US20140023621A1 - Bone marrow derived cd271 precursor cells for cardiac repair - Google Patents

Bone marrow derived cd271 precursor cells for cardiac repair Download PDF

Info

Publication number
US20140023621A1
US20140023621A1 US13/819,154 US201113819154A US2014023621A1 US 20140023621 A1 US20140023621 A1 US 20140023621A1 US 201113819154 A US201113819154 A US 201113819154A US 2014023621 A1 US2014023621 A1 US 2014023621A1
Authority
US
United States
Prior art keywords
cells
heart
cell
msc
cardiac
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/819,154
Other languages
English (en)
Inventor
Joshua M. Hare
Ian K. McNiece
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hare Joshua M
Original Assignee
University of Miami
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Miami filed Critical University of Miami
Priority to US13/819,154 priority Critical patent/US20140023621A1/en
Assigned to UNIVERSITY OF MIAMI reassignment UNIVERSITY OF MIAMI ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HARE, JOSHUA M., MCNIECE, IAN K.
Publication of US20140023621A1 publication Critical patent/US20140023621A1/en
Assigned to HARE, JOSHUA M reassignment HARE, JOSHUA M ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF MIAMI
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Definitions

  • Embodiments of the invention are directed to methods for treating cardiovascular diseases using bone marrow derived mesenchymal-precursor cells.
  • Heart failure is responsible for $33.2 billion in direct and indirect costs to the US healthcare system (1, 2) and the majority of patients with this diagnosis have scarred myocardium from previous MI.
  • Left ventricular function is the most important determinant of survival and quality of life in patients who have suffered a myocardial infarction (MI) (1, 3).
  • MI myocardial infarction
  • the myocardium has very limited regenerative potential after infarction and a major quest in medicine presently is that of cell-based tissue regeneration. This quest holds the promise of transforming the treatment of numerous chronic illnesses.
  • chronic ischemic cardiomyopathy a disorder affecting more than 4 million Americans
  • a successful cell-based therapeutic will have an enormous impact on patient morbidity and mortality, and reduce the societal burdens of this disorder.
  • Embodiments of the invention relate to compositions comprising adult bone marrow derived precursors of mesenchymal stem cells. These cells are administered in vivo, such as for example, the heart and regenerate the myocardium.
  • a method of preventing or treating cardiovascular diseases or disorders comprises isolating CD271 ⁇ mesenchymal stem cell precursors (MSCs) from bone marrow of a subject; administering to a patient a therapeutically effective amount of isolated CD271 + mesenchymal stem cell (MSC) precursors.
  • MSCs mesenchymal stem cell precursors
  • the CD271 + MSCs are isolated from bone marrow cells having a low affinity nerve growth receptor (NGFR; CD271).
  • the CD271 + stem cells are isolated from donors (or sources) comprising: autologous, syngeneic, allogeneic, or xenogeneic.
  • the MSC precursor cells differentiate into at least one lineage comprising: myocardial, vascular, or endothelial lineages.
  • the isolated precursor mesenchymal stem cells are cultured ex vivo and expanded prior to administration to a patient.
  • the non-adherent stem cells are expanded and administered to a patient.
  • the precursor mesenchymal stem cells are optionally administered to a patient in varying concentrations over a period of time.
  • the wherein the precursor mesenchymal stem cells are optionally conditioned with media conditioned by heart derived stromal cells.
  • one or more agents are optionally administered to the patient, the agents comprising at least one of: cytokines, chemotactic factors, growth factors, or differentiation factors.
  • an adult stem cell comprises a bone marrow mesenchymal stem cell (MSC) precursor derived cell having a CD271 ⁇ phenotype.
  • MSC bone marrow mesenchymal stem cell
  • FIG. 1 shows the morphology of CD271 + cells. Cytospins were prepared and stained with Wright Giemsa.
  • FIG. 2A -2C shows MSC formation from CD271 + cells ( FIG. 2A ; 170,000 cells per T75 cm 2 flask); BM MNC ( FIG. 2B ; 15 million cells per T75 cm 2 flask) and C) CD271 ⁇ cells ( FIG. 2C ; 17 million cells per T75 cm 2 flask).
  • FIG. 3 shows a typical CFU-F colony at day 10 of culture.
  • FIG. 4 shows the culture of CD 271 + cells in Teflon bags for 7, 14 and 21 days.
  • FIG. 5 shows the flow analysis of non-adherent mesenchymal stem cells (NA-MSC). Isotype control staining is shown in the green line and CD105-FITC in the shaded area.
  • NA-MSC non-adherent mesenchymal stem cells
  • FIGS. 6A and 6B show the osteogenic and adipocyte differentiation from human bone marrow CD271 + cells. Osteogenic and adipocyte differentiation were performed as described in the methods. Presence of osteogenic differentiation was shown by expression of alkaline phosphatase (AP) stained by FAST BCIP/NBT ( FIG. 6A ; x/100) on day 14. Differentiation to adipocyte was shown by Oil Red O staining ( FIG. 6B ; x/100) on day 11. A representative example of three experiments is shown.
  • AP alkaline phosphatase
  • FIG. 7 shows the expression of cardiac markers in cultured CD271 ⁇ cells.
  • FIG. 8 shows the echocardiographic comparison of treatment groups.
  • FIGS. 9A-9D show the immunohistochemistry of heart sections from NOD/SCID mice injected with CD271 + cells.
  • the heart sections were stained with Alu sequence and co stained with cardiac markers ( ⁇ SA, troponin I (TnI), Connexin 43 (Cx)(40 ⁇ ).
  • FIG. 9A shows a slice from Border zone with positive cells embedded in vascular wall and also between host myocytes.
  • FIG. 9B shows a remote zone towards the base of the heart still showing injected human cells.
  • FIG. 9C shows a large blood vessel with alu positive cells in remote area.
  • FIG. 9D shows the borderzone of another heart with numerous positive cells.
  • Stem cells show potential for many different areas of health and medical research. Some of the most serious medical conditions, such as cancer and birth defects, are caused by problems that occur somewhere in the process of stem cell differentiation or maintenance. Broadly, there are two different types of stem cells, embryonic stem cells and adult stem cells. Embryonic stem cells are found in blastocysts and have the ability to differentiate into all of the specialized embryonic tissues. Adult stem cells are undifferentiated cells found throughout the body after embryonic development. Adult stem cells are able to divide and replenish dying cells and regenerate damaged tissue. Furthermore, adult stem cells can maintain the normal turnover of regenerative organs such as blood, skin and intestinal tissue. Adult stem cells have the ability to divide and self-renew indefinitely and are able to generate all of the cell types of the organ from which they originate.
  • Stem cells can be classified as being totipotent, pluripotent, multipotent or unipotent based on their potential to differentiate into different cell types.
  • Totipotent stem cells are produced from the fusion of gametes and the first few divisions of the fertilized egg. These cells can differentiate into embryonic and extra-embryonic cell types.
  • Pluripotent stem cells can differentiate into cells from any of the three germ layers.
  • Multipotent calls can produce only cells of a closely related family.
  • Unipotent cells can produce only one cell type, but have the property of self-renewal which distinguishes them from non-stem cells. Most adult stem cells are lineage restricted multipotent stem cells, and are referred to by their tissue of origin.
  • Pluripotent adult stem cells are rare and generally small in number but can be found in a number of tissues including umbilical cord blood (Ratajczak M. Z., et al., Leukemia 21(5): 860-867 (2007)).
  • There are several different types of adult stem cells including, but not limited to, adipose derived stem cells (Zuk, P. A., et al., Tissue Engineering 7:211-216) (2001)), epithelial stem cells, hematopoietic stem cells, mammary stem cells (Shackleton, M., et al., Breast Cancer R E.
  • mesenchymal stem cells mesenchymal stem cells
  • endothelial stem cells neural stem cells
  • olfactory stem cells e.g., testicular stem cells, dental pulp derived stem cells, and umbilical cord blood hematopoietic progenitor cells.
  • an adult stem cell When an adult stem cell divides, it creates another cell like itself and a cell more differentiated than itself. This process of asymmetric cell division, gives rise to one identical daughter cell and one early transient-amplifying cell (early TA), which possesses high proliferative capacity. Through a series of cell divisions, the early TA cell gives rise to a late TA cell followed by a tissue-specific progenitor cell and finally to the bulk of differentiated cells that make up the organ or tissue (Ribacka, C., et al. Ann. Med. epub ahead of print: 1-10 (2008)).
  • TA early transient-amplifying cell
  • Embodiments of the invention may be practiced without the theoretical aspects presented. Moreover, the theoretical aspects are presented with the understanding that Applicants do not seek to be bound by the theory presented.
  • the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.
  • a “stem cell” as used herein is an undifferentiated cell which is capable of essentially unlimited propagation either in vivo or ex vivo and capable of differentiation to other cell types. This can be to certain differentiated, committed, immature, progenitor, or mature cell types present in the tissue from which it was isolated, or dramatically differentiated cell types, such as for example the erythrocytes and lymphocytes that derive from a common precursor cell, or even to cell types at any stage in a tissue completely different from the tissue from which the stem cell is obtained.
  • blood stem cells may become brain cells or liver cells
  • neural stem cells can become blood cells, such that stem cells are pluripotential, and given the appropriate signals from their environment, they can differentiate into any tissue in the body.
