient's original cells and donor cells.
  • Non-myeloablative transplantations relies on the principal of the "graft- versus-malignancy" (GVM), "graft-versus-tumor,” or "graft-versus- leukemia” effect.
  • GVM graft- versus-malignancy
  • graft-versus-tumor graft-versus- leukemia
  • Transplantations preceded by non-myeloablative therapy can be used in patients who are older or who have other medical conditions that would make them unable to tolerate the toxic chemotherapy effects of regular transplants.
  • Transplantations preceded by non- myeloablative therapy can also have a role in treating patients who are in remission with a high- risk cancer, such as acute myelogenous leukemia, or who have had a relapse after a previous transplant.
  • cord blood transplantations can be highly dependent on the number of cells transplanted.
  • One disadvantage of cord blood transplants is that the number of cells contained in any one cord blood unit is small.
  • banked cord blood units have 120 x 10 7 nucleated cells, whereas the nucleated cell dose required for successful engraftment is 3.7 x 10 7 nucleated cells/kg.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a single cord blood transplantation.
  • a single cord blood transplantation a single unit of cord blood from a single individual is transplanted to a patient. As discussed above, the single unit must have a sufficient number of cells for the patient. The use of a single cord blood transplant can be advantageous, given that only a single type of donor cells is introduced.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a manipulated cord blood transplantation (e.g., manipulated to deplete T-cells or treated with prostaglandins). T-cell depletion can reduce the risk and/or severity of GVHD. Methods for depleting T-cells ex vivo can rely on negative selection with anti-T cell antibodies and are well known in the art as described in Ho, V.
  • manipulated cord blood comprises cord blood treated with prostaglandins.
  • Treatment with prostaglandins can expand the population of hematopoietic stem cells in the cord blood.
  • Experimental data in zebrafish and mouse models has demonstrated that pretreatment with prostaglandin can be used to increase hematopoietic stem cell formation ex vivo (for example, see North, T. et al., "Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis", Nature 447, 1007-1011, 2007).
  • cord blood cells are treated with PGE2.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving an expanded cord blood transplantation.
  • expanded cord blood comprises cord blood in which hematopoietic stem cells in cord blood are expanded ex vivo prior to administration by selection for CD34+ cells, as is well known in the art and described, for example, in Shpall, E. et al, "Transplantation of Ex Vivo Expanded Cord Blood," Biology of Blood and Marrow
  • expanded cord blood comprises cord blood in which hematopoietic stem cells in cord blood are expanded ex vivo prior to
  • expanded cord blood comprises cord blood in which hematopoietic stem cells in cord blood are expanded ex vivo prior to administration by treatment with nicotinamide, as described, for example, in Peled, T.
  • an angiotensin (1-7) peptide described herein can be administered to treat or reduce the risk of GVHD in patients receiving a double cord blood transplantation.
  • Methods for double cord blood transplantation are well known in the art, as described, for example, by Sideri, A. et al, in "An overview of the progress on double umbilical cord blood transplantation", (Haematologica. 2011 Aug;96(8): 1213-20) and MacMillan, M. et al. in "Acute graft-versus-host disease after unrelated donor umbilical cord blood transplantation: analysis of risk factors," (Blood, 2009, 113(11): 2410-2415).
  • a double cord blood transplantation comprises simultaneous administration of two units of cord blood from separate donors. When such a transplant is performed, there can be an initial state of chimerism, with blood cells from both of the donors being present, followed by predominance of blood from one of the donors. While double cord blood transplantations over single cord blood transplantations can increase cells for transplantation, they can also increase risk of graft versus graft disease.
  • HLA typing One important consideration in cord blood transplantation is HLA typing.
  • the risk of GVHD has been shown to decrease with increased HLA matching.
  • a match of at least 4 of 6 markers is required at HLA-A,-B, and-DRBl .
  • These minimum requirements are based on research studies of transplant outcomes. Patients receiving transplants preceded with non- myeloablative therapy can often develop at least a mild form of chronic GVHD that is thought to be associated with the "graft-versus-tumor" effect.
  • GVHD can also occur when a patient receives additional types of transplants known in the art. Accordingly, in some embodiments, an angiotensin (1-7) peptide described herein is administered to treat or reduce the risk of GVHD in patients receiving these types of transplants.
  • an angiotensin (1-7) peptide described herein is administered to treat or reduce the risk of GVHD in patients receiving these types of transplants.
  • transplants include, for example, bone marrow transplantation (see, for example, Blazar, B. et al., "Bone marrow transplantation and approaches to avoid graft-versus-host disease (GVHD),” Philos Trans R Soc Lond B Biol Sci., 2005, 360(1461): 1747-67) adult stem cell transplantation (see, for example, Krstevska, S. et al, "Acute graft versus host disease in hematopoietic stem cell allotransplant recipients," Med Arh.
  • mucositis can occur.
  • mucositis is caused by damage to mucosal tissues throughout the body including, but not limited to, oral, buccal, sublingual, nasal, vaginal, rectal, aural, lung, and gastrointestinal mucosa.
  • damage to mucosal tissues are caused by inflammation and involves redness and ulcerative sores in the soft tissues of the mucosa.
  • mucositis can occur anywhere along the gastrointestinal (GI) tract.
  • GI gastrointestinal
  • mucositis occurs in the mouth, also referred to as oral mucositis.
  • mucositis usually occurs as an adverse effect of chemotherapy and radiotherapy treatment for cancer.
  • Mucositis can be categorized into clinical grade depending on clinical severity.
  • Various methods an instruments have been developed to stage and measure mucositis severity including, but not limited to, National Cancer Institute's Common Toxicity Criteria (NCI CTC), World Health Organization's (WHO's) Oral Toxicity Scale (OTS).
  • NCI CTC National Cancer Institute's Common Toxicity Criteria
  • WHO's World Health Organization's
  • OTS Oral Toxicity Scale
  • WHO's oral toxicity scale includes Grade 1, 2,3 and 4.
  • Grade 1 involves soreness with or without erythema.
  • Grade 2 includes erythema and ulcers. Patients can swallow solid food.
  • Grade 3 involves ulcers with extensive erythema. Patient cannot swallow food.
  • Grade 4 involves mucositis to the extent that alimentation is not possible.
  • Grade 3 and 4 mucositis are considered severe. Exemplary detailed grading is further described in the Example section.
  • angiotensin (1-7) peptide refers to both naturally- occurring Angiotensin (1-7) and any functional equivalent, analogue or derivative of naturally- occurring Angiotensin (1-7).
  • peptide and polypeptide are interchangeable terms and refer to two or more amino acids bound together by a peptide bond.
  • the terms “peptide” and “polypeptide” include both linear and cyclic peptide.
  • the terms “angiotensin-(l-7)", “Angiotensin ⁇ 1-7)", “Ang-(l-7)", and "TXA-127" are used
  • Naturally-occurring Angiotensin (1-7) (also referred to as Ang-(l-7)) is a seven amino acid peptide shown below:
  • Angiotensinogen which is an ⁇ -2-globulin that is produced constitutively and released into the circulation mainly by the liver.
  • Angiotensinogen is a member of the serpin family and also known as renin substrate.
  • Human angiotensinogen is 452 amino acids long, but other species have angiotensinogen of varying sizes. Typically, the first 12 amino acids are the most important for angiotensin activity:
  • angiotensin may be formed by the action of various enzymes.
  • Angiotensin (1-7) is generated by action of Angiotensin-converting enzyme 2 (ACE 2).
  • ACE 2 Angiotensin-converting enzyme 2
  • Ang-(l-7) is an endogenous ligand for Mas receptors.
  • Mas receptors are G- protein coupled receptor containing seven transmembrane spanning regions.
  • angiotensin-(l-7) receptor' encompasses the G Protein-Coupled Mas Receptors.
  • Angiotensin (1-7) peptide purified from natural sources and any recombinantly produced or chemically synthesized peptides that have an amino acid sequence identical to that of the naturally-occurring Angiotensin (1-7).
  • an angiotensin (1-7) peptide suitable for the present invention is a functional equivalent of naturally-occurring Ang-(l-7).
  • a functional equivalent of naturally-occurring Ang-(l-7) refers to any peptide that shares amino acid sequence identity to the naturally-occurring Ang-(l-7) and retain substantially the same or similar activity as the naturally-occurring Ang-(l-7).
  • a functional equivalent of naturally-occurring Ang-(l-7) described herein has pro-angiogenic activity as determined using methods described herein or known in the art, or an activity such as nitric oxide release, vasodilation, improved endothelial function, antidiuresis, or one of the other properties discussed herein, that positively impacts angiogenesis.
  • a functional equivalent of naturally-occurring Ang-(l-7) described herein can bind to or activate an angiotensin ⁇ 1-7) receptor (e.g., the G protein-coupled Mas receptor) as determined using various assays described herein or known in the art.
  • angiotensin ⁇ 1-7) receptor e.g., the G protein-coupled Mas receptor
  • a functional equivalent of Ang-(l-7) is also referred to as an angiotensin (1-7) analogue or derivative, or functional derivative.
  • a functional equivalent of angiotensin (1-7) shares amino acid sequence similarity to the naturally-occurring Ang-(l-7).
  • a functional equivalent of Ang-(l-7) according to the invention contains a sequence that includes at least 3 (e.g., at least 4, at least 5, at least 6, at least 7) amino acids from the seven amino acids that appear in the naturally-occurring Ang-(l-7), wherein the at least 3 (e.g., at least 4, at least 5, at least 6, or at least 7) amino acids maintain their relative positions and/or spacing as they appear in the naturally-occurring Ang-(l-7).
  • a functional equivalent of Ang-(l-7) also encompasses any peptide that contains a sequence at least 50% (e.g., at least 60%, 70%>, 80%>, or 90%>) identical to the amino acid sequence of naturally-occurring Ang-(l-7). Percentage of amino acid sequence identity can be determined by alignment of amino acid sequences. Alignment of amino acid sequences can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • the WU-BLAST-2 software is used to determine amino acid sequence identity (Altschul et ah, Methods in Enzymology 266, 460-480 (1996); http://blast.wustl/edu/blast/README.html). WU-BLAST-2 uses several search parameters, most of which are set to the default values.
  • HSP score (S) and HSP S2 parameters are dynamic values and are established by the program itself, depending upon the composition of the particular sequence, however, the minimum values may be adjusted and are set as indicated above.
  • Ang-(l-7) is a fragment of the naturally-occurring Ang-(l-7).
  • a functional equivalent, analogue or derivative of Ang-(l-7) contains amino acid substitutions, deletions and/or insertions in the naturally-occurring Ang-(l-7).
  • Ang-(l-7) functional equivalents, analogues or derivatives can be made by altering the amino acid sequences by substitutions, additions, and/or deletions.
  • one or more amino acid residues within the sequence of the naturally-occurring Ang-(l-7) SEQ ID NO: l