  • Propagation can be determined, for example, by the ability of an isolated stem cell to be propagated through at least 50, preferably 100, and even up to 200 or more cell divisions in a cell culture system.
  • Stem cells can be “totipotent,” meaning that they can give rise to all the cells of an organism as for germ cells.
  • Stem cells can also be “pluripotent,” meaning that they can give rise to many different cell types, but not all the cells of an organism. When a stem cell differentiates it generally gives rise to a more adult cell type, which may be a partially differentiated cell such as a progenitor cell, a differentiated cell, or a terminally differentiated cell.
  • Stem cells can be highly motile.
  • Isolating a stem cell refers to the process of removing a stem cell from a tissue sample and separating away other cells which are not stem cells of the tissue.
  • An isolated stem cell will be generally free from contamination by other cell types, i.e. “homogeneity” or purity” and will generally have the capability of propagation and differentiation to produce mature cells of the tissue from which it was isolated.
  • An isolated stem cell can exist in the presence of a small fraction of other cell types which do not interfere with the utilization of the stem cell for analysis or production of other, differentiated cell types.
  • Isolated stem cells will generally be at least 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 98%, 99%, or 99.9% pure.
  • isolated stem cells according to the invention will be at least 98% or at least 99% pure.
  • culturing refers to propagating or nurturing a cell, collection of cells, tissue, or organ, by incubating for a period of time in an environment and under conditions which support cell viability or propagation. Culturing can include one or more of the steps of expanding and proliferating a cell, collection of cells, tissue, or organ according to the invention.
  • BMDC single marrow derived progenitor cell
  • bone marrow derived stem cell refers to a primitive stem cell with the machinery for self-renewal constitutively active. Included in this definition are stem cells that are totipotent, pluripotent and precursors.
  • a “precursor cell” can be any cell in a cell differentiation pathway that is capable of differentiating into a more mature cell.
  • the term “precursor cell population” refers to a group of cells capable of developing into a more mature cell.
  • a precursor cell population can comprise cells that are totipotent, cells that are pluripotent and cells that are stem cell lineage restricted (i.e. cells capable of developing into less than all hematopoietic lineages, or into, for example, only cells of erythroid lineage).
  • autologous is meant to refer to any material derived from the same individual to whom it is later to be re-introduced into the individual.
  • xenogeneic cell refers to a cell that derives from a different animal species than the animal species that becomes the recipient animal host in a transplantation or vaccination procedure.
  • allogeneic cell refers to a cell that is of the same animal species but genetically different in one or more genetic loci as the animal that becomes the “recipient host”. This usually applies to cells transplanted from one animal to another non-identical animal of the same species.
  • genotypic cell refers to a cell which is of the same animal species and has the same genetic composition for most genotypic and phenotypic markers as the animal who becomes the recipient host of that cell line in a transplantation or vaccination procedure. This usually applies to cells transplanted from identical twins or may be applied to cells transplanted between highly inbred animals.
  • safe and effective amount refers to the quantity of a component which is sufficient to yield a desired therapeutic response without undue adverse side effects (such as toxicity, irritation, or allergic response) commensurate with a reasonable benefit/risk ratio when used in the manner of this invention.
  • therapeutically effective amount is meant an amount of a compound of the present invention effective to yield the desired therapeutic response.
  • the specific safe and effective amount or therapeutically effective amount will vary with such factors as the particular condition being treated, the physical condition of the patient, the type of mammal or animal being treated, the duration of the treatment, the nature of concurrent therapy (if any), and the specific formulations employed and the structure of the compounds or its derivatives.
  • Patient or “subject” refers to mammals and includes human and veterinary subjects.
  • diagnosis refers to classifying a disease or a symptom, determining a severity of the disease, monitoring disease progression, forecasting an outcome of a disease and/or prospects of recovery.
  • detecting may also optionally encompass any of the above.
  • a “biological sample obtained from the subject” may also optionally comprise a sample that has not been physically removed from the subject, as described in greater detail below.
  • Treatment is an intervention performed with the intention of preventing the development or altering the pathology or symptoms of a disorder. Accordingly, “treatment” refers to both therapeutic treatment and prophylactic or preventative measures. Those in need of treatment include those already with the disorder as well as those in which the disorder is to be prevented.
  • ameliorated refers to a symptom which is approaches a normalized value (for example a value obtained in a healthy patient or individual), e.g., is less than 50% different from a normalized value, preferably is less than about 25% different from a normalized value, more preferably, is less than 10 % different from a normalized value, and still more preferably, is not significantly different from a normalized value as determined using routine statistical tests.
  • a normalized value for example a value obtained in a healthy patient or individual
  • a therapeutically effective amount” of an agent or compound, cells etc. means an amount sufficient to produce a therapeutically (e.g., clinically) desirable result.
  • the compositions can be administered one from one or more times per day to one or more times per week; including once every other day.
  • certain factors can influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present.
  • treatment of a subject with a therapeutically effective amount of the compounds of the invention can include a single treatment or a series of treatments.
  • a “prophylactically effective amount” may refer to the amount of precursor mesenchymal stem cells sufficient to prevent the recurrence of heart diseases or disorders, for example, ischemia, or the occurrence of such in a patient, including but not limited to those predisposed to heart disease, for example those genetically predisposed to heart disease, stroke, etc.
  • a prophylactically effective amount may also refer to the amount of the prophylactic agent that provides a prophylactic benefit in the prevention of disease.
  • sample refers to a biological sample, such as, for example; one or more cells, tissues, or fluids (including, without limitation, plasma, serum, whole blood, cerebrospinal fluid, lymph, tears, urine, saliva, milk, pus, and tissue exudates and secretions) isolated from an individual or from cell culture constituents, as well as samples obtained from, for example, a laboratory procedure.
  • a biological sample such as, for example; one or more cells, tissues, or fluids (including, without limitation, plasma, serum, whole blood, cerebrospinal fluid, lymph, tears, urine, saliva, milk, pus, and tissue exudates and secretions) isolated from an individual or from cell culture constituents, as well as samples obtained from, for example, a laboratory procedure.
  • a biological sample may comprise chromosomes isolated from cells (e.g., a spread of metaphase chromosomes), organelles or membranes isolated from cells, whole cells or tissues, nucleic acid such as genomic DNA in solution or bound to a solid support such as for Southern analysis, RNA in solution or bound to a solid support such as for Northern analysis, cDNA in solution or bound to a solid support, oligonucleotides in solution or bound to a solid support, polypeptides or peptides in solution or bound to a solid support, a tissue, a tissue print and the like.
  • nucleic acid such as genomic DNA in solution or bound to a solid support such as for Southern analysis, RNA in solution or bound to a solid support such as for Northern analysis, cDNA in solution or bound to a solid support, oligonucleotides in solution or bound to a solid support, polypeptides or peptides in solution or bound to a solid support, a tissue, a tissue print and the like.
  • tissue or fluid collection methods can be utilized to collect the biological sample from the subject in order to determine the level of DNA, RNA and/or polypeptide of the variant of interest in the subject. Examples include, but are not limited to, fine needle biopsy, needle biopsy, core needle biopsy and surgical biopsy (e.g., brain biopsy), and lavage. Regardless of the procedure employed, once a biopsy/sample is obtained the level of the variant can be determined and a diagnosis can thus be made.
  • heart disease or disorders or “cardiovascular diseases or disorders” refer to any type of heart disease or disorders including cardiomyopathy, hypertrophic cardiomyopathy, dilated cardiomyopathy, atherosclerosis, coronary artery disease, ischemic heart disease, myocarditis, viral infection, wounds, hypertensive heart disease, valvular disease, congenital heart disease, myocardial infarction, congestive heart failure, arrhythmias, diseases resulting in remodeling of the heart, heart failure, ischemia, myocardial infarction, transplantation, hypertension, restenosis, angina pectoris, rheumatic heart disease, or congenital cardiovascular defects.
  • Diseases or disorders of the heart can be due to any reason, such as for example, damage to cardiac tissue such as a loss of contractility (e.g., as might be demonstrated by a decreased ejection fraction).
  • Cardiac damage or disorders characterized by insufficient cardiac function includes any impairment or absence of a normal cardiac function or presence of an abnormal cardiac function.
  • Abnormal cardiac function can be the result of disease, injury, and/or aging.
  • abnormal cardiac function includes morphological and/or functional abnormality of a cardiomyocyte, a population of cardiomyocytes, or the heart itself.
  • Non-limiting examples of morphological and functional abnormalities include physical deterioration and/or death of cardiomyocytes, abnormal growth patterns of cardiomyocytes, abnormalities in the physical connection between cardiomyocytes, under- or over-production of a substance or substances by cardiomyocytes, failure of cardiomyocytes to produce a substance or substances which they normally produce, and transmission of electrical impulses in abnormal patterns or at abnormal times.
  • Abnormalities at a more gross level include dyskinesis, reduced ejection fraction, changes as observed by echocardiography (e.g., dilatation), changes in EKG, changes in exercise tolerance, reduced capillary perfusion, and changes as observed by angiography.