  • Substitution for an amino acid within the sequence may be selected from other members of the class to which the amino acid belongs.
  • the positively charged (basic) amino acids include arginine, lysine, and histidine.
  • the nonpolar (hydrophobic) amino acids include leucine, isoleucine, alanine, phenylalanine, valine, proline, tryptophan, and methionine.
  • the uncharged polar amino acids include serine, threonine, cysteine, tyrosine, asparagine, and glutamine.
  • the negatively charged (acid) amino acids include glutamic acid and aspartic acid.
  • the amino acid glycine may be included in either the nonpolar amino acid family or the uncharged (neutral) polar amino acid family. Substitutions made within a family of amino acids are generally understood to be conservative substitutions. For example, the amino acid sequence of a peptide inhibitor can be modified or substituted.
  • angiotensin-(l-7) peptide can be of any length.
  • an angiotensin-(l-7) peptide according to the present invention can contain, for example, from 5-25 amino acid residues, such as 5-20, 5-15 or 5-10 amino acid residues.
  • an Ang-(l-7) peptide according to the present invention contain 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24 or 25 residues.
  • an angiotensin-(l-7) peptide contains one or more modifications to increase protease resistance, serum stability and/or bioavailability.
  • suitable modifications are selected from pegylation, acetylation, glycosylation, biotinylation, substitution with D-amino acid and/or un-natural amino acid, and/or cyclization of the peptide.
  • amino acid in its broadest sense, refers to any compound and/or substance that can be incorporated into a polypeptide chain.
  • an amino acid has the general structure H 2 N-C(H)(R)-COOH.
  • an amino acid is a naturally-occurring amino acid.
  • an amino acid is a synthetic or un-natural amino acid (e.g., ⁇ , ⁇ -disubstituted amino acids, N-alkyl amino acids); in some embodiments, an amino acid is a D-amino acid; in certain embodiments, an amino acid is an L-amino acid.
  • Standard amino acid refers to any of the twenty standard amino acids commonly found in naturally occurring peptides including both L- and D- amino acids which are both incorporated in peptides in nature.
  • Nonstandard or “unconventional amino acid” refers to any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetically or obtained from a natural source.
  • synthetic or unnatural amino acid encompasses chemically modified amino acids, including but not limited to salts, amino acid derivatives (such as amides), and/or substitutions.
  • Amino acids including carboxy- and/or amino-terminal amino acids in peptides, can be modified by methylation, amidation, acetylation, and/or substitution with other chemical groups that can change the peptide's circulating half-life without adversely affecting its activity. Examples of
  • unconventional or un-natural amino acids include, but are not limited to, citrulline, ornithine, norleucine, norvaline, 4-(E)-butenyl-4(i?)-methyl-N-methylthreonine (MeBmt), N-methyl- leucine (MeLeu), aminoisobutyric acid, statine, and N-methyl-alanine (MeAla).
  • Amino acids may participate in a disulfide bond.
  • amino acid is used interchangeably with "amino acid residue,” and may refer to a free amino acid and/or to an amino acid residue of a peptide. It will be apparent from the context in which the term is used whether it refers to a free amino acid or a residue of a peptide.
  • angiotensin-(l-7) peptides contain one or more L-amino acids, D-amino acids, and/or un-natural amino acids.
  • peptidomimetics or peptide analogs are also encompassed by the present invention.
  • Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide.
  • the non-peptide compounds are termed "peptide mimetics" or peptidomimetics (Fauchere et al, Infect. Immun. 54:283-287 (1986); Evans et al, J. Med. Chem. 30:1229-1239 (1987)).
  • Peptide mimetics that are structurally related to
  • therapeutically useful peptides and may be used to produce an equivalent or enhanced
  • paradigm polypeptide i.e., a polypeptide that has a biological or pharmacological activity
  • Such peptide mimetics may have significant advantages over naturally- occurring polypeptides including more economical production, greater chemical stability, enhanced pharmacological properties (e.g., half-life, absorption, potency, efficiency, etc.), reduced antigenicity and others.
  • Angiotensin (1-7) peptides also include other types of peptide derivatives containing additional chemical moieties not normally part of the peptide, provided that the derivative retains the desired functional activity of the peptide.
  • Examples of such derivatives include (1) N-acyl derivatives of the amino terminal or of another free amino group, wherein the acyl group may be an alkanoyl group (e.g., acetyl, hexanoyl, octanoyl) an aroyl group (e.g., benzoyl) or a blocking group such as F-moc (fluorenylmethyl-O-CO-); (2) esters of the carboxy terminal or of another free carboxy or hydroxyl group; (3) amide of the carboxy-terminal or of another free carboxyl group produced by reaction with ammonia or with a suitable amine; (4) phosphorylated derivatives; (5) derivatives conjugated to an antibody or other biological ligand and other types of derivatives; and
  • Angiotensin (1-7) peptides may be obtained by any method of peptide synthesis known to those skilled in the art, including synthetic (e.g., exclusive solid phase synthesis, partial solid phase synthesis, fragment condensation, classical solution synthesis, native-chemical ligation) and recombinant techniques.
  • the peptides or peptides derivatives can be obtained by solid phase peptide synthesis, which in brief, consist of coupling the carboxyl group of the C-terminal amino acid to a resin (e.g., benzhydrylamine resin, chloromethylated resin, hydroxymethyl resin) and successively adding N-alpha protected amino acids.
  • the protecting groups may be any such groups known in the art.
  • angiotensin (1-7) peptide it may be desirable to protect sensitive reactive groups on any of the molecule concerned. This may be achieved by means of conventional protecting groups such as those described in Protective Groups In Organic Synthesis by T.W. Greene & P.G.M. Wuts, 1991, John Wiley and Sons, New- York; and Peptides: chemistry and Biology by Sewald and Jakubke, 2002, Wiley- VCH, Wheinheim p.142.
  • alpha amino protecting groups include acyl type protecting groups (e.g., trifluoroacetyl, formyl, acetyl), aliphatic urethane protecting groups (e.g., t- butyloxycarbonyl (BOC), cyclohexyloxycarbonyl), aromatic urethane type protecting groups (e.g., fluorenyl-9-methoxy-carbonyl (Fmoc), benzyloxycarbonyl (Cbz), Cbz derivatives) and alkyl type protecting groups (e.g., triphenyl methyl, benzyl).
  • acyl type protecting groups e.g., trifluoroacetyl, formyl, acetyl
  • aliphatic urethane protecting groups e.g., t- butyloxycarbonyl (BOC), cyclohexyloxycarbonyl
  • aromatic urethane type protecting groups e.g., fluorenyl-9
  • the amino acids side chain protecting groups include benzyl (for Thr and Ser), Cbz (Tyr, Thr, Ser, Arg, Lys), methyl ethyl, cyclohexyl (Asp, His), Boc (Arg, His, Cys) etc.
  • the protecting groups may be removed at a convenient subsequent stage using methods known in the art.
  • Angiotensin (1-7) peptides may be synthesized according to the FMOC protocol in an organic phase with protective groups. Desirably, the peptides are purified with a yield of 70% with high-pressure liquid chromatography (HPLC) on a CI 8 chromatography column and eluted with an acetonitrile gradient of 10-60%. The molecular weight of a peptide can be verified by mass spectrometry (reviewed in Fields, G.B. "Solid-Phase Peptide Synthesis” Methods in Enzymology. Vol. 289, Academic Press, 1997).
  • Angiotensin (1-7) peptides may be prepared in recombinant systems using, for example, polynucleotide sequences encoding the polypeptides. It is understood that a polypeptide may contain more than one of the above-described modifications within the same polypeptide.
  • peptides may be effective in eliciting a biological activity in vitro, their effectiveness in vivo might be reduced by the presence of proteases.
  • Serum proteases have specific substrate requirements. The substrate must have both L-amino acids and peptide bonds for cleavage.
  • exopeptidases which represent the most prominent component of the protease activity in serum, usually act on the first peptide bond of the peptide and require a free N-terminus (Powell et al, Pharm. Res. 10: 1268-1273 (1993)).
  • modified versions of peptides The modified peptides retain the structural characteristics of the original L-amino acid peptides that confer the desired biological activity of Ang-(l-7) but are advantageously not readily susceptible to cleavage by protease and/or exopeptidases.
  • D-amino acid of the same type may be used to generate more stable peptides.
  • a peptide derivative or peptidomimetic of the present invention may be all L, all D or mixed D, L peptide, in either forward or reverse order.
  • the presence of an N-terminal or C-terminal D-amino acid increases the in vivo stability of a peptide since peptidases cannot utilize a D-amino acid as a substrate (Powell et al., Pharm. Res. 10:1268-1273 (1993)).
  • Reverse- D peptides are peptides containing D-amino acids, arranged in a reverse sequence relative to a peptide containing L-amino acids.
  • the C-terminal residue of an L-amino acid peptide becomes N-terminal for the D-amino acid peptide, and so forth.
  • Reverse D-peptides retain the same secondary conformation and therefore similar activity, as the L-amino acid peptides, but are more resistant to enzymatic degradation in vitro and in vivo, and thus can have greater therapeutic efficacy than the original peptide (Brady and Dodson, Nature 368:692-693 (1994); Jameson et al, Nature 368:744-746 (1994)).
  • a reverse-L peptide may be generated using standard methods where the C-terminus of the parent peptide becomes takes the place of the N-terminus of the reverse-L peptide. It is contemplated that reverse L-peptides of L-amino acid peptides that do not have significant secondary structure (e.g., short peptides) retain the same spacing and conformation of the side chains of the L-amino acid peptide and therefore often have the similar activity as the original L-amino acid peptide. Moreover, a reverse peptide may contain a combination of L- and D-amino acids. The spacing between amino acids and the conformation of the side chains may be retained resulting in similar activity as the original L- amino acid peptide.
  • Another effective approach to confer resistance to peptidases acting on the N- terminal or C-terminal residues of a peptide is to add chemical groups at the peptide termini, such that the modified peptide is no longer a substrate for the peptidase.
  • One such chemical modification is glycosylation of the peptides at either or both termini.
  • Certain chemical modifications, in particular N-terminal glycosylation, have been shown to increase the stability of peptides in human serum (Powell et al., Pharm. Res. 10: 1268-1273 (1993)).
  • N- terminal alkyl group consisting of a lower alkyl of from one to twenty carbons, such as an acetyl group, and/or the addition of a C-terminal amide or substituted amide group.
  • the present invention includes modified peptides consisting of peptides bearing an N-terminal acetyl group and/or a C-terminal amide group.
  • Substitution of non-naturally-occurring amino acids for natural amino acids in a subsequence of the peptides can also confer resistance to proteolysis. Such a substitution can, for instance, confer resistance to proteolysis by exopeptidases acting on the N-terminus without affecting biological activity.
  • non-naturally-occurring amino acids include ⁇ , ⁇ - disubstituted amino acids, N-alkyl amino acids, C-a-methyl amino acids, ⁇ -amino acids, and ⁇ - methyl amino acids.