  • ischemic heart disease e.g., angina pectoris, myocardial infarction, chronic ischemic heart disease, hypertensive heart disease, pulmonary heart disease (cor pulmonale), valvular heart disease, e.g., rheumatic fever, mitral valve prolapse, calcification of mitral annulus, carcinoid heart disease, infective endocarditis, congenital heart disease, myocardial disease, e.g., myocarditis, dilated cardiomyopathy, hypertensive cardiomyopathy, cardiac disorders which result in congestive heart failure, and tumors of the heart, e.g., primary sarcomas and secondary tumors.
  • Heart damage also includes wounds, such as for example, knife wound; biological (e.g. viral; autoimmune diseases) or chemical (e.g. chemotherapy, drugs); surgery; transplantation and the like.
  • Myocardial ischemia refers to a lack of oxygen flow to the heart which results in myocardial ischemic damage.
  • myocardial ischemic damage includes damage caused by reduced blood flow to the myocardium.
  • Non-limiting examples of causes of myocardial ischemia and myocardial ischemic damage include: decreased aortic diastolic pressure, increased intraventricular pressure and myocardial contraction, coronary artery stenosis (e.g., coronary ligation, fixed coronary stenosis, acute plaque change (e.g., rupture, hemorrhage), coronary artery thrombosis, vasoconstriction), aortic valve stenosis and regurgitation, and increased right atrial pressure.
  • coronary artery stenosis e.g., coronary ligation, fixed coronary stenosis, acute plaque change (e.g., rupture, hemorrhage), coronary artery thrombosis, vasocons
  • Non-limiting examples of adverse effects of myocardial ischemia and myocardial ischemic damage include: myocyte damage (e.g., myocyte cell loss, myocyte hypertrophy, myocyte cellular hyperplasia), angina (e.g., stable angina, variant angina, unstable angina, sudden cardiac death), myocardial infarction, and congestive heart failure. Damage due to myocardial ischemia may be acute or chronic, and consequences may include scar formation, cardiac remodeling, cardiac hypertrophy, wall thinning, dilatation, and associated functional changes.
  • the existence and etiology of acute or chronic myocardial damage and/or myocardial ischemia may be diagnosed using any of a variety of methods and techniques well known in the art including, e.g., non-invasive imaging (e.g., MRI, echocardiography), angiography, stress testing, assays for cardiac-specific proteins such as cardiac troponin, and clinical symptoms. These methods and techniques as well as other appropriate techniques may be used to determine which subjects are suitable candidates for the treatment methods described herein.
  • Ischemic cardiomyopathy is the leading cause of heart failure in developed countries, few therapies exist to improve cardiac function once infarct remodeling has occurred, and treatments that actually reverse deleterious remodeling of the heart after a myocardial infarction (MI) are lacking
  • MI myocardial infarction
  • BM bone marrow
  • MSCs mesenchymal stem cells
  • a precursor of MSCs can be isolated from the bone marrow based upon expression of the low affinity nerve growth factor receptor (NGFR; CD271) and the CD271 + cells are a readily available cell source for therapeutic use.
  • NGFR low affinity nerve growth factor receptor
  • CD271 + cells can be obtained from a bone marrow aspiration and isolated to sufficient quantities in 4 to 5 hours, providing a logistical advantage for immediate use.
  • these cells are dramatically superior in efficacy to cultured MSCs.
  • the goal of the studies is to conduct preclinical trials testing BM-CD271 + cells in a rodent and swine model of myocardial infarction and translate this work to a clinical trial of direct surgical injections of CD-271 + cells following coronary artery bypass surgery.
  • the central hypothesis is that BM-CD271 + cells delivered by surgical injection will engraft, improve cardiac function, and reduce scar size.
  • CD271 + cells are indeed precursors of MSC and have the potential to repair ischemic tissue in a mouse model of myocardial infarction (MI).
  • MI myocardial infarction
  • CD271 + cells are more potent and have greater capacity for differentiation into cardiomyocytes than do cultured MSCs.
  • adequate numbers of CD271 + cells can be isolated from BM in four to five hours providing a major logistic advance to treat patients immediately with autologous bone marrow derived therapy.
  • bone marrow derived (BM) derived precursor mesenchymal stem CD271 + cells are utilized in the treatment of ischemic tissue in patients with heart failure.
  • a method of preventing or treating cardiovascular diseases or disorders comprises administering to a patient an effective amount of CD271 + stem cells.
  • the CD271 + cells are isolated from bone marrow cells having a low affinity nerve growth receptor (NGFR; CD271).
  • the CD271 + stem cells are autologous, syngeneic, allogeneic, xenogeneic or combinations thereof.
  • the administered stem cells populate and repair damaged tissue, for example, cardiac tissue. These cells differentiate into the various lineages resulting in the regeneration and repair of damaged tissue.
  • one or more agents are optionally administered to the patient, the agents comprising at least one of: cytokines, chemotactic factors, growth factors, or differentiation factors.
  • a method for treating a patient with a heart disease or injury comprising administering a therapeutic cell composition to a patient with a disease or injury of the heart or circulatory system, and evaluating the patient for improvements in cardiac function, wherein said cell composition comprises CD271 + as described herein.
  • the heart disease is a cardiomyopathy.
  • the cardiomyopathy is either idiopathic or a cardiomyopathy with a known cause.
  • the cardiomyopathy is either ischemic or nonischemic in nature.
  • the disease of the heart or circulatory system comprises one or more of angioplasty, aneurysm, angina (angina pectoris), aortic stenosis, aortitis, arrhythmias, arteriosclerosis, arteritis, asymmetric septal hypertrophy (ASH), atherosclerosis, atrial fibrillation and flutter, bacterial endocarditis, Barlow's Syndrome (mitral valve prolapse), bradycardia, Buerger's Disease (thromboangiitis obliterans), cardiomegaly, cardiomyopathy, carditis, carotid artery disease, coarctation of the aorta, congenital heart diseases (congenital heart defects), congestive heart failure (heart failure), coronary artery disease, Eisenmenger's Syndrome, embolism, endocarditis, erythromelalgia, fibrillation, fibromuscular dysplasia, heart block, heart murmur, hypertension, hypotension,
  • the disease of the heart or circulatory system comprises one or more of acute rheumatic fever, acute rheumatic pericarditis, acute rheumatic endocarditis, acute rheumatic myocarditis, chronic rheumatic heart diseases, diseases of the mitral valve, mitral stenosis, rheumatic mitral insufficiency, diseases of aortic valve, diseases of other endocardial structures, ischemic heart disease (acute and subacute), angina pectoris, diseases of pulmonary circulation (acute pulmonary heart disease, pulmonary embolism, chronic pulmonary heart disease), kyphoscoliotic heart disease, myocarditis, endocarditis, endomyocardial fibrosis, endocardial fibroelastosis, atrioventricular block, cardiac dysrhythmias, myocardial degeneration, diseases of the circulatory system including cerebrovascular disease, occlusion and stenosis of precerebral
  • treatment comprises treatment of a patient with a cardiomyopathy with a therapeutic cell composition comprising CD271 + cells, either with or without another cell type.
  • the patient experiences benefits from the therapy for example from the ability of the cells to support the growth of other cells, including stem cells or progenitor cells present in the heart, from the tissue ingrowth or vascularization of the tissue, and from the presence of beneficial cellular factors, chemokines, cytokines and the like.
  • Improvement in an individual having a disease or disorder of the circulatory system, wherein the individual is administered the CD271 + cells or therapeutic compositions provided herein, can be assessed or demonstrated by detectable improvement in one or more symptoms of the disease or disorder of the circulatory system.
  • improvement in an individual having a disease or disorder of the circulatory system, wherein the individual is administered the CD271 ⁇ cells or therapeutic compositions provided herein can be assessed or demonstrated by detectable improvement in one or more, indicia of cardiac function, for example, demonstration of detectable improvement in one or more of chest cardiac output (CO), cardiac index (CI), pulmonary artery wedge pressures (PAWP), and cardiac index (CI), % fractional shortening (% FS), ejection fraction (EF), left ventricular ejection fraction (LVEF); left ventricular end diastolic diameter (LVEDD), left ventricular end systolic diameter (LVESD), contractility (e.g.
  • CO chest cardiac output
  • CI cardiac index
  • PAWP pulmonary artery wedge pressures
  • CI cardiac index
  • % FS % fractional shortening
  • EF ejection fraction
  • LVEF left ventricular ejection fraction
  • LVEDD left ventricular end diastolic diameter
  • LVESD left
  • dP/dt pressure-volume loops, measurements of cardiac work, an increase in atrial or ventricular functioning; an increase in pumping efficiency, a decrease in the rate of loss of pumping efficiency, a decrease in loss of hemodynamic functioning; and a decrease in complications associated with cardiomyopathy, as compared to the individual prior to administration of CD271 + cells.
  • Improvement in an individual receiving the therapeutic compositions provided herein can also be assessed by subjective metrics, e.g., the individual's self-assessment about his or her state of health following administration.
  • the methods of treatment provided herein comprise inducing the therapeutic CD271 ⁇ cells to differentiate along mesenchymal lineage, e.g., towards a cardiomyogenic, angiogenic or vasculogenic phenotype, or into cells such as myocytes, cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells).
  • a cardiomyogenic, angiogenic or vasculogenic phenotype or into cells such as myocytes, cardiomyocytes, endothelial cells, myocardial cells, epicardial cells, vascular endothelial cells, smooth muscle cells (e.g. vascular smooth muscle cells).