  • Amino acids analogs useful in the present invention may include, but are not limited to, ⁇ -alanine, norvaline, norleucine, 4-aminobutyric acid, orithine, hydroxyproline, sarcosine, citrulline, cysteic acid, cyclohexylalanine, 2-aminoisobutyric acid, 6-aminohexanoic acid, t-butylglycine, phenylglycine, o-phosphoserine, N-acetyl serine, N-formylmethionine, 3- methylhistidine and other unconventional amino acids. Furthermore, the synthesis of peptides with non-naturally-occurring amino acids is routine in the art.
  • constrained peptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods well known in the art (Rizo and Gierasch, Ann. Rev. Biochem. 61 :387-418 (1992)).
  • constrained peptides may be generated by adding cysteine residues capable of forming disulfide bridges and, thereby, resulting in a cyclic peptide.
  • Cyclic peptides can be constructed to have no free N- or C-termini. Accordingly, they are not susceptible to proteolysis by exopeptidases, although they may be susceptible to endopeptidases, which do not cleave at peptide termini.
  • amino acid sequences of the peptides with N-terminal or C-terminal D-amino acids and of the cyclic peptides are usually identical to the sequences of the peptides to which they correspond, except for the presence of N-terminal or C-terminal D-amino acid residue, or their circular structure, respectively.
  • a functional equivalent, analogue or derivative of naturally-occurring Ang-(l-7) is a cyclic peptide.
  • a cyclic peptide has an intramolecular covalent bond between two non-adjacent residues.
  • the intramolecular bond may be a backbone to backbone, side-chain to backbone or side-chain to side-chain bond (i.e., terminal functional groups of a linear peptide and/or side-chain functional groups of a terminal or interior residue may be linked to achieve cyclization).
  • Typical intramolecular bonds include disulfide, amide and thioether bonds.
  • Cyclic peptides as described herein may comprise residues of L-amino acids, D- amino acids, or any combination thereof.
  • Amino acids may be from natural or non-natural sources, provided that at least one amino group and at least one carboxyl group are present in the molecule; a- and ⁇ -amino acids are generally preferred.
  • Cyclic peptides may also contain one or more rare amino acids (such as 4-hydroxyproline or hydroxy lysine), organic acids or amides and/or derivatives of common amino acids, such as amino acids having the C-terminal carboxylate esterified (e.g., benzyl, methyl or ethyl ester) or amidated and/or having
  • N-terminal amino group e.g., acetylation or alkoxycarbonylation
  • side-chain modifications and/or substitutions e.g., methylation, benzylation, t-butylation, tosylation, alkoxycarbonylation, and the like.
  • Suitable derivatives include amino acids having an N-acetyl group (such that the amino group that represents the N- terminus of the linear peptide prior to cyclization is acetylated) and/or a C-terminal amide group (i.e., the carboxy terminus of the linear peptide prior to cyclization is amidated).
  • Residues other than common amino acids that may be present with a cyclic peptide include, but are not limited to, penicillamine, ⁇ , ⁇ -tetramethylene cysteine, ⁇ , ⁇ -pentamethylene cysteine, ⁇ - mercaptopropionic acid, ⁇ , ⁇ -pentamethylene- ⁇ -mercaptopropionic acid, 2-mercaptobenzene, 2- mercaptoaniline, 2-mercaptoproline, ornithine, diaminobutyric acid, a-aminoadipic acid, m- aminomethylbenzoic acid and ⁇ , ⁇ -diaminopropionic acid.
  • cyclization may be achieved by any of a variety of techniques well known in the art.
  • a bond may be generated between reactive amino acid side chains.
  • a disulfide bridge may be formed from a linear peptide comprising two thiol-containing residues by oxidizing the peptide using any of a variety of methods.
  • air oxidation of thiols can generate disulfide linkages over a period of several days using either basic or neutral aqueous media.
  • the peptide is used in high dilution to minimize aggregation and intermolecular side reactions.
  • cyclization may be achieved by amide bond formation.
  • a peptide bond may be formed between terminal functional groups (i.e., the amino and carboxy termini of a linear peptide prior to cyclization).
  • the linear peptide comprises a D-amino acid.
  • cyclization may be accomplished by linking one terminus and a residue side chain or using two side chains, with or without an N-terminal acetyl group and/or a C-terminal amide.
  • Residues capable of forming a lactam bond include lysine, ornithine (Orn), a-amino adipic acid, m-aminomethylbenzoic acid, ⁇ , ⁇ -diaminopropionic acid, glutamate or aspartate. Methods for forming amide bonds are generally well known in the art.
  • carbodiimide -mediated lactam formation can be accomplished by reaction of the carboxylic acid with DCC, DIC, ED AC or DCCI, resulting in the formation of an O-acylurea that can be reacted immediately with the free amino group to complete the cyclization.
  • cyclization can be performed using the azide method, in which a reactive azide intermediate is generated from an alkyl ester via a hydrazide.
  • cyclization can be accomplished using activated esters. The presence of electron withdrawing substituents on the alkoxy carbon of esters increases their susceptibility to aminolysis.
  • a thioether linkage may be formed between the side chain of a thiol-containing residue and an appropriately derivatized a-amino acid.
  • a lysine side chain can be coupled to bromoacetic acid through the carbodiimide coupling method (DCC, ED AC) and then reacted with the side chain of any of the thiol containing residues mentioned above to form a thioether linkage.
  • DCC carbodiimide coupling method
  • ED AC carbodiimide coupling method
  • any two thiol containing side-chains can be reacted with dibromoethane and diisopropylamine in DMF.
  • the invention provides linear angiotensin-(l-7) peptides.
  • the structure of naturally-occurring Ang-(l-7) is as follows:
  • peptides and peptide analogs of the invention can be generally represented by the following sequence:
  • Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa ⁇ Xaa 7 (SEQ ID NO:5), or a pharmaceutically acceptable salt thereof.
  • Xaa 1 is any amino acid or a dicarboxylic acid. In certain embodiments, Xaa 1 is
  • Xaa 1 is a negatively-charged amino acid, such as Asp or Glu, typically Asp.
  • Xaa 2 is Arg, Lys, Ala, Cit (citrulline), Orn (ornithine), acetylated Ser, Sar, D-Arg and D-Lys.
  • Xaa 2 is a positively-charged amino acid such as Arg or Lys, typically Arg.
  • Xaa 3 is Val, Ala, Leu, Nle (norleucine), He, Gly, Lys, Pro, HydroxyPro
  • Xaa 3 is an aliphatic amino acid such as Val, Leu, He or Nle, typically Val or Nle.
  • Xaa 4 is Tyr, Tyr(P0 3 ), Thr, Ser, homoSer (homoserine), azaTyr (aza-c ⁇ -homo-L- tyrosine) or Ala.
  • Xaa 4 is a hydroxyl-substituted amino acid such as Tyr, Ser or Thr, typically Tyr.
  • Xaa 5 is He, Ala, Leu, norLeu, Val or Gly.
  • Xaa 5 is an aliphatic amino acid such as Val, Leu, He or Nle, typically He.
  • Xaa 6 is His, Arg or 6-NH 2 -Phe (6-aminophenylalanine). In certain embodiments,
  • Xaa 6 is a fully or partially positively-charged amino acid such as Arg or His.
  • Xaa 7 is Cys, Pro or Ala.
  • one or more of Xaa ⁇ Xaa 7 is identical to the
  • all but one or two of Xaa 1 -Xaa 7 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7). In other embodiments, all of Xaa x -Xaa 6 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7).
  • Xaa 3 is Nle.
  • Xaa 3 is Nle, one or more of Xaa 1 -
  • Xaa 2 and Xaa 4"7 are optionally identical to the corresponding amino acid in naturally-occurring Ang-(l-7). In certain such embodiments, all but one or two of Xaa 1 -Xaa 2 and Xaa 4"7 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7). In other words,
  • all of Xaa*-Xaa 2 and Xaa 4"7 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7), resulting in the amino acid sequence: Asp 1 -Arg 2 -Nle 3 -Tyr 4 -Ile 5 - His 6 -Pro 7 (SEQ ID NO:2).
  • the peptide has the amino acid sequence Asp 1 -Arg 2 -Val 3 -
  • Ser 4 -Ile 5 -His 6 -Cys 7 (SEQ ID NO:3) or Asp 1 -Arg 2 -Val 3 -ser 4 -Ile 5 -His 6 -Cys7 (SEQ ID NO:6).
  • the invention provides a cyclic angiotensin-(l-7) (Ang-(l-7)) peptide analog comprising a linkage, such as between the side chains of amino acids
  • peptide analogs typically comprise 7 amino acid residues, but can also include a cleavable sequence.
  • the invention includes fragments and analogs where one or more amino acids are substituted by another amino acid (including fragments), for example, Asp 1 -Arg 2 -Val 3 -Ser 4 -Ile 5 - His 6 -Cys 7 (SEQ ID NO:22), wherein a linkage is formed between Ser 4 and Cys 7 .
  • a thioether bridge is also referred to as a monosulfide bridge or, in the case of Ala-S- Ala, as a lanthionine bridge.
  • Thioether bridge-containing peptides can be formed by two amino acids having one of the following formulas:
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are independently -H, an alkyl (e.g.,
  • R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are each independently -H or -CH 3 , such where all are -H.
  • the invention provides an Ang analog or derivative comprising a thioether bridge according to formula (I).
  • R 1 , R 2 , R 3 and R 4 are independently selected from -H and -CH 3 .
  • Peptides comprising a thioether bridge according to formula (I) can be produced, for example, by lantibiotic enzymes or by sulfur extrusion of a disulfide.
  • the disulfide from which the sulfur is extruded can be formed by D- cysteine in position 4 and L-cysteine in position 7 or by D-cysteine in position 4 and L- penicillamine in position 7 (see, e.g., Gaisme, Trent and Spatola (2003) Biopolymers 71, 534- 551).
  • the linkage of the two amino acids can be the bridges depicted in Formula (II) or Formula (III).
  • Peptides comprising a thioether bridge according to Formula (II) can be made, for example, by sulfur extrusion of a disulfide formed by D- homocysteine in position 4 and L-cysteine in position 7.
  • peptides comprising a thioether bridge as in Formula (III) can be made, for example, by sulfur extrusion of a disulfide formed by D-cysteine in position 4 and L-homocysteine in position 7.
  • the Ang analogs and derivatives of the invention vary in length and amino acid composition.
  • the Ang analogs and derivatives of the invention preferably have biological activity or are an inactive precursor molecule that can be proteolytically activated (such as how angiotensin (I), with 10 amino acids, is converted to active fragments by cleavage of 2 amino acids).
  • the size of an Ang analog or derivative can vary but is typically between from about 5 to 10 amino acids, as long as the "core" pentameric segment comprising the 3-7 Nle-thioether-ring structure is encompassed.
  • the amino acid sequence of an analog or derivative of the invention can vary, typically provided that it is biologically active or can become proteolytically activated.
  • Bioactivity of an analog or derivative can be determined using methods known in the art, including radioligand binding studies, in vitro cell activation assays and in vivo experiments. See, for example, Godeny and Sayeski, (2006) Am. J. Physiol. Cell. Physiol. 291 :0297-1307; Sarr et al, Cardiovasc. Res. (2006) 71 :794-802; and Koziarz et ah, (1933) Gen. Pharmacol. 24:705- 713.
  • Ang analogs and derivatives where only the length of the peptide is varied include the following: a 4,7-cyclized analog designated [Cyc 4_7 ]Ang-(l-7), which is derived from natural Ang- (1-7) (Asp ⁇ Arg ⁇ Val ⁇ Cyc ⁇ Ile ⁇ His ⁇ Cyc 7 , SEQ ID NO:7).
  • the amino acids at positions 4 and 7 of the Cyc 4"7 analog are modified to allow introduction of the thioether-ring structures shown above.
  • the amino acids at positions other than 3, 4 and 7 can be the same or different from the naturally-occurring peptide, typically provided that the analog retains a biological function.
  • One example is Asp 1 - Arg 2 -Val 3 -Ser 4 -Ile 5 -His 6 -Cys 7 (SEQ ID NO:22).
  • analogs of inactive precursors like
  • Ang-(l-10) biological function refers to one or both of an analog's susceptibility to angiotensin-converting enzymes that can cleave it to a biologically active fragment (e.g. Ang-(1- 8) or Ang-(l-7)) or the biological activity of the fragment itself.
  • a biologically active fragment e.g. Ang-(1- 8) or Ang-(l-7)
  • an Ang analog or derivative of the invention has no intrinsic function but inhibits the effects of one or more naturally-occurring angiotensin compounds.
  • an Ang analog of the invention is represented by
  • Xaa 1 is any amino acid, but typically a negatively-charged amino acid such as Glu or Asp, more typically Asp.
  • Xaa 2 is a positively-charged amino acid such as Arg or Lys, typically Arg.
  • Xaa 3 is an aliphatic amino acid, such as Leu, He or Val, typically Val.
  • Cyc 4 forms a thioether bridge in conjunction with Cyc 7 .
  • Cyc 4 can be a D- stereoisomer and/or a L-stereoisomer, typically a D-stereoisomer.
  • Examples of Cyc 4 (taken with Cyc 7 ) are shown in Formulas (I), (II) and (III).
  • the R groups in Formulae (I), (II) and (III) are -H or -CH 3 , especially -H.
  • Xaa 5 is an aliphatic amino acid, such as Leu, He or Val, typically He.
  • Xaa 6 is His.
  • Cyc 7 forms a thioether bridge in conjunction with Cyc 4 , such as in Formula (I),
  • Cyc 7 can be a D-stereoisomer and/or a L-stereoisomer, typically a L-stereoisomer. Examples of Cyc 7 (taken with Cyc 4 ) are shown in Formulas (I), (II) and (III). Typically, the R groups in Formulas (I), (II) and (III) are -H or -CH 3 , especially -H.
  • one or more of Xaa ⁇ Xaa 6 (excluding Cyc 4 and Cyc 7 ) is identical to the corresponding amino acid in naturally-occurring Ang-(l-7). In certain such embodiments, all but one or two of Xaa ⁇ Xaa 6 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7). In other embodiments, all of Xaa ⁇ Xaa 6 are identical to the corresponding amino acid in naturally-occurring Ang-(l-7).
  • Cyc 4 and Cyc 7 are independently selected from Abu (2- aminobutyric acid) and Ala (alanine), where Ala is present in at least one position.
  • cyclic analogs can have a thioether linkage formed by -Ala 4 -S-Ala 7 - (Formula (I), where R -R 4 are each -H); -Ala 4 -S-Abu 7 - (Formula (I): R l -R 3 are -H and R 4 is -CH 3 ) or -Abu 4 -S-Ala 7 - (Formula (I): R 1 , R 3 and R 4 are -H and R 2 is -CH 3 ).
  • Specific examples of cyclic analogs comprise a -Abu 4 -S- Ala 7 - or -Ala 4 -S-Ala 7 - linkage.
  • the invention provides an Ang-(l-7) analog with a thioether-bridge between position 4 and position 7 having the amino acid sequence Asp ⁇ Arg 2 - Val 3 -Abu 4 -Ile 5 -His 6 -Ala 7 (SEQ ID NO: 15) or the amino acid sequence Asp 1 -Arg 2 -Val 3 -Ala 4 -Ile 5 - His 6 -Ala 7 (SEQ ID NO: 16), which are represented by the following structural diagrams:
  • an Ang analog or derivative of the invention is represented by Formula (V):
  • Xaa 1 , Xaa 2 , Xaa 8 , Xaa 9 and Xaa 10 are absent in certain embodiments.
  • (1) Xaa 10 is absent, (2) Xaa 9 and Xaa 10 are absent, (3) Xaa 8 , Xaa 9 and Xaa 10 are absent, (4) Xaa 1 is absent, (5) Xaa 1 and Xaa 10 are absent, (6) Xaa 1 , Xaa 9 and Xaa 10 are absent, (7) Xaa 1 , Xaa 8 , Xaa 9 and Xaa 10 are absent, (8) Xaa 1 and Xaa 2 are absent, (9) Xaa 1 ,
  • Xaa 2 and Xaa 10 are absent, (10) Xaa 1 , Xaa2 , Xaa 9 and Xaa 10 are absent, or (11) Xaa 1 , Xaa2 , Xaa 8 , Xaa 9 and Xaa 10 are absent.
  • the remaining amino acids have the values described below.
  • Xaa 1 when present, is any amino acid, but typically a negatively charged amino acid such as Glu or Asp, more typically Asp.
  • Xaa 2 when present, is a positively charged amino acid such as Arg or Lys, typically Arg.
  • Nle 3 is norleucine.
  • Cyc 4 forms a thioether bridge in conjunction with Cyc 7 .
  • Cyc 4 can be a D- stereoisomer and/or a L-stereoisomer, typically a D-stereoisomer.
  • Examples of Cyc 4 (taken with Cyc 7 ) are shown in Formulas (I), (II) and (III).
  • the R groups in Formulae (I), (II) and (III) are -H or -CH 3 , especially -H.
  • Xaa 5 is an aliphatic amino acid, such as Leu, Nle, He or Val, typically He.
  • Xaa 6 is His.
  • Cyc 7 forms a thioether bridge in conjunction with Cyc 4 , such as in Formula (I),
  • Cyc 7 can be a D-stereoisomer and/or a L-stereoisomer, typically a L-stereoisomer. Examples of Cyc 7 (taken with Cyc 4 ) are shown in Formulas (I), (II) and (III). Typically, the R groups in Formulae (I), (II) and (III) are -H or -CH 3 , especially -H.
  • Xaa 8 when present, is an amino acid other than Pro, typically Phe or He. In certain embodiments, He results in an inhibitor of Ang(l-8). In certain embodiments, Phe maintains the biological activity of Ang(l-8) or Ang(l-lO).
  • Xaa 9 when present, is His.
  • Xaa 10 when present, is an aliphatic residue, for example, He, Val or Leu, typically
  • one or more of Xaa ⁇ Xaa 10 (excluding Nle 3 , Cyc 4 and
  • Cyc 7 is identical to the corresponding amino acid in naturally-occurring Ang (including Ang-(1- 7), Ang(l-8), Ang(l-9), Ang(l-lO), Ang(2-7), Ang(2-8), Ang(2-9), Ang(2-10), Ang(3-8), Ang(3- 9) and Ang(3-10).
  • all but one or two of Xaa ⁇ Xaa 10 are identical to the corresponding amino acid in naturally-occurring Ang.
  • all of Xaa ⁇ Xaa 10 are identical to the corresponding amino acid in naturally-occurring Ang.
  • Cyc 4 and Cyc 7 are independently selected from Abu (2- aminobutyric acid) and Ala (alanine), where Ala is present at at least one position.
  • cyclic analogs comprising a thioether linkage formed by -Ala 4 -S-Ala 7 - (Formula (I), where R -R 4 are each -H); -Ala 4 -S-Abu 7 - (Formula (I): R J -R 3 are -H and R 4 is -CH 3 ) or -Abu 4 -S-Ala 7 - (Formula (I): R 1 , R 3 and R 4 are -H and R 2 is -CH 3 ).
  • Specific cyclic analogs comprise a -Abu 4 -S-Ala 7 - or -Ala 4 -S-Ala 7 - linkage.
  • the invention provides an Ang-(l-7) analog or derivative with a thioether-bridge between position 4 and position 7 having the amino acid sequence Asp ⁇ Arg 2 - Nle 3 -Abu 4 -Ile 5 -His 6 -Ala 7 (SEQ ID NO: 18) or the amino acid sequence Asp ⁇ Arg ⁇ Nle ⁇ Ala ⁇ Ile 5 - His 6 -Ala 7 (SEQ ID NO: 19).
  • the invention provides an Ang-(l-8) analog or derivative with a thioether-bridge between position 4 and position 7 having Ang-(l-8) antagonistic activity, in particular an Ang(l-8) analog or derivative having the amino acid sequence Asp ⁇ Arg ⁇ Nle 3 - Abu 4 -Ile 5 -His 6 -Ala 7 -Ile 8 (SEQ ID NO:20) or the amino acid sequence Asp ⁇ Arg ⁇ Nle ⁇ Ala ⁇ Ile 5 - His 6 -Ala 7 -Ile 8 (SEQ ID NO:21).
  • the present invention provides methods of treating GVHD including administering to a subject who is suffering from or susceptible to GVHD an angiotensin (1-7) receptor agonist.
  • angiotensin-(l-7) receptor agonist encompasses any molecule that has a positive impact in a function of an angiotensin-(l-7) receptor, in particular, the G-protein coupled Mas receptor.
  • an angiotensin-(l-7) receptor agonist directly or indirectly enhances, strengthens, activates and/or increases an angiotensin-(l-7) receptor (i.e., the Mas receptor) activity.
  • an angiotensin-(l-7) receptor agonist directly interacts with an angiotensin-(l-7) receptor (i.e., the Mas receptor).
  • angiotensin-(l-7) receptor i.e., the Mas receptor.
  • Such agonists can be peptidic or non-peptidic including, e.g., proteins, chemical compounds, small molecules, nucleic acids, antibodies, drugs, ligands, or other agents.
  • the angiotensin (1-7) receptor agonist is a non-peptidic agonist.
  • an exemplary class of angiotensin-(l-7) receptor agonists are l-(p- thienylbenzyl)imidazoles. Examples of these non-peptide angiotensin-(l-7) receptor agonists are represented by Structural Formula (VI):
  • R 1 is halogen, hydroxyl, (Ci-C4)-alkoxy, (Ci-Cg)-alkoxy wherein 1 to 6 carbon atoms are replaced by the heteroatoms O, S, or NH (preferably by O), (Ci-C 4 )-alkoxy substituted by a saturated cyclic ether such as tetrahydropyran or tetrahydrofuran, 0-(Ci-C 4 )-alkenyl, 0-(Ci-C 4 )- alkylaryl, or aryloxy that is unsubstituted or substituted by a substituent selected from halogen, (Ci-C3)-alkyl, (Ci-C3)-alkoxy and trifluoromethyl;
  • R 2 is CHO, COOH, or (3) CO-0-(Ci-C 4 )-alkyl
  • R 3 is (Ci-C 4 )-alkyl or aryl
  • R 4 is hydrogen, halogen (chloro, bromo, fluoro), or (Ci-C 4 )-alkyl;
  • X is oxygen or sulfur;
  • Y is oxygen or -NH-;
  • R 5 is hydrogen, (Ci-C 6 )-alkyl; or (Ci-C 4 )-alkylaryl, where R 5 is hydrogen when Y is -NH-;
  • R 6 is (Ci-Cs)-alkyl.
  • R 1 is not halogen when R 2 is COOH or CO-0-(Ci-C 4 )- alkyl.
  • an angiotensin-(l-7) receptor agonist is AVE 0991, 5- formyl-4-methoxy-2 -phenyl- 1 [ [4- [2-(ethylaminocarbonylsulfonamido)-5 -isobutyl-3 -thienyl]- phenyl]-methyl]-imidazole, which is represented by the following structure:
  • angiotensin-(l-7) receptor agonists are p- thienylbenzylamides. Examples of these non-peptide angiotensin-(l-7) receptor agonists are represented by Structural Formula (VII):
  • R 1 is (Ci-C 5 )-alkyl that is unsubstituted or substituted by a radical chosen from NH 2 , halogen, 0-(Ci-C 3 )-alkyl, CO-0-(Ci-C 3 )-alkyl and C0 2 H, (C 3 -C 8 )-cycloalkyl, (Ci-C 3 )-alkyl-(C 3 - C 8 )-cycloalkyl, (C 6 -Ci 0 )-aryl that is unsubstituted or substituted by a radical chosen from halogen and 0-(Ci-C 3 )-alkyl, (Ci-C 3 )-alkyl-(C 6 -Cio)-aryl where the aryl radical is unsubstituted or substituted by a radical chosen from halogen and 0-(Ci-C 3 )-alkyl, (Ci-Cs)-heteroaryl, or (
  • R 2 is hydrogen, (Ci-C 6 )-alkyl that is unsubstituted or substituted by a radical chosen from halogen and 0-(Ci-C 3 )-alkyl, (C 3 -C 8 )-cycloalkyl, (Ci-C 3 )-alkyl-(C 3 -C 8 )-cycloalkyl, (C 6 -Ci 0 )-aryl that is unsubstituted or substituted by a radical chosen from among halogen, 0-(Ci-C 3 )-alkyl and CO-0-(Ci-C 3 )-alkyl, or (Ci-C 3 )-alkyl-(C 6 -Cio)-aryl that is unsubstituted or substituted by a radical chosen from halogen and 0-(Ci-C 3 )-alkyl;
  • R 3 is hydrogen, COOH, or COO-(Ci-C 4 )-alkyl
  • R 4 is hydrogen, halogen; or (Ci-C 4 )-alkyl
  • R 5 is hydrogen or (Ci-C 6 )-alkyl
  • R 6 is hydrogen, (Ci-C 6 )-alkyl, (Ci-C 3 )-alkyl-(C 3 -C 8 )-cycloalkyl, or (C 2 -C 6 )-alkenyl; and X is oxygen or NH.
  • angiotensin-(l-7) receptor agonists described above can be present as pharmaceutically acceptable salts.
  • a pharmaceutically acceptable salt refers to salts that retain the desired activity of the peptide or equivalent compound, but preferably do not detrimentally affect the activity of the peptide or other component of a system, which uses the peptide.
  • examples of such salts are acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like.
  • Salts may also be formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, and the like.
  • Salts formed from a cationic material may utilize the conjugate base of these inorganic and organic acids.
  • Salts may also be formed with polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel and the like or with an organic cation formed from ⁇ , ⁇ '- dibenzylethylenediamme or ethylenediamine, or combinations thereof (e.g., a zinc tannate salt).
  • polyvalent metal cations such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel and the like or with an organic cation formed from ⁇ , ⁇ '- dibenzylethylenediamme or ethylenediamine, or combinations thereof (e.g., a zinc tannate salt).
  • organic cation formed from ⁇ , ⁇ '- dibenzylethylenediamme or ethylenediamine, or combinations thereof (e.g., a zinc tannate salt).
  • the non-toxic, physiologically acceptable salts are preferred.
  • the salts can be formed by conventional means such as by reacting the free acid or free base forms of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble, or in a solvent such as water which is then removed in vacuo or by freeze-drying, or by exchanging the cations of an existing salt for another cation on a suitable ion exchange resin.
  • An alkyl group is a straight chained or branched non-aromatic hydrocarbon that is completely saturated. Typically, a straight chained or branched alkyl group has from 1 to about 20 carbon atoms, preferably from 1 to about 10. Examples of straight chained and branched alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl and octyl.