  • CD271 + cells, or therapeutic compositions comprising such cells, to an individual in need thereof can be accomplished, e.g., by transplantation, implantation (e.g., of the cells themselves or the cells as part of a matrix-cell combination), injection (e.g., directly to the site of the disease or condition, for example, directly to an ischemic site in the heart of an individual who has had a myocardial infarction), infusion, delivery via catheter, or any other means known in the art for providing cell therapy.
  • the therapeutic cell compositions are provided to an individual in need thereof, for example, by injection into one or more sites in the individual.
  • the therapeutic cell compositions are provided by intracardiac injection, e.g., to an ischemic area in the heart.
  • the cells are injected onto the surface of the heart, into an adjacent area, or even to a more remote area.
  • the cells can home to the diseased or injured area.
  • an individual having a disease or condition of the coronary or vascular systems can be administered CD271 ⁇ cells at any time the cells would be therapeutically beneficial.
  • the cells or therapeutic compositions of the invention are administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 days of the myocardial infarction.
  • Administration proximal in time to a myocardial infarction e.g., within 1-3 or 1-7 days, is preferable to administration distal in time, e.g., after 3 or 7 days after a myocardial infarction.
  • the cells or therapeutic compositions of the invention are administered within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 days of initial diagnosis of the disease or condition.
  • kits for use in the treatment of myocardial infarction provide the therapeutic cell composition which can be prepared in a pharmaceutically acceptable form, for example by mixing with a pharmaceutically acceptable carrier, and an applicator, along with instructions for use.
  • the kit can be used in the field, for example in a physician's office, or by an emergency care provider to be applied to a patient diagnosed as having had a myocardial infarction or similar cardiac event.
  • the CD271 + cells are administered with stem cells that are not CD271 + cells, myoblasts, myocytes, cardiomyoblasts, cardiomyocytes, or progenitors of myoblasts, myocytes, cardiomyoblasts, and/or cardiomyocytes.
  • the methods of treatment provided herein comprise administering CD271 + cells, e.g., a therapeutic composition comprising the cells, to a patient with a disease of the heart or circulatory system; and evaluating the patient for improvements in cardiac function, wherein the therapeutic cell composition is administered as a matrix-cell complex.
  • the matrix is a scaffold, preferably bioabsorbable, comprising at least the cells.
  • populations of CD271 + cells are incubated or are administered to a patient in the presence of one or more factors which stimulate stem or progenitor cell differentiation along a cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathway.
  • factors are known in the art; determination of suitable conditions for differentiation can be accomplished with routine experimentation.
  • factors include, but are not limited to factors, such as growth factors, chemokines, cytokines, cellular products, demethylating agents, and other stimuli which are now known or later determined to stimulate differentiation, for example of stem cells, along cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathways or lineages.
  • CD271 + cells may be differentiated along cardiogenic, angiogenic, hemangiogenic, or vasculogenic pathways or lineages by culture of the cells in the presence of factors comprising at least one of a demethylation agent, a BMP, FGF, Wnt factor protein, Hedgehog, and/or anti-Wnt factors.
  • Inclusion of demethylation agents tends to allow the cells to differentiate along mesenchymal lines, toward a cardiomyogenic pathway. Differentiation can be determined by, for example, expression of at least one of cardiomyosin, skeletal myosin, or GATA4; or by the acquisition of a beating rhythm, spontaneous or otherwise induced; or by the ability to integrate at least partially into a patient's cardiac muscle without inducing arrhythmias.
  • Demethylation agents that can be used to initiate such differentiation include, but are not limited to, 5-azacytidine, 5-aza-2′-deoxycytidine, dimethylsulfoxide, chelerythrine chloride, retinoic acid or salts thereof, 2-amino-4-(ethylthio)butyric acid, procainamide, and procaine.
  • cells become cardiomyogenic, angiogenic, hemangiogenic, or vasculogenic cells, or progenitors.
  • cells integrate into a recipient's cardiovascular system, including but not limited to heart muscle, vascular and other structures of the heart, cardiac or peripheral blood vessels, and the like.
  • the CD271 + cells differentiate into cells acquiring two or more of the indicia of cardiomyogenic cells or their progenitors, and able to integrate into a recipient's heart or vasculature.
  • the cells, which administered to an individual result in no increase in arrhythmias, heart defects, blood vessel defects or other anomalies of the individual's circulatory system or health.
  • the CD271 + cells act to promote the differentiation of stem cells naturally present in the patient's cardiac muscle, blood vessels, blood and the like to themselves differentiate into for example, cardiomyocytes, or at least along cardiomyogenic, angiogenic, hemangiogenic, or vasculogenic lines.
  • CD271 + cells, and populations of such cells can be provided therapeutically or prophylactically to an individual, e.g., an individual having a disease, disorder or condition of, or affecting, the heart or circulatory system.
  • diseases, disorders or conditions can include congestive heart failure due to atherosclerosis, cardiomyopathy, or cardiac injury, e.g., an ischemic injury, such as from myocardial infarction or wound (acute or chronic).
  • the CD271 + cells may be administered to an individual in the form of a therapeutic composition comprising the cells and another therapeutic agent, such as insulin-like growth factor (IGF), platelet-derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), IL-8, an antithrombogenic agent (e.g., heparin, heparin derivatives, urokinase, and PPack (dextrophenylalanine proline arginine chloromethylketone); antithrombin compounds, platelet receptor antagonists, anti-thrombin antibodies, anti-platelet receptor antibodies, aspirin, dipyridamole, protamine, hirudin, prostaglandin inhibitors, and/or platelet inhibitors), an antiapoptotic agent (e.g., EPO, EPO derivatives and analogs, and their salts, TPO, IGF-I, IGF-II
  • therapeutic compositions comprising the CD271 + cells further comprise one or more additional cell types, e.g., adult cells (for example, fibroblasts or endodermal cells), or stem or progenitor cells.
  • additional cell types e.g., adult cells (for example, fibroblasts or endodermal cells), or stem or progenitor cells.
  • Such therapeutic agents and/or one or more additional cells can be administered to an individual in need thereof individually or in combinations or two or more such compounds or agents.
  • the individual to be treated is a mammal. In a specific embodiment the individual to be treated is a human. In specific embodiments, the individual is a livestock animal or a domestic animal. In other specific embodiments, the individual to be treated is a horse, sheep, cow or steer, pig, dog or cat.
  • the population of stem cells is at least about 80% pure as compared to a control sample of cells isolated from a heart tissue, preferably, the stem cell population is about 90% pure as compared to a control sample of cells, preferably, the stem cell population is about 95%, 96%, 97%, 98%, 99%, 99.9% pure as compared to a control sample of cells.
  • the isolated CD 271 ’ stem cell populations can be used in any assay desired by the end user, such as for example, expressing a non-native or foreign molecule, a native molecule which may or may not be activated in the cells.
  • examples of such molecules can be growth factors, receptors, ligands, therapeutic agents, etc.
  • the molecules can be selected by the end user for expression by the isolated stem cells depending on the end user's need.
  • the molecules comprise, for example, a polypeptide, a peptide, an oligonucleotide, a polynucleotide, an organic or inorganic molecule.
  • stem cells may be embryonic stem cells, adult stem cells, umbilical cord blood stem cells, somatic stem cells or cancer stem cells.
  • the stem cells are adult stem cells, preferably cardiac stem cells.
  • the stem cells of the current invention may be hematopoietic stem cells, or mesenchymal stem cells.
  • the stem cells of the current invention may be totipotent, pluripotent, multipotent or unipotent stem cells.
  • Stem cells according to the current invention may be selected for by the presence of one or more stem cell markers including but not limited to: CD133, CD34, CD38, CD117/c-kit, OCT3/4, Nanog, RUNX2, SOX9, Integrin, SPARC, osteocalcin, endoglin and STRO-1.
  • the stem cells of the current invention may be primary stem cells or may be derived from an established stem cell line, premalignant stem cell line, cancer cell line, or any cell line that manifests any stem cell marker.
  • Primary stem cells may be derived from a cancer patient or a healthy patient.
  • Isolation of CD271 + stem cell populations is useful many types of applications, for example, transplantation into heart or other organs for the treatment of cardiac diseases or disorders, such as damaged myocardium.
  • damaged myocardium refers to myocardial cells which have been exposed to ischemic conditions. These ischemic conditions may be caused by a myocardial infarction, or other cardiovascular disease or related complaint. The lack of oxygen causes the death of the cells in the surrounding area, leaving an infarct, which will eventually scar.
  • age-related cardiomyopathy refers to the deterioration of the myocardium as a result of intrinsic mechanisms occurring as the organism ages.
  • the stem cells are used in methods of repairing and/or regenerating damaged myocardium or age-related cardiomyopathy in a subject in need thereof by administering isolated stem cells to areas of damaged myocardium, wherein the administered stem cells differentiate into one or more of myocytes, endothelial cells, or smooth muscle cells.
  • the differentiated cells may proliferate and form various cardiac structures including coronary arteries, arterioles, capillaries, and myocardium, which are all structures essential for proper function in the heart.
  • the ability to restore both functional and structural integrity is yet another aspect of this invention.
  • the stem cells are adult cardiac stem cells.
  • adult cardiac stem cells are isolated from cardiac tissue harvested from the subject in need of therapeutic treatment for one of the cardiac or vasculature conditions and implanted back into the subject.
  • the isolated CD271 + stem cells are cultured and expanded ex vivo prior to administration of the stem cells to a patient.
  • the cells can be for example, autologous, syngeneic, allogeneic, xenogeneic or any combination thereof.
  • the same source of stem cells does not have to be used if successive administrations are required.
  • the invention involves administering a therapeutically effective dose or amount of stem cells to the heart.
  • An effective dose is an amount sufficient to effect a beneficial or desired clinical result.
  • the dose could be administered in one or more administrations.
  • the precise determination of what would be considered an effective dose may be based on factors individual to each patient, including their size, age, area of myocardial damage, and amount of time since damage.
  • One skilled in the art specifically a physician or cardiologist, would be able to determine the number of stem cells that would constitute an effective dose without undue experimentation.
  • the isolated stem cells are activated prior to administration to a subject.
  • Activation of the stem cells may be accomplished by exposing the isolated stem cells to one or more cytokines, such as hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), or variant thereof.
  • HGF positively influences stem cell migration and homing through the activation of the c-Met receptor (Kollet et al. (2003) J. Clin. Invest. 112: 160-169; Linke et al. (2005) Proc. Natl. Acad. Sci. USA 102: 8966-8971; Rosu-Myles et al. (2005) J. Cell. Sci. 118: 4343-4352; Urbanek et al.
  • HGF hepatocyte growth factor
  • IGF-1 insulin-like growth factor-1
  • IGF-1 and its corresponding receptor induce cardiac stem cell division, upregulate telomerase activity, hinder replicative senescence and preserve the pool of functionally-competent cardiac stem cells in the heart (Kajstura et al. (2001) Diabetes 50: 1414-1424; Torella et al. (2004) Circ. Res. 94: 514-524; Davis et al. (2006) Proc. Natl. Acad. Sci. USA 103: 8155-8160).
  • the isolated stem cells are contacted with hepatocyte growth factor (HGF) and/or insulin-like growth factor-1 (IGF-1).
  • HGF hepatocyte growth factor
  • IGF-1 insulin-like growth factor-1
  • cytokines that are suitable for the activation of the isolated stem cells include Activin A, Bone Morphogenic Protein 2, Bone Morphogenic Protein 4, Bone Morphogenic Protein 6, Cardiotrophin-1, Fibroblast Growth Factor 1, Fibroblast Growth Factor 4, Flt3 Ligand, Glial-Derived Neurotrophic Factor, Heparin, Insulin-like Growth Factor-II, Insulin-Like Growth Factor Binding Protein-3, Insulin-Like Growth Factor Binding Protein-5, Interleukin-3, Interleukin-6, Interleukin-8, Leukemia Inhibitory Factor, Midkine, Platelet-Derived Growth Factor AA, Platelet-Derived Growth Factor BB, Progesterone, Putrescine, Stem Cell Factor, Stromal-Derived Factor-1, Thrombopoietin, Transforming Growth Factor- ⁇ , Transforming Growth Factor- ⁇ 1, Transforming Growth Factor-P2, Transforming Growth
  • Functional variants of the above-mentioned cytokine variants can also be employed in the invention. Functional cytokine variants would retain the ability to bind and activate their corresponding receptors. Variants can include amino acid substitutions, insertions, deletions, alternative splice variants, or fragments of the native protein.
  • NK1 and NK2 are natural splice variants of HGF, which are able to bind to the c-MET receptor.
  • the administration of stem cells to a subject in need thereof is accompanied by the administration of one or more cytokines to the heart.
  • the cytokines may be selected from the group consisting of stem cell factor (SCF), granulocyte-colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), stromal cell-derived factor-1, steel factor, vascular endothelial growth factor, macrophage colony stimulating factor, granulocyte-macrophage stimulating factor, hepatocyte growth factor (HGF), insulin-like growth factor-1 (IGF-1), Interleukin-3, or any cytokine capable of the stimulating and/or mobilizing stem cells.
  • SCF stem cell factor
  • G-CSF granulocyte-colony stimulating factor
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • stromal cell-derived factor-1 steel factor
  • vascular endothelial growth factor macrophage colony stimulating factor
  • the cytokines are selected from HGF, IGF-1, functional variants of HGF or IGF-1, or combinations thereof.
  • the cytokines may be delivered simultaneously with the CD271 ⁇ population of stem cells.
  • the administration of the cytokines may either precede or follow the administration of the stem cells by a specified time period.
  • the time period may be about 15 minutes, about 30 minutes, about 1 hour, about 3 hours, about 6 hours, about 12 hours, about 24 hours, about 36 hours, about 1 week, about 2 weeks, about 1 month, or about 6 months.
  • the cytokines may be delivered to the heart by one or more administrations. In one embodiment, cytokines are delivered by a single administration. In another embodiment, multiple administrations of the same dosage of cytokines are delivered to the heart.
  • a preferred embodiment of the invention includes administration of multiple doses of the cytokines to the heart, such that a chemotactic gradient is formed.
  • a chemotactic gradient extending from the atria and/or apex of the heart to the mid-region of the left ventricle may be established by administering multiple doses of increasing cytokine concentration.
  • the chemotactic gradient can be formed from the site of implantation of the stem cells to the mid-region of the left ventricle or the border region of infarcted myocardium.
  • At least two cytokines are used in the formation of the chemotactic gradient.
  • the concentration of the first cytokine remains constant while the concentration of the second cytokine is variable, thereby forming the chemotactic gradient.
  • the chemotactic gradient is formed by multiple administrations of IGF-1 and HGF, wherein the concentration of IGF-1 remains constant and the concentration of HGF is variable.
  • the variable concentrations of HGF may range from about 0.1 to about 400 ng/ml. In other embodiments, the concentration of IGF-1 may be from about 0.1 to about 500 ng/ml.
  • the isolated CD271 + population of stem cells and cytokines may be administered to the heart by injection.
  • the injection is preferably intramyocardial. As one skilled in the art would be aware, this is the preferred method of delivery for stem cells and/or cytokines as the heart is a functioning muscle. Injection by this route ensures that the injected material will not be lost due to the contracting movements of the heart.
  • the stem cells and/or cytokines are administered by injection transendocardially or trans-epicardially.
  • This preferred embodiment allows the cytokines to penetrate the protective surrounding membrane, necessitated by the embodiment in which the cytokines are injected intramyocardially.
  • Another preferred embodiment of the invention includes use of a catheter-based approach to deliver the trans-endocardial injection.
  • the use of a catheter precludes more invasive methods of delivery wherein the opening of the chest cavity would be necessitated. As one skilled in the art would appreciate, optimum time of recovery would be allowed by the more minimally invasive procedure.
  • a catheter approach involves the use of such techniques as the NOGA catheter or similar systems.
  • the NOGA catheter system facilitates guided administration by providing electromechanic mapping of the area of interest, as well as a retractable needle that can be used to deliver targeted injections or to bathe a targeted area with a therapeutic. Any of the embodiments of the present invention can be administered through the use of such a system to deliver injections or provide a therapeutic.
  • One of skill in the art will recognize alternate systems that also provide the ability to provide targeted treatment through the integration of imaging and a catheter delivery system that can be used with the present invention.
  • Information regarding the use of NOGA and similar systems can be found in, for example, Sherman (2003) Basic Appl. Myol. 13: 11-14; Patel et at (2005) The Journal of Thoracic and Cardiovascular Surgery 130:1631-38; and Perrin et al.
  • the isolated cardiac stem cells are administered by an intracoronary route of administration.
  • One of skill in the art will recognize other useful methods of delivery or implantation which can be utilized with the present invention, including those described in Dawn et al. (2005) Proc. Natl. Acad. Sci. USA 102, 3766-3771, the contents of which are incorporated herein in their entirety.
  • the methods of the present invention are useful for the treatment of cardiovascular disease, including, but not limited to, atherosclerosis, ischemia, hypertension, restenosis, angina pectoris, rheumatic heart disease, congenital cardiovascular defects, age-related cardiomyopathy, and arterial inflammation and other disease of the arteries, arterioles and capillaries.
  • cardiovascular disease including, but not limited to, atherosclerosis, ischemia, hypertension, restenosis, angina pectoris, rheumatic heart disease, congenital cardiovascular defects, age-related cardiomyopathy, and arterial inflammation and other disease of the arteries, arterioles and capillaries.
  • the methods of the present invention provide for the repair and/or regeneration of damaged myocardium resulting from one of the diseases listed above or from the general decline of myocardial cells with age.
  • the present invention also encompasses methods of preventing or treating heart failure in a subject comprising administering an isolated, CD271 + population of adult cardiac stem cells into the subject's heart and administering an angiotensin II receptor antagonist.
  • the CD271 + population adult cardiac stem cells are activated prior to administration by exposure to one or more cytokines as described herein.
  • one or more cytokines are administered to the heart to form a chemotactic gradient causing the administered adult cardiac stem cells to migrate to areas of myocardial damage.
  • the one or more cytokines are HGF, IGF-1, or variants thereof.