  • a C1-C4 straight chained or branched alkyl group is also referred to as a "lower alkyl" group.
  • An alkenyl group is a straight chained or branched non-aromatic hydrocarbon that is includes one or more double bonds. Typically, a straight chained or branched alkenyl group has from 2 to about 20 carbon atoms, preferably from 2 to about 10. Examples of straight chained and branched alkenyl groups include ethenyl, n-propenyl, and n-butenyl.
  • Aromatic (aryl) groups include carbocyclic aromatic groups such as phenyl, naphthyl, and anthracyl, and heteroaryl groups such as imidazolyl, thienyl, furyl, pyridyl, pyrimidyl, pyranyl, pyrazolyl, pyrrolyl, pyrazinyl, thiazolyl, oxazolyl, and tetrazolyl.
  • Aromatic groups also include fused polycyclic aromatic ring systems in which a carbocyclic aromatic ring or heteroaryl ring is fused to one or more other heteroaryl rings.
  • Examples include benzothienyl, benzofuryl, indolyl, quinolinyl, benzothiazole, benzoxazole, benzimidazole, quinolinyl, isoquinolinyl and isoindolyl.
  • Aromatic (aryl) groups include carbocyclic aromatic groups such as phenyl, naphthyl, and anthracyl, and heteroaryl groups such as imidazolyl, thienyl, furyl, pyridyl, pyrimidyl, pyranyl, pyrazolyl, pyrrolyl, pyrazinyl, thiazolyl, oxazolyl, and tetrazolyl.
  • Aromatic groups also include fused polycyclic aromatic ring systems in which a carbocyclic aromatic ring or heteroaryl ring is fused to one or more other heteroaryl rings.
  • Examples include benzothienyl, benzofuryl, indolyl, quinolinyl, benzothiazole, benzoxazole, benzimidazole, quinolinyl, isoquinolinyl and isoindolyl.
  • the present invention provides methods and compositions for treatment of
  • GVHD GVHD
  • treatment with an angiotensin (1-7) peptide described herein can reduce the severity of GVHD, such as by reducing a stage of severity of GVHD.
  • treatment of GVHD with an angiotensin (1-7) peptide described herein reduces GVHD by one, two, three, or four stages (e.g., stage 4 to stage 3, 2, 1, or 0) as compared to a control.
  • treatment with an angiotensin (1-7) peptide results in substantially free of Stage 4, 3, 2, or 1 acute GVHD in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • treatment with an angiotensin (1-7) peptide results in results in substantially free of Grade IV, III, II, or I acute GVHD in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • treatment with an angiotensin (1-7) peptide results in substantially free of acute GVHD symptom in the subject 100 days (e.g., at least 120 days, 130 days, 140 days, 150 days, 160 days, 170 days, 180 days or more) following transplantation.
  • treatment with an angiotensin (1-7) peptide results in substantially free of chronic GVHD symptoms in the subject at least 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 1.5 years, 2 years, 2.5 years, 3 years, 4 years, 5 years or more following transplantation.
  • the present invention may be used as a monotherapy or as part of a combination therapy with one or more other prophylactic or therapeutic materials routinely used with transplantation or used to treat or reduce risk of GVHD.
  • the present invention comprises a method of treating or reducing the risk of GVHD by administering to a subject who is suffering from or susceptible to GVHD an angiotensin (1-7) peptide described herein.
  • a subject is any multicellular organism.
  • a subject is a mouse, rat, dog, non-human primate or other animal commonly used for laboratory experiments.
  • a subject is an individual.
  • an individual is a human.
  • a subject has or is susceptible to a disease, disorder, or condition.
  • a subject has or is susceptible to
  • a subject who is susceptible to GVHD comprises a subject who has had a transplantation. In some embodiments, a subject who is susceptible to GVHD comprises a subject who is going to have a transplantation. In some embodiments, a subject who is susceptible to GVHD comprises a subject having a disease, disorder, or condition that may require a transplantation. In some embodiments, a subject who is susceptible to GVHD comprises a subject at risk for a disease, disorder, or condition that may require a transplantation. In some embodiments, a transplantation is a cord blood transplantation. In some embodiments, a transplantation is a double cord blood transplantation. In some embodiments, a transplantation is a bone marrow transplantation. In some embodiments, a transplantation is an adult stem cell transplantation. In some embodiments, a transplantation is an embryonic stem cell
  • a transplantation is an organ transplantation. In some embodiments, a transplantation is a blood transfusion.
  • an angiotensin (1-7) peptide described herein is administered to a subject having or at risk of GVHD at an effective dose periodically at an administration interval such that at least one symptom or feature of GVHD is reduced in intensity, severity, duration, or frequency or has delayed in onset.
  • an angiotensin (1-7) peptide described herein of the invention can be administered to a subject alone (e.g., as a purified peptide), or as a component of a composition or medicament ⁇ e.g., in the manufacture of a medicament for the treatment of the disease), as described herein.
  • the compositions can be formulated with a physiologically acceptable carrier or excipient to prepare a pharmaceutical composition.
  • the carrier and composition can be sterile.
  • the formulation should suit the mode of administration. Methods of formulating compositions are known in the art (see, e.g.,
  • Suitable pharmaceutically acceptable carriers include but are not limited to water, salt solutions ⁇ e.g., NaCl), saline, buffered saline, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, sugars such as mannitol, sucrose, or others, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc. , as well as combinations thereof.
  • the pharmaceutical preparations can, if desired, be mixed with auxiliary agents ⁇ e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like) which do not deleteriously react with the active compounds or interference with their activity.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like
  • a water-soluble carrier suitable for intravenous administration is used.
  • composition or medicament can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • the composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder.
  • the composition can also be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, polyvinyl pyrollidone, sodium saccharine, cellulose, magnesium carbonate, etc.
  • composition or medicament can be formulated in accordance with the routine procedures as a pharmaceutical composition adapted for administration to human beings.
  • a composition for intravenous administration typically is a solution in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water, saline or dextrose/water.
  • an ampule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions comprising those provided as pharmaceutical formulations, comprise a liquid carrier such as but not limited to water, saline, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solutions, Hank's solution, other aqueous physiologically balanced solutions, oils, esters and glycols.
  • a liquid carrier such as but not limited to water, saline, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solutions, Hank's solution, other aqueous physiologically balanced solutions, oils, esters and glycols.
  • An angiotensin (1-7) peptide and/or angiotensin (1-7) receptor agonist as described herein can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include, but are not limited to, those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • an angiotensin (1-7) peptide described herein (or a composition or medicament containing an angiotensin (1-7) peptide described herein) is administered by any appropriate route.
  • an angiotensin (1-7) peptide described herein is administered subcutaneously.
  • the term "subcutaneous tissue”, is defined as a layer of loose, irregular connective tissue immediately beneath the skin.
  • the subcutaneous administration may be performed by injecting a composition into areas including, but not limited to, thigh region, abdominal region, gluteal region, or scapular region.
  • an angiotensin (1-7) peptide described herein is administered intravenously.
  • an angiotensin (1-7) peptide described herein is administered orally.
  • an angiotensin (1-7) peptide described herein is administered by direct administration to a target tissue, such as heart or muscle ⁇ e.g., intramuscular), tumor (intratumorallly), nervous system ⁇ e.g., direct injection into the brain; intra ventricularly; intrathecally).
  • a target tissue such as heart or muscle ⁇ e.g., intramuscular), tumor (intratumorallly), nervous system ⁇ e.g., direct injection into the brain; intra ventricularly; intrathecally).
  • an angiotensin (1-7) peptide described herein (or a composition or medicament containing an angiotensin (1-7) peptide described herein) can be administered by inhalation, parenterally, intradermally, transdermally, or transmucosally ⁇ e.g., orally or nasally). More than one route can be used concurrently, if desired.
  • a composition is administered in a therapeutically effective amount and/or according to a dosing regimen that is correlated with a particular desired outcome (e.g., with treating or reducing risk for GVHD).
  • Particular doses or amounts to be administered in accordance with the present invention may vary, for example, depending on the nature and/or extent of the desired outcome, on particulars of route and/or timing of administration, and/or on one or more characteristics (e.g., weight, age, personal history, genetic characteristic, lifestyle parameter, severity of cardiac defect and/or level of risk of cardiac defect, etc., or combinations thereof). Such doses or amounts can be determined by those of ordinary skill.
  • an appropriate dose or amount is determined in accordance with standard clinical techniques.
  • an appropriate dose or amount is a dose or amount sufficient to reduce severity of GVHD by one, two, three, or four stages.
  • an appropriate dose or amount is determined through use of one or more in vitro or in vivo assays to help identify desirable or optimal dosage ranges or amounts to be
  • angiotensin (1-7) peptides or angiotensin (1-7) receptor agonists, including derivatives, analogs, and/or salts are administered at a therapeutically effective amount.
  • therapeutically effective amount is largely determined based on the total amount of the therapeutic agent contained in the pharmaceutical compositions of the present invention. Generally, a therapeutically effective amount is sufficient to achieve a meaningful benefit to the subject (e.g., treating, modulating, curing, preventing and/or ameliorating the underlying disease or condition).
  • appropriate doses or amounts to be administered may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • a therapeutically effective dosage amount of angiotensin (1-7) peptides or angiotensin (1-7) receptor agonists, including derivatives, analogs, and/or salts can be, for example, between about 0.001-10 mg/kg, about 0.005-1.5 mg/kg, about 0.1-1 mg/kg, or 300-1,000 ⁇ g/kg.
  • the therapeutically effective dosage amount of angiotensin (1-7) peptides or angiotensin (1-7) receptor agonists, including derivatives, analogs, and/or salts can be, for example, about 1 ⁇ g/kg, 2.5 ⁇ /kg, 5 ⁇ /kg, 10 , 20 , 30 , 40 , 50 , 60 , 70 , 80 %, 90 , 100 , 150 , 200 , 250 , 300 , 400 , 500 , 600 ⁇ g/kg, 700 ⁇ g/kg, 800 ⁇ g/kg, 900 ⁇ g/kg, 1000 ⁇ g/kg, or 1500 ⁇ g/kg.
  • the effective dose for a particular individual can be varied ⁇ e.g., increased or decreased) over time, depending on the needs of the individual.
  • Therapeutically effective dosage amounts of angiotensin (1-7) peptides or angiotensin (1-7) receptor agonists, including derivatives, analogs, and/or salts may be present in varying amounts in various embodiments.