  • the renin-angiotensin system is a hormone system that facilitates the regulation of blood pressure and extracellular volume in the body.
  • RAS renin-angiotensin system
  • renin cleaves angiotensinogen, an inactive precursor peptide secreted by the liver, into angiotensin I.
  • Angiotensin I is subsequently converted into angiotensin II (Ang II) by angiotensin-converting enzyme (ACE), which is predominantly found in the lungs.
  • ACE angiotensin-converting enzyme
  • Ang II produces many effects, including vasoconstriction and secretion of aldosterone and vasopressin, through activation of the AT1 receptor.
  • Ang II has been implicated in the age-dependent accumulation of oxidative damage in the heart (Fiordaliso et al. (2001) Diabetes 50: 2363-2375; Kajstura et al. (2001) Diabetes 50: 1414-1424), and has been reported to induce senescence and decrease the number and function of endothelial progenitor cells (Kobayashi et al. (2006) Hypertens. Res. 29: 449-455).
  • Ang II triggers apoptosis in myocytes (Leri et al. (1998) J. Clin. Invest. 101: 1326-1342) and may contribute to the progression of heart failure (McMurray et al. (2003) Lancet 362: 767-771).
  • inhibition of AT1 receptors has been shown to improve the clinical outcome of patients with chronic heart failure and prolong life in humans (McMurray et al. (2003) Lancet 362: 767-771).
  • the invention provides for methods of preventing heart failure and/or treating chronic heart failure in a subject by administering an Ang II receptor antagonist in combination with administration of adult cardiac stem cells to the subject's heart.
  • the Ang II receptor antagonist is an antagonist of the AT1 receptor.
  • Some non-limiting examples of Ang II receptor antagonists that would be encompassed by the invention include Valsartan, Telmisartan, Losartan, Irbesartan, Olmesartan, Candesartan, and Eprosartan.
  • ACE angiotensin converting enzyme
  • ACE inhibitors which may be used in the methods of the prevent invention include, but are not limited to, Benazepril, Enalapril, Lisinopril, Captopril, Fosinopril, Ramipril, Perindopril, Quinapril, Moexipril, and Trandolapril.
  • the Ang II receptor antagonists or ACE inhibitors may be administered to the subject in multiple doses subsequent to the administration of the adult cardiac stem cells.
  • the antagonists or inhibitors may be taken on a routine schedule for a set period of time. For example, the inhibitors may be taken once daily for about 1 month, about 2 months, about 3 months, about 6 months, about 12 months, or about 24 months after administration of the adult cardiac stem cells. Other dosing schedules may be employed.
  • One of skill in the art, particularly a physician or cardiologist would be able to determine the appropriate dose and schedule for the administration of the ACE inhibitors or Ang II receptor antagonists.
  • one or more symptoms of heart failure is reduced or alleviated following administration of the cardiac stem cells and the angiotensin II receptor antagonist and/or ACE inhibitor.
  • Symptoms of heart failure include, but are not limited to, fatigue, weakness, rapid or irregular heartbeat, dyspnea, persistent cough or wheezing, edema in the legs and feet, and swelling of the abdomen.
  • compositions such as pharmaceutical compositions, including adult stem cells and/or at least one cytokine, for instance, for use in inventive methods for treating cardiovascular disease, heart failure or other cardiac conditions.
  • the pharmaceutical composition comprises isolated human cardiac stem cells and a pharmaceutically acceptable carrier.
  • the methods and/or compositions, including pharmaceutical compositions comprise effective amounts of adult cardiac stem cells or two or more cytokines in combination with an appropriate pharmaceutical agent useful in treating cardiac and/or vascular conditions.
  • the pharmaceutical compositions of the present invention are delivered via injection.
  • routes for administration include, but are not limited to, subcutaneous or parenteral including intravenous, intraarterial (e.g. intracoronary), intramuscular, intraperitoneal, intramyocardial, transendocardial, trans-epicardial, intranasal administration as well as intrathecal, and infusion techniques.
  • the pharmaceutical composition is preferably in a form that is suitable for injection.
  • it will generally be formulated in a unit dosage injectable form (solution, suspension, emulsion).
  • the pharmaceutical formulations suitable for injection include sterile aqueous solutions or dispersions and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • the carrier can be a solvent or dispersing medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • Nonaqueous vehicles such as cottonseed oil, sesame oil, olive oil, soybean oil, corn oil, sunflower oil, or peanut oil and esters, such as isopropyl myristate, may also be used as solvent systems for compound compositions.
  • various additives which enhance the stability, sterility, and isotonicity of the compositions including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added.
  • compositions of the present invention can be administered to the subject in an injectable formulation containing any compatible carrier, such as various vehicles, adjuvants, additives, and diluents; or the compounds utilized in the present invention can be administered parenterally to the subject in the form of slow-release subcutaneous implants or targeted delivery systems such as monoclonal antibodies, iontophoretic, polymer matrices, liposomes, and microspheres.
  • the pharmaceutical compositions utilized in the present invention can be administered orally to the subject. Conventional methods such as administering the compounds in tablets, suspensions, solutions, emulsions, capsules, powders, syrups and the like are usable. Known techniques which deliver the compound orally or intravenously and retain the biological activity are preferred.
  • a composition of the present invention can be administered initially, and thereafter maintained by further administration.
  • a composition of the invention can be administered in one type of composition and thereafter further administered in a different or the same type of composition.
  • a composition of the invention can be administered by intravenous injection to bring blood levels to a suitable level.
  • the subject's levels are then maintained by an oral dosage form, although other forms of administration, dependent upon the subject's condition, can be used.
  • the quantity of the pharmaceutical composition to be administered will vary for the subject being treated. In a preferred embodiment, 2 ⁇ 10 4 to about 1 ⁇ 10 5 adult cardiac stem cells and, optionally, 50-500 ⁇ g/kg per day of a cytokine or variant of said cytokine are administered to the subject.
  • any additives in addition to the active stem cell(s) and/or cytokine(s) are present in an amount of 0.001 to 50 wt % solution in phosphate buffered saline, and the active ingredient is present in the order of micrograms to milligrams, such as about 0.0001 to about 5 wt %, preferably about 0.0001 to about 1 wt %, most preferably about 0.0001 to about 0.05 wt % or about 0.001 to about 20 wt %, preferably about 0.01 to about 10 wt %, and most preferably about 0.05 to about 5 wt %.
  • any composition to be administered to an animal or human it is preferred to determine therefore: toxicity, such as by determining the lethal dose (LD) and LD 50 in a suitable animal model e.g., rodent such as mouse; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response.
  • toxicity such as by determining the lethal dose (LD) and LD 50 in a suitable animal model e.g., rodent such as mouse
  • LD 50 lethal dose
  • LD 50 low-d dose
  • compositions comprising a therapeutic of the invention include liquid preparations for orifice, e.g., oral, nasal, anal, vaginal, peroral, intragastric, mucosal (e.g., perlingual, alveolar, gingival, olfactory or respiratory mucosa) etc., administration such as suspensions, syrups or elixirs; and, preparations for parenteral, subcutaneous, intradermal, intramuscular or intravenous administration (e.g., injectable administration), such as sterile suspensions or emulsions.
  • Such compositions may be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose or the like.
  • compositions can also be lyophilized.
  • the compositions can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired. Standard texts, such as “REMINGTON'S PHARMACEUTICAL SCIENCE”, 17th edition, 1985, incorporated herein by reference, may be consulted to prepare suitable preparations, without undue experimentation.
  • compositions of the invention are conveniently provided as liquid preparations, e.g., isotonic aqueous solutions, suspensions, emulsions or viscous compositions which may be buffered to a selected pH.
  • suitable carriers and other additives will depend on the exact route of administration and the nature of the particular dosage form, e.g., liquid dosage form (e.g., whether the composition is to be formulated into a solution, a suspension, gel or another liquid form), or solid dosage form (e.g., whether the composition is to be formulated into a pill, tablet, capsule, caplet, time release form or liquid-filled form).
  • Solutions, suspensions and gels normally contain a major amount of water (preferably purified water) in addition to the active compound.
  • compositions can be isotonic, i.e., they can have the same osmotic pressure as blood and lacrimal fluid.
  • the desired isotonicity of the compositions of this invention may be accomplished using sodium chloride, or other pharmaceutically acceptable agents such as dextrose, boric acid, sodium tartrate, propylene glycol or other inorganic or organic solutes.
  • Sodium chloride is preferred particularly for buffers containing sodium ions.
  • Viscosity of the compositions may be maintained at the selected level using a pharmaceutically acceptable thickening agent.
  • Methylcellulose is preferred because it is readily and economically available and is easy to work with.
  • suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like.
  • a pharmaceutically acceptable preservative can be employed to increase the shelf-life of the compositions.
  • Benzyl alcohol may be suitable, although a variety of preservatives including, for example, parabens, thimerosal, chlorobutanol, or benzalkonium chloride may also be employed.
  • a suitable concentration of the preservative will be from 0.02% to 2% based on the total weight although there may be appreciable variation depending upon the agent selected.
  • the components of the compositions should be selected to be chemically inert with respect to the active compound. This will present no problem to those skilled in chemical and pharmaceutical principles, or problems can be readily avoided by reference to standard texts or by simple experiments (not involving undue experimentation), from this disclosure and the documents cited herein.
  • compositions can be administered in dosages and by techniques well known to those skilled in the medical and veterinary arts taking into consideration such factors as the age, sex, weight, and condition of the particular subject, and the composition form used for administration (e.g., solid vs. liquid).
  • Dosages for humans or other mammals can be determined without undue experimentation by the skilled artisan, from this disclosure, the documents cited herein, and the knowledge in the art.
  • Suitable regimes for initial administration and further doses or for sequential administrations also are variable, may include an initial administration followed by subsequent administrations; but nonetheless, may be ascertained by the skilled artisan, from this disclosure, the documents cited herein, and the knowledge in the art.
  • compositions of the present invention are used to treat heart failure and cardiovascular diseases, including, but not limited to, atherosclerosis, ischemia, hypertension, restenosis, angina pectoris, rheumatic heart disease, congenital cardiovascular defects and arterial inflammation and other diseases of the arteries, arterioles and capillaries or related complaint.
  • cardiovascular diseases including, but not limited to, atherosclerosis, ischemia, hypertension, restenosis, angina pectoris, rheumatic heart disease, congenital cardiovascular defects and arterial inflammation and other diseases of the arteries, arterioles and capillaries or related complaint.
  • the invention involves the administration of adult stem cells as herein discussed, alone or in combination with one or more cytokines or variant of said cytokine, as herein discussed, for the treatment or prevention of any one or more of these conditions or other conditions involving heart disease or disorders.
  • Advantageous routes of administration involves those best suited for treating these conditions, such as via injection, including, but are not limited to subcutaneous or parenteral including intravenous, intraarterial, intramuscular, intraperitoneal, intramyocardial, transendocardial, trans-epicardial, intranasal administration as well as intrathecal, and infusion techniques.
  • subcutaneous or parenteral including intravenous, intraarterial, intramuscular, intraperitoneal, intramyocardial, transendocardial, trans-epicardial, intranasal administration as well as intrathecal, and infusion techniques.
  • Non-Standard Abbreviations and Acronyms Bone Marrow (BM); Colony Forming Unit Fibroblast (CFU-F); Coronary Artery Bypass Grafting (CABG); Ejection Fraction (EF); End Diastolic Volume (EDV); End Systolic Volume (ESV); Mesenchymal Stem Cell (MSC); Mononuclear Cell (MNC); Mouse Heart Stromal Cell Conditioned Media (MsHrtStr CM); Myocardial Infarction (MI); Non adherent MSC (NA-MSC); Recombinant human basic fibroblast growth factor (rhbFGF).
  • mice Human cells were transplanted into NOD/SCID mice subject to left coronary artery ligation and induction of myocardial infarction (MI). Only male, 2 month old mice were studied. The cohorts were as follows: 1) 10 mice injected with human MSC; 2) 10 mice injected with human BM-CD271 + cells.
  • mice were anesthetized with 5% isoflurane for induction and then etomidate 20 mg/kg i.p. Endotracheal intubation was performed and the mouse was then placed on a cardiac monitor and ventilated mechanically.
  • the skin over the site of left lateral thoracotomy was prepped and draped in sterile fashion using providone-iodine 10% solution.
  • a heating pad was used to keep mice warm during procedures to prevent heat loss.
  • Surgically sterile non-medicated ophthalmic ointment was applied to the eyes preoperatively to prevent corneal drying.
  • the chest was opened via left lateral thoracotomy.
  • the left coronary artery was exposed and ligated to produce an anterior MI.
  • 10 minutes after occlusion erythropoietin (2-6 ⁇ g/kg) was infused through an internal jugular catheter.
  • the chest was closed in layers with 0 and 3.0 (for muscle) absorbable suture, and buprenorphine (0.05-0.1 mg/kg s.c.) were given post-operatively for pain.
  • Mice were bandaged and kept in standard sterile isolated housing where they recover for 3 to 4 days following surgery. Pain control was achieved with buprenorphine 0.05-0.1 mg/kg s.c. q12 h ⁇ 6 doses.
  • mice Sham-operated mice that experience all but the placement of the coronary artery ligature serve as controls. Perioperative mortality was low in the sham group ( ⁇ 15%), and slightly higher for the infarcted cohorts. Of the remaining mice, about 75% survived at least two months post-MI. The mice were typically studied for four weeks after surgery.
  • Stem cells were administered by direct intra-myocardial injection using a 30 gauge needle. On the day of infarction, while the chest was open for MI induction, the cells were injected directly into the myocardium. Three injections of 100 ⁇ l were delivered.
  • Model of chronic ischemic cardiomyopathy Gottingen miniswine (25-30 kg, female, 10-12 months of age) were used and subjected to a large animal model of ischemic heart failure.
  • the minipig was given ketamine for induction of anesthesia, endotracheal intubation was performed, and isoflurane given for maintenance of general anesthesia.
  • the pigs have continuous monitoring of noninvasive BP, heart rate, temperature, pulse oximetery, and capnography. A longitudinal incision in the mid-neck is made and the right common carotid artery and internal jugular vein were exposed.
  • Proximal and distal control with vessel loops is obtained, and a 7 Fr vascular access sheath was then placed in both the right common carotid artery and right internal jugular vein. Pressure volume loops with IVC occlusion were obtained pre and post MI. Left and right coronary angiograms were conducted with a JR 4 catheter. Then the minipigs undergo experimental anterior wall infarction by balloon occlusion of the left anterior descending (LAD) coronary artery just distal to the first diagonal branch for 2.5 hours. The pig was monitored for arrhythmias and Advanced Cardiac Life Support initiated if necessary.
  • LAD left anterior descending
  • the carotid artery is repaired with 6-0 prolene sutures and the internal jugular vein ligated.
  • the neck incision is closed in 3 layers; the fascia, subcutaneous tissue and skin are re-approximated with 3-0 polysorb.
  • the mini pig was then recovered and the scar, typically a transmural infarction approximately 20% of the left ventricle and localized to the anteroseptal wall, was allowed to heal for 3 months as the heart undergoes remodeling.
  • BM-MSCs or human BM-CD271 ⁇ cells Delivery of human BM-MSCs or human BM-CD271 ⁇ cells: At 3 months post-MI, the minipig was given ketamine for induction of anesthesia, placed supine on the operating table, given isoflurane via a mask, endotracheal intubation was performed, and continued on isoflurane for maintenance of general anesthesia. Vascular access of the left common carotid artery and internal jugular vein was obtained as described above to conduct hemodynamic assessment with pressure-volume loops.
  • a left anterior-lateral thoracotomy incision was made in the 5-6th intercostal space with a number 10 scalpel; the soft tissues were dissected with electrocautery to control bleeding; the parietal pleura was identified; using metzenbaum scissors it was opened to enter the left pleural cavity with care not to injure the lung parenchyma; a rib retractor was used to spread the ribs.
  • the pericardium was identified and a longitude incision was made with metzenbaum scissors with care to stay anterior to the phrenic nerve and not injure the myocardium.
  • the heart was exposed and 10 injections to the area of scar and border zone was done with a syringe filled with 0.5 cc of MSC or CD271 + cells. Total injected volume was 5 cc. Any areas of bleeding were controlled with pledgeted sutures.
  • the thoracotomy incision was closed in three layers with 2-0 polysorb sutures with an 18Fr chest tube to underwater suction placed in the lateral aspect of the incision. The pig was then weaned from the ventilator and recovered. The chest tube was removed the next day.
  • the cardiac MR protocol includes ECG-gated cine images, first-pass gadolinium perfusion imaging, tagged MR images, and delayed hyperenhancement images.
  • the typical MR scan obtains approximately 1,200 images of the heart that requires extensive post-processing analysis. All images were maintained on the University of Miami WebPax system and each study downloaded to a Dell Precision 690 workstation. Two FDA approved software programs were used for the analysis, Segment (Medviso AB and Lund University, Lund, Sweden) and Diagnosoft (Cary, N.C.). The cine and delayed enhancement images were analyzed using the Segment Software.
  • the tagged images and first pass perfusion images were analyzed with the Diagnosoft program to obtain peak eularian circumferential strain and myocardial upslope and area under the curve, respectively.
  • Sacrifice and Histology At 3 months after injection of cells each animal was sacrificed under deep anesthesia as discussed above. The heart was arrested by central infusion of 40 mEq of potassium chloride which arrests the heart in diastole. The pig's hearts were harvested and preserved in formaldehyde. Each heart was sectioned in the short axis and biopsies of the infarct, border zone, and remote zone were analyzed with confocal microscopy for engraftment and differentiation. In addition, whole body necropsy of the pigs was done to evaluate for ectopic tissue formation.
  • Mouse Surgeries Mouse surgeries were performed with concern for the prevention of pain and discomfort. As indicated above, appropriate agents were used during any surgical procedures to provide anesthesia and analgesia. For coronary artery ligation, the mice were anesthetized with 5% isoflurane for induction and then etomidate 20 mg/kg i.p. Pain control was achieved with buprenorphine 0.05-0.1 mg/kg s.c. q12 h. In addition, mouse body temperature was maintained using warming blankets or lamps. Although some mice developed cardiac hypertrophy and heart failure due to the different surgical procedures, mice that experienced excessive weight loss, labored breathing, cyanosis and non-responsiveness were assessed, and if appropriate, euthanized early.