  • a therapeutically effective dosage amount can be, for example, about 1-10,000 ⁇ g/kg, about 5-1,500 ⁇ g/kg, about 100- 1 ,000 ⁇ /kg, or 50-500 ⁇ /kg.
  • the therapeutically effective dosage amount can be, for example, about 1 ⁇ g/kg, 2.5 ⁇ g/kg, 5 ⁇ g/kg, 10 ⁇ g/kg, 20 ⁇ g/kg, 30 ⁇ g/kg, 40 ⁇ /kg, 50 ⁇ /kg, 60 ⁇ /kg, 70 , 80 , 90 , 100 , 150 , 200 , 250 ⁇ /kg, 300 ⁇ /kg, 400 , 500 , 600 , 700 , 800 , 900 ⁇ ⁇ ⁇ ⁇ , 1 ,000 ⁇ ⁇ , or 1 ,500 ⁇ /kg.
  • the effective dose for a particular individual can be varied (e.g.,
  • the therapeutically effective amount described herein is provided in one dose. In some embodiments, the therapeutically effective amount described herein is provided in one day.
  • a therapeutically effective dosage amount may be, for example, about 0.001 mg/kg weight to 500 mg/kg weight, e.g., from about 0.001 mg/kg weight to 400 mg/kg weight, from about 0.001 mg/kg weight to 300 mg/kg weight, from about 0.001 mg/kg weight to 200 mg/kg weight, from about 0.001 mg/kg weight to 100 mg/kg weight, from about 0.001 mg/kg weight to 90 mg/kg weight, from about 0.001 mg/kg weight to 80 mg/kg weight, from about 0.001 mg/kg weight to 70 mg/kg weight, from about 0.001 mg/kg weight to 60 mg/kg weight, from about 0.001 mg/kg weight to 50 mg/kg weight, from about 0.001 mg/kg weight to 40 mg/kg weight, from about 0.001 mg/kg weight to 30 mg/kg weight, from about 0.001 mg/kg weight to 25 mg/kg weight, from about 0.001 mg/kg weight to 20 mg/kg weight, from about 0.00 0.001 mg/kg weight to
  • a therapeutically effective dosage amount may be, for example, about 0.001 mg/kg weight to about 1 mg/kg weight, e.g. from about 0.001 mg/kg weight to about 0.9 mg/kg weight, from about 0.001 mg/kg weight to about 0.8 mg/kg weight, from about 0.001 mg/kg weight to about 0.8 mg/kg weight, from about 0.001 mg/kg weight to about 0.7 mg/kg weight, from about 0.001 mg/kg weight to about 0.6 mg/kg weight, from about 0.001 mg/kg weight to about 0.5 mg/kg weight, from about 0.01 mg/kg weight to about 1 mg/kg weight, from about 0.01 mg/kg weight to about 0.9 mg/kg weight, from about 0.01 mg/kg weight to about 0.8 mg/kg weight, from about 0.01 mg/kg weight to about 0.7 mg/kg weight, from about 0.01 mg/kg weight to about 0.6 mg/kg weight, from about 0.01 mg/kg weight to about 0.5 mg/kg weight, from about 0.01 mg/kg weight to about 1
  • a therapeutically effective dosage amount may be, for example, about 0.0001 mg/kg weight to 0.1 mg/kg weight, e.g. from about 0.0001 mg/kg weight to 0.09 mg/kg weight, from about 0.0001 mg/kg weight to 0.08 mg/kg weight, from about 0.0001 mg/kg weight to 0.07 mg/kg weight, from about 0.0001 mg/kg weight to 0.06 mg/kg weight, from about 0.0001 mg/kg weight to 0.05 mg/kg weight, from about 0.0001 mg/kg weight to about 0.04 mg/kg weight, from about 0.0001 mg/kg weight to 0.03 mg/kg weight, from about 0.0001 mg/kg weight to 0.02 mg/kg weight, from about 0.0001 mg/kg weight to 0.019 mg/kg weight, from about 0.0001 mg/kg weight to 0.018 mg/kg weight, from about 0.0001 mg/kg weight to 0.017 mg/kg weight, from about 0.0001 mg/kg weight to 0.016 mg/kg weight, from about
  • the therapeutically effective dose may be 0.0001 mg/kg weight, 0.0002 mg/kg weight, 0.0003 mg/kg weight, 0.0004 mg/kg weight, 0.0005 mg/kg weight, 0.0006 mg/kg weight, 0.0007 mg/kg weight, 0.0008 mg/kg weight, 0.0009 mg/kg weight, 0.001 mg/kg weight, 0.002 mg/kg weight, 0.003 mg/kg weight, 0.004 mg/kg weight, 0.005 mg/kg weight, 0.006 mg/kg weight, 0.007 mg/kg weight, 0.008 mg/kg weight, 0.009 mg/kg weight, 0.01 mg/kg weight, 0.02 mg/kg weight, 0.03 mg/kg weight, 0.04 mg/kg weight, 0.05 mg/kg weight, 0.06 mg/kg weight, 0.07 mg/kg weight, 0.08 mg/kg weight, 0.09 mg/kg weight, or 0.1 mg/kg weight.
  • the effective dose for a particular individual can be varied (e.g., increased or decreased) over time, depending on the needs of the individual.
  • the therapeutically effective amount described herein is provided in one dose. In some embodiments, the therapeutically effective amount described herein is provided in one day.
  • the angiotensin (1-7) peptide is administered at an effective dose ranging from about 1-1 ,000 ⁇ g/kg/day (e.g., ranging from about 1-900 ⁇ g/kg/day, 1-800 ⁇ g/kg/day, 1-700 ⁇ g/kg/day, 1-600 ⁇ g/kg/day, 1-500 ⁇ g/kg/day, 1-400 ⁇ g/kg/day, 1-300 ⁇ g/kg/day, 1-200 ⁇ g/kg/day, 1-100 ⁇ g/kg/day, 1-90 ⁇ g/kg/day, 1-80 ⁇ g/kg/day, 1-70 ⁇ g/kg/day, 1-60 ⁇ g/kg/day, 1-50 ⁇ g/kg/day, 1-40 ⁇ g/kg/day, 1-30 ⁇ g/kg/day, 1-20 ⁇ g/kg/day, 1-10 ⁇ g/kg/day).
  • an effective dose ranging from about 1-1 ,000 ⁇ g/kg/day (e.g., ranging from
  • the angiotensin (1-7) peptide is administered at an effective dose ranging from about 1-500 ⁇ g/kg/day. In some embodiments, the angiotensin (1-7) peptide is administered at an effective dose ranging from about 50-500 ⁇ g/kg/day. In some
  • the angiotensin (1-7) peptide is administered at an effective dose ranging from about 1-100 ⁇ g/kg/day. In some embodiments, the angiotensin (1-7) peptide is administered at an effective dose ranging from about 1-60 ⁇ g/kg/day. In some embodiments, the angiotensin (1- 7) peptide is administered at an effective dose selected from about 1 , 2, 4, 6, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, or 1 ,000 ug/kg/day.
  • a provided composition is provided as a pharmaceutical formulation.
  • a pharmaceutical formulation is or comprises a unit dose amount for administration in accordance with a dosing regimen correlated with achievement of the reduced incidence or risk of GVHD.
  • a formulation comprising an angiotensin (1-7) peptide described herein administered as a single dose.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered at regular intervals. Administration at an "interval,” as used herein, indicates that the therapeutically effective amount is
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered bimonthly, monthly, twice monthly, triweekly, biweekly, weekly, twice weekly, thrice weekly, daily, twice daily, or every six hours.
  • the administration interval for a single individual need not be a fixed interval, but can be varied over time, depending on the needs of the individual.
  • the term “monthly” means administration once per month;
  • the term “triweekly” means administration once per three weeks (i.e., once every three weeks);
  • the term “biweekly” means administration once per two weeks (i.e., once every two weeks);
  • the term “weekly” means administration once per week; and the term “daily” means administration once per day.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered at regular intervals indefinitely. In some embodiments, a formulation comprising angiotensin (1-7) peptide described herein is administered at regular intervals for a defined period. In some embodiments, a formulation comprising an angiotensin (1-7) peptide described herein is administered at regular intervals for 5 years, 4, years, 3, years, 2, years, 1 year, 11 months, 10 months, 9 months, 8 months, 7 months, 6 months, 5 months, 4 months, 3 months, 2 months, a month, 3 weeks, 2, weeks, a week, 6 days, 5 days, 4 days, 3 days, 2 days or a day.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered prior to transplantation.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered for at least 1 year, 11 months, 10 months, 9 months, 8 months, 7 months, 6 months, 5 months, 4 months, 3 months, 2 months, a month, 3 weeks, 2, weeks, a week, 6 days, 5 days, 4 days, 3 days, 2 days or a day prior to transplantation.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered after transplantation.
  • a formulation comprising an angiotensin (1-7) peptide described herein is administered for at least 1 year, 11 months, 10 months, 9 months, 8 months, 7 months, 6 months, 5 months, 4 months, 3 months, 2 months, a month, 3 weeks, 2, weeks, a week, 6 days, 5 days, 4 days, 3 days, 2 days or a day after transplantation.
  • an angiotensin (1-7) peptide described herein is administered concurrently with (i.e., at about the same time as) a transplantation.
  • an angiotensin (1-7) peptide can be administered on the same day as a transplantation.
  • an angiotensin (1-7) peptide is administered in
  • the known therapeutic agent(s) is/are administered according to its standard or approved dosing regimen and/or schedule. In some embodiments, the known therapeutic agent(s) is/are administered according to a regimen that is altered as compared with its standard or approved dosing regimen and/or schedule.
  • an altered regimen differs from the standard or approved dosing regimen in that one or more unit doses is altered (e.g., reduced or increased) in amount, and/or in that dosing is altered in frequency (e.g., in that one or more intervals between unit doses is expanded, resulting in lower frequency, or is reduced, resulting in higher frequency).
  • an angiotensin (1-7) peptide is administered in combination with one or more immunosuppressants.
  • An "immunosuppressant" is any molecule that reduces or eliminates an immune response in a host when the host is challenged with an immunogenic molecule, including immunogenic molecules present on transplanted organs, tissues or cells. Examples of immunosuppressants include but are not limited to antithymocyte globulin (ATG), anti-TNF agents, azathioprine (or other inosine 5 '-monophosphate
  • dehydrogenase inhibitors azodiacarbonide, bisindolyl maleimide VIII, brequinar, chlorambucil, CTLA4-Ig, corticosteroids, cyclophosphamide, cyclosporine A, deoxyspergualin,
  • dexamethasone glucocorticoids, IL-2 antagonists (e.g., daclizumab and basiliximab), leflunomide, mercaptopurine, 6-mercaptopurine (6-MP), methotrexate, methylprednisolone, mizoribine, mizoribine monophosphate, muromonab CD3, mycophenolate mofetil, OKT3, prednisone, sirolimus, rapamycin, rho (D) immune globin, tacrolimus (FK506), vitamin D analogs (e.g., MC1288), etc.
  • IL-2 antagonists e.g., daclizumab and basiliximab
  • leflunomide mercaptopurine
  • 6-MP 6-mercaptopurine
  • methotrexate methylprednisolone
  • mizoribine mizoribine monophosphate
  • muromonab CD3, mycophenolate mofetil OKT3, pred
  • provided compositions comprise growth factors (e.g., filgrastim). In some embodiments, provided compositions comprise DNA nucleoside analogs (e.g., fludarabine). In some embodiments, provided compositions comprise antineoplastic alkylating agents (e.g., melphalan, thiotepa).
  • growth factors e.g., filgrastim
  • DNA nucleoside analogs e.g., fludarabine
  • provided compositions comprise antineoplastic alkylating agents (e.g., melphalan, thiotepa).
  • compositions are administered in combination with one or more conventional treatments for GVHD.
  • Conventional treatments for GVHD includes steroid (e.g., methylprednisolone) therapy.
  • Chronic GVHD is treated with a steroid (e.g., methylprednisolone) therapy.
  • the present invention also provides methods and compositions for treatment of mucositis.
  • the present invention can be used to treat mucositis caused by transplantation (e.g., allogeneic transplantation).
  • mucositis it is meant that the compositions of the present invention may reduce the intensity, severity, duration or frequency, or delay the onset of one or more symptom of mucositis.
  • treatment with an angiotensin (1-7) peptide described herein can reduce the severity of mucositis, such as by reducing a stage or grade of severity of mucositis.
  • treatment with an angiotensin (1-7) peptide described herein reduces mucositis by one, two, three, or four stages (e.g., stage 4 to stage 3, 2, 1, or 0) or grades as compared to a control.
  • treatment with an angiotensin (1-7) peptide results in substantially free of Stage or Grade 4, 3, 2, or 1 mucositis in the subject following transplantation.
  • treatment with an angiotensin (1-7) peptide results in substantially free of Stage or Grade 4, 3 or 2 mucositis in the subject following transplantation.
  • treatment with an angiotensin (1-7) peptide results in substantially free of Stage or Grade 4 or 3 mucositis in the subject following transplantation. In some embodiments, treatment with an angiotensin (1-7) peptide results in substantially free of mucositis following transplantation.
  • a suitable control is a baseline measurement, such as a measurement in the same individual prior to initiation of the treatment described herein, or a measurement in a control individual (or multiple control individuals) in the absence of the treatment described herein.
  • a “control individual” is an individual afflicted with GVHD, mucositis or a recipient of transplant, who is about the same age and/or gender as the individual being treated (to ensure that the stages of the disease in the treated individual and the control individual(s) are comparable).
  • the individual (also referred to as "patient” or “subject") being treated is an individual (fetus, infant, child, adolescent, or adult human) suffering from or at risk of developing GVHD or mucositis.
  • kits or other articles of manufacture which contains an angiotensin (1-7) peptide or a formulation containing the same and provides instructions for its reconstitution (if lyophilized) and/or use.
  • Kits or other articles of manufacture may include a container, a syringe, vial and any other articles, devices or equipment useful in administration (e.g., subcutaneous, oral, by inhalation).
  • Suitable containers include, for example, bottles, vials, syringes (e.g., pre-filled syringes), ampules, cartridges, reservoirs, or lyo-jects.
  • the container may be formed from a variety of materials such as glass or plastic.
  • a container is a pre-filled syringe.
  • Suitable pre-filled syringes include, but are not limited to, borosilicate glass syringes with baked silicone coating, borosilicate glass syringes with sprayed silicone, or plastic resin syringes without silicone.
  • the container may holds formulations and a label on, or associated with, the container that may indicate directions for reconstitution and/or use.
  • the label may indicate that the formulation is reconstituted to concentrations as described above.
  • the label may further indicate that the formulation is useful or intended for, for example, subcutaneous administration.
  • a container may contain a single dose of a stable formulation containing an angiotensin (1-7) peptide.
  • a single dose of the stable formulation is present in a volume of less than about 15 ml, 10 ml, 5.0 ml, 4.0 ml, 3.5 ml, 3.0 ml, 2.5 ml, 2.0 ml, 1.5 ml, 1.0 ml, or 0.5 ml.
  • a container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the formulation.
  • Kits or other articles of manufacture may further include a second container comprising a suitable diluent (e.g., BWFI, saline, buffered saline).
  • kits or other articles of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. In some embodiments, kits or other articles of manufacture may include an instruction for self-administration.
  • Example 1 Clinical Trial of Angiotensin-(l-7) treated double cord blood transplant recipients Subject selection criteria
  • Human subjects were selected from subjects undergoing double cord blood transplantation for the treatment of a variety of malignancies. Malignancies included: having ⁇ 10% bone marrow blasts; having Acute Myelogenous Leukemia (AML) past first remission, in first or subsequent relapse, induction failure, or in first remission with high-risk for relapse (with high-risk cytogenetics or presence of flt3 mutation or with secondary leukemia from prior chemotherapy); having Myelodysplastic Syndrome or Myelofibrosis of intermediate or high-risk; having Acute Lymphoblastic Leukemia (ALL) with induction failure, in first complete remission with Philadelphia chromosome or translocation (4: 11), hypodiploidy and or evidence of minimal residual disease by flow cytometry, in second or third complete remission or second relapse; having Chronic Myelocytic leukemia (CML) in second chronic phase or accelerated phase;
  • AML Acute Myelogenous Leukemia
  • CML Chronic Mye
  • Non-Hodgkin's Lymphoma (NHL) with induction failure, in second or third complete remission or relapse; or having Chronic Lymphocytic leukemia (CLL) with progressive disease following standard therapy.
  • NHL Non-Hodgkin's Lymphoma
  • CLL Chronic Lymphocytic leukemia
  • Subjects were selected of either gender ranging in age from 18-60 years with adequate major organ function and with an Eastern Cooperative Oncology Group (ECOG) status of ⁇ 2. Adequate major organ function was demonstrated by: left ventricular ejection fraction of at least 40%: pulmonary function test (PFT) demonstrating a diffusion capacity of at least 50% predicted: creatinine ⁇ 1.6 mg/dL; Serum Glutamic Pyruvic Transaminase (SGPT)/ Serum Glutamic Oxaloacetic Transaminase (SGOT), alkaline phosphatase, and LDH ⁇ 3.0 times the upper limit of normal (ULN); and bilirubin ⁇ 1.5 times ULN.
  • PFT pulmonary function test
  • Female subjects capable of reproduction were required to have a negative beta human chorionic gonadotropin (PHCG) serum or urine pregnancy test result prior to starting the conditioning regimen.
  • Female subjects who were surgically sterilized or who had not experienced menses for > 2 years were not required to have a pregnancy test.
  • Subjects were selected for whom a 7/8 or 8/8 HLA-match related or 8/8 HLA- match unrelated bone marrow donor was not available, or if the progress of the subject's disease dictated that it was not in the subject's best interest to wait for an unrelated marrow donor to be procured.
  • Subjects were selected with two cord blood units matched with the subject at 4/6, 5/6, or 6/6 HLA class I (serological) and II (molecular) antigens available, with each unit containing at least 1 x 10 7 total nucleated cells/kg recipient body weight (pre-thaw).
  • the conditioning regimen was administered starting on Day -8 or Day -6 and was completed on Day -1. All subjects received one of the following two conditioning regimens shown in tables 1-2.
  • Acute GVHD Prophylaxis and Hematopoietic Cytokine Support are considered standard practice in subjects receiving allogeneic transplant.
  • Tacrolimus was administered in a starting dose of 0.03 mg/kg (ideal body weight) daily starting on Day -2, and was changed to oral dosing when tolerated. Tacrolimus was tapered around Day 180, if no acute GVHD was present.
  • Mycophenolate mofetil (MMF) was administrated at 1 gram twice daily. If the subject was ⁇ 50 kg, they were given 15 mg/kg orally, twice daily, adjusted to tablet size; intravenous administration at the same dosage was used if oral administration was not tolerated. MMF was started on day -3. If renal failure was present and glomerular filtration rate (GFR) was ⁇ 25 mL/min, 1 gram MMF twice daily was not exceeded. No dose adjustment was made for liver disease.
  • GFR glomerular filtration rate
  • MMF was stopped at day 100 if no acute GVHD was present. If the subject had acute GVHD requiring systemic therapy, MMF was optionally stopped 7 days after control of acute GVHD.
  • G-CSF was administered at a dose of 5 ⁇ g/kg/day (rounded up to the nearest vial) subcutaneously beginning on Day 0, and continuing until the absolute neutrophil count (ANC) was > 2.5 x 10 9 /L.
  • Ang-(l-7) was supplied as a sterile, non-pyrogenic solution for injection at concentrations of 30 mg/mL.
  • the parenteral formulation was produced as a pH-buffered solution, adjusted for the proper osmolality with the addition of 4% mannitol to provide a final osmolality of 295-415 mOsm.
  • the product was packaged in a 2-mL, single-use, stoppered vial with a 1.0-mL fiU.
  • Ang-(l-7) concentrations were standardized so that a ⁇ - ⁇ volume of solution was dispensed per kilogram of subject body mass for subjects receiving 300 ⁇ g/kg, and 33 ⁇ was dispensed per kilogram of subject body mass for subjects receiving 1000 ⁇ g/kg. The dose was calculated for each subject based on the subject's mass.
  • Ang-(l-7) was aseptically drawn into a syringe prior to injection. Qualified site personnel administered Ang-(l-7) at the investigational site. A new, unused vial or vials was used for each dose. Ang-(l-7) was administered once daily with a subcutaneous needle into sites located in the abdomen or thigh. Given the potential discomfort associated with a larger volume administered subcutaneously to subjects treated at the 1000 ⁇ g/kg/day dose, the full dose was, at the Investigator's discretion, given as two injections into different sites.
  • the treatment period begins on day 0, the day of the cord blood transplant.
  • the following assessments are performed on day 0: physical exam including weight (the Day 0 weight measurement will determine the dose of Ang-(l-7) to be administered throughout the treatment period); vital signs (blood pressure, heart rate, respirations, and temperature); severity grading of oral mucositis; acute GVHD assessment at engraftment; adverse events and concomitant therapies (including blood transfusion and hematopoietic growth factors) within 30 days of first Ang-(l-7) dose; pregnancy test for women of childbearing potential; CBC with differential and platelet count; chemistry panel (sodium, chloride, potassium, magnesium, bicarbonate, BUN, creatinine, glucose, calcium, phosphorus, total bilirubin, ALT, AST, total protein, albumin, and alkaline phosphatase); and amylase and LDH.
  • the following assessments were performed daily during the treatment period: vital signs (blood pressure, heart rate, respirations, and temperature);
  • concomitant medications including blood transfusion and hematopoietic growth factors
  • CBC hematopoietic growth factors
  • the following assessments were performed weekly at days 6, 13, 20 and 27 of the treatment period: a symptom-directed physical exam; weight;
  • chemistry panel sodium, chloride, potassium, magnesium, bicarbonate, BUN, creatinine, glucose, calcium, phosphorus, total bilirubin, ALT, AST, total protein, albumin, and alkaline phosphatase); amylase and LDH; PT/PTT and INR (day 6 only); and chest X-Ray (Day 27 only). For all weekly assessments during the treatment period, there was a +/- 1 day window.
  • the follow-up period begins one week after the point at which a subject completes the treatment period (i.e., one week after Day 27) and lasts up to day 100 after transplantation.
  • the initial follow-up visit was performed +1- 2 days following subject treatment period completion. There was no allowable window at Engraftment. All other assessments during the follow-up period were performed in a +/- 3 day window.
  • the follow-up period assessment timepoints are days 34 (Month 1), 41, 48, 55, 62 (Month 2), 69, 76, 83, and 90.
  • concomitant medications including blood transfusion and hematopoietic growth factors
  • GVHD was assessed using the criteria shown in tables 3 and 4.
  • the skin, liver and gut were staged from 1-4 according to degree of involvement.
  • Exemplary criteria for staging are shown in table 3.
  • Skin staging was assessed by extent of maculopapular rash, generalized erythroderma, generalized exfoliative dermatitis, ulcerative dermatitis, and/or bullous formation.
  • Liver staging was assessed by bilirubin level.
  • Gut staging was assessed by volume of diarrhea and/or presence of nausea, abdominal cramping, abdominal pain and/or ileus.
  • a clinical grade from 0 to IV was determined based on skin stage, liver stage, gut stage, and/or functional impairment.
  • Exemplary criteria for clinical grading are shown in table 4.
  • Table 5 shows clinical trial data for 9 subjects treated with 300 ⁇ g/kg/day Ang-(1-
  • Incidence of mucositis is defined by the occurrence of least one adverse event with Medical Dictionary for Regulatory Activities (MedDRA) preferred term that includes “mucositis” or “stomatitis”.
  • MedDRA Medical Dictionary for Regulatory Activities
  • Grade 3 severe
  • Grade 4 lifethreatening
  • Grades 3-4 severe or life-threatening
  • the severity grade was determined by NCI-CTCAE.
  • the incidence of Grade 3 mucositis will be summarized based on the proportion of subjects with at least one episode of Grade 3 mucositis. Similar analyses will be performed for Grade 4 and Grade 3-4 events.

Landscapes

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

Abstract

L'invention concerne des compositions et des procédés destinés au traitement et/ou à la prévention de la maladie du greffon contre l'hôte. En particulier, l'invention porte sur des compositions et des procédés qui permettent de traiter et/ou prévenir la maladie du greffon contre l'hôte à l'aide de peptides de l'angiotensine-(1-7) ou d'équivalents fonctionnels de ces derniers. En particulier, l'invention peut servir à faciliter une transplantation et à réduire ou prévenir l'apparition de la maladie du greffon contre l'hôte, de la mucosite et d'autres maladies, troubles et états.
PCT/US2013/037308 2012-04-19 2013-04-19 Compositions et procédés pour le traitement de la maladie du greffon contre l'hôte WO2013158959A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261635786P 2012-04-19 2012-04-19
US61/635,786 2012-04-19

Publications (1)

Publication Number Publication Date
WO2013158959A1 true WO2013158959A1 (fr) 2013-10-24

Family

ID=49384093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/037308 WO2013158959A1 (fr) 2012-04-19 2013-04-19 Compositions et procédés pour le traitement de la maladie du greffon contre l'hôte

Country Status (1)

Country Link
WO (1) WO2013158959A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018075888A1 (fr) * 2016-10-21 2018-04-26 Regents Of The University Of Minnesota Matériaux et procédés pour traiter ou prévenir une maladie du greffon contre l'hôte
WO2019236472A1 (fr) * 2018-06-04 2019-12-12 Oncoimmune, Inc. Procédés d'utilisation de cd24 pour la prévention et le traitement de la maladie du greffon contre l'hôte et de la mucosite

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0739212A1 (fr) * 1994-01-21 1996-10-30 Genetics Institute, Inc. Utilisation de l'interleukine-12 pour prevenir la reaction du greffon contre l'hote
US20070213307A1 (en) * 2001-01-19 2007-09-13 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
JP2011088920A (ja) * 2002-11-06 2011-05-06 Cbio Ltd シャペロニン10免疫抑制

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0739212A1 (fr) * 1994-01-21 1996-10-30 Genetics Institute, Inc. Utilisation de l'interleukine-12 pour prevenir la reaction du greffon contre l'hote
US20070213307A1 (en) * 2001-01-19 2007-09-13 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
JP2011088920A (ja) * 2002-11-06 2011-05-06 Cbio Ltd シャペロニン10免疫抑制

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018075888A1 (fr) * 2016-10-21 2018-04-26 Regents Of The University Of Minnesota Matériaux et procédés pour traiter ou prévenir une maladie du greffon contre l'hôte
WO2019236472A1 (fr) * 2018-06-04 2019-12-12 Oncoimmune, Inc. Procédés d'utilisation de cd24 pour la prévention et le traitement de la maladie du greffon contre l'hôte et de la mucosite
CN112512640A (zh) * 2018-06-04 2021-03-16 肿瘤免疫股份有限公司 Cd24用于预防和治疗移植物抗宿主病和粘膜炎的使用方法

Similar Documents

Publication Publication Date Title
JP6472914B2 (ja) 脳状態の処置におけるアンジオテンシン
EP3373951B1 (fr) Méthodes et compositions pour le traitement d'une épidermolyse bulleuse
US20230110554A1 (en) Methods and compositions for treatment of coronavirus infection
CA2961375A1 (fr) Compositions et methodes permettant le traitement d'un syndrome d'irradiation aigue
US20180050079A1 (en) Angiotensin peptides in treating marfan syndrome and related disorders
US9333233B2 (en) Methods and compositions for the delayed treatment of stroke
WO2013158959A1 (fr) Compositions et procédés pour le traitement de la maladie du greffon contre l'hôte
US20160199436A1 (en) Methods and compositions for the treatment of amyotrophic lateral sclerosis
JP2023541403A (ja) ビオチンをミトコンドリアに送達するための方法及び組成物
AU2014332346B2 (en) Novel peptide compositions
WO2015057403A2 (fr) Compositions et méthodes de traitement d'une maladie inflammatoire chronique de l'intestin
KR102666922B1 (ko) 미토콘드리아에 비오틴의 전달을 위한 방법 및 조성물
US9133241B2 (en) Peptide compositions
KR20240073137A (ko) 미토콘드리아에 비오틴의 전달을 위한 방법 및 조성물
EP2334324A2 (fr) Bbeta (15-42) et ses analogues combine avec la cyclosporine pour le traitement des lesions de la reperfusion
WO2016010844A1 (fr) Procédés et compositions pour le traitement de l'accident vasculo-cérébral

Legal Events

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

Ref document number: 13778593

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13778593

Country of ref document: EP

Kind code of ref document: A1