  • Swine Surgeries All pig procedures were conducted with concern for the prevention of pain and suffering. As discussed above all procedures are conducted under general anesthesia. Post operative pain was controlled with a postoperative injection of buprenophine 0.05-0.1 mg/kg subcutaneous and a fentanyl 25 mcg patch is placed on the dorsum of the pig for 72 hours. If at any time during the course of the study a pig was in distress it was assessed by the veterinarian and investigative team. Many cases of distress are due to an infection of the wound or lungs which can be managed with oral or intramuscular antibiotics.
  • Euthanasia These methods are consistent with the recommendations of the 2007 American Veterinary Medical Association Guidelines on Euthanasia.
  • mice All swine and mice were euthanized for one of two indications: 1) exhibiting significant post-operative pain or distress which was not alleviated by analgesics, antibiotics and other measures; 2) completing the amount of time designated for the study of physiology. Mice were euthanized with an overdose of ketamine (150 mg/kg) and xylazine (10 mg/kg) and then by cervical dislocation. Swine were placed under deep anesthesia as discussed previously and the heart is arrested with 40 mEq of potassium chloride.
  • Differentiated stem cells were tracked by immunostaining Human cells were detected within the mouse hearts using alu staining. Staining for troponin I, ⁇ -sarcomeric actin, cardiac myocyte, desmin, ⁇ -cardiac actinin, connexin-43, GATA-4, Nkx-2.5, and MEF2 indicated myocytes. CD31 and vimentin staining was used to identify endothelial cells, while ⁇ -smooth muscle actin and SMA22 were used to identify smooth muscle cells. Thus, all cells resulting from injection of human CD271 or MSC and the potential for these cells to regenerate different lineages can be identified.
  • myocyte cell numbers and size were measured by counting nuclei using the method of Bruel and Nyengaard.
  • slides were stained with fluorescein-conjugated wheat germ agglutinin (Invitrogen) and Hoechst 33258.
  • Myocyte volume was calculated using a combination of the Cavalieri and dissector principles.
  • individual myocytes can be measured by confocal microscopy after acute dissociation using morphometric software.
  • Results were corroborated by Masson's trichrome.
  • paraffin-embedded sections were stained using the Apoptag Red In Situ Apoptosis Detection Kit (Millipore) based on the indirect TUNEL method.
  • Other assays include immunohistochemistry with an antibody specific for cleaved caspase-3, detection by Western blotting of caspase-dependent PKC ⁇ cleavage, and DNA laddering.
  • CD271 cells When evaluated histologically, CD271 cells exhibited a high degree of engraftment and evidence of myocyte differentiation, supporting the idea that cardiac recovery is due to cell engraftment in the injured myocardium. These studies definitively test the hypothesis that CD271 MSC precursors are capable of myocyte and vascular differentiation, and repair of the injured heart to a greater degree than cultured MSCs.
  • a large animal model of chronic ischemic cardiomyopathy was used and is well-established in this laboratory (7, 8) to test whether CD271 cells engraft and differentiate into myocytes, endothelial cells, and vascular smooth muscle cells. Cardiac MRI was also used to test the effects of CD271 + cells on cardiac function, scar size and myocardial perfusion. The inventors have extensive experience in creating models of ischemic cardiomyopathy in various breeds of swine (8, 9).
  • CsA oral cyclosporine A
  • MRI serial cardiac magnetic resonance imaging
  • This imaging modality allowed detailed phenotypic analysis of global cardiac function, regional cardiac function employing HARP, scar size using delayed hyper-enhancement imaging, and myocardial perfusion upslope and area under the curve by first pass perfusion of gadolinium.
  • the animals were sacrificed and the hearts harvested for immunohistochemical analysis to determine engraftment and differentiation of CD271 ⁇ cells.
  • Whole body necropsy was performed on each animal to evaluate for ectopic tissue formation.
  • cardiac MRI revealed that intramyocardial injections of MSCs not only reduced the scar burden (mass of LV) by 21.8+3.9% (p ⁇ 0.05 vs. placebo and week 12 vs. week 24), but also significantly improved regional contractility, global LV function, ejection fraction, and myocardial blood flow. Importantly, the therapy produced reverse remodeling and reduced the circumferential extent of the infarct scar. This constellation of effects evidences highly effective repair in ischemic cardiomyopathy.
  • Allogeneic Mesenchymal Stem Cells Restore Cardiac Function in Chronic Ischemic Cardiomyopathy Via Trilineage Differentiating Capacity: The hypothesis was tested whether MSC based cardiac repair regenerates the heart via mechanisms comprising long-term engraftment and by differentiation into both myocardial and vascular elements. Allogeneic MSCs were generated from a male swine donor, and administered sex mismatched cells by transendocardial injection into female swine 12 weeks post-MI. Animals were followed with serial MRI, and 12 weeks later the hearts were collected for immunohistological evaluation.
  • Y-chromosome Y pos
  • MSCs engrafted in infarct and border zones and differentiated into cardiomyocytes as ascertained by co-localization with GATA-4, Nkx2.5 and ⁇ -sarcomeric actin markers.
  • Y pos MSCs exhibited vascular smooth muscle and endothelial cell differentiation, contributing to large and small vessel formation. The number of cells engrafting correlated with the functional changes that occurred.
  • phase I/II clinical trial [called “Prospective Randomized Study of CD271 + Cell Therapy in Patients Undergoing Cardiac Surgery” (PROMETHEUS-II)]. It is a double-blind, randomized, placebo-controlled trial, comparing CD271 + cells to placebo in 15 patients undergoing bypass surgery.
  • the endpoints of this trial are safety and efficacy.
  • the efficacy endpoint utilizes cardiac magnetic resonance imaging (MRI) to assess myocardial infarct size, regional and global left ventricular (LV) function, and myocardial perfusion.
  • MRI cardiac magnetic resonance imaging
  • LV left ventricular
  • a biorepository of patient bone marrow and circulating blood samples is also collected to determine biomarkers which may predict successful response to therapy.
  • the CD271 + cell products used in this trial are isolated in the Cell Manufacturing Program facilities at the University of Miami.
  • the CD271 + cell to use in this trial is a bone marrow derived adult stem cell that is a precursor to the MSC.
  • TAC-HFT Transendocardial Autologous Cells in Ischemic Heart Failure Trial
  • the ambulatory ECG recordings demonstrated a reduced number of ventricular tachycardia episodes in hMSC treated patients compared to placebo. Echocardiography in anterior MI patients showed improved ejection fraction in hMSC treated patients. Importantly, this trial provided pivotal safety data for the use of allogeneic MSCs in ischemic heart disease.
  • MSC injection via catheter into infarcted tissue reduces myocardial infarct size, improves global and regional LV function, normalizes cardiac energetics, and restores tissue perfusion (7, 8, 16).
  • CD271 + Cells from Normal Human BM Bone marrow aspirates were obtained from a patient's iliac crest under conscious sedation and local analgesia by an experienced Hematologist/Oncologist. The BM mononuclear cells were isolated using density gradient centrifugation and then the cells were labeled with microbeads attached to an antibody to CD271 and the CD271 + cells isolated on the CliniMACS clinical device (Miltenyi Biotech, Cologne, Germany). The cells were then washed and prepared for infusion. This process takes approximately 4 to 5 hours. All CD271 + cells (1-5 M) were used for patient injection.
  • CD271 + cells were isolated from normal donor bone marrow cells using an indirect labeling of cells with anti CD271-APC and anti-APC Microbeads.
  • the isolated CD271 + cells demonstrated a primitive morphology with a low cytoplasm to nuclear ratio.
  • CD271 + cells When placed into liquid culture, the CD271 + cells formed MSC within 7 days, while whole BM MNCs formed fewer MSC.
  • the generation of MSCs from CD271 + cells was compared to CD271 ⁇ cells and while 170,000 CD271 + cells generated significant MSCs after 7 days of culture in a T162 cm 2 flask, one hundred times more CD271 ⁇ cells (1.7 ⁇ 10 7 cells in a T162 cm 2 flask) failed to generate significant MSCs in 7 day. There were some adherent cells in the culture of CD271 ⁇ cells, however, these appeared to be endothelial cells not MSCs.
  • the CD271 + cells were enriched for CFU-F, with 1 in 222 CD271 + cells forming CFU-F compare to 1 in 12,500 BM MNC. Also the CD271 negative fraction contained very few CFU-F with less than 1 in 100,000 cells.
  • the autologous BM-CD271 + cells or placebo were injected by the cardiac surgeon under direct vision via the epicardium to the area of scar and borderzone. Any areas of bleeding were controlled with pledgeted sutures.
  • Cardiac MRI This group has extensive experience in determining cardiac structure and function by exploiting cutting-edge, non-invasive imaging techniques (17, 19, 20). Cardiac MR was used to evaluate efficacy of cell therapy. Myocardial function, tissue perfusion and non-invasive determination of infarct size, were determined by Harmonic Phase (HARP) Tissue Tagging, gadolinium uptake kinetics, and Delayed Contrast-Enhancement MRI protocols, respectively. Patients lay supine on the magnet table and all images were obtained during a 12-15 heartbeat breathhold at end-expiration, averaging 10-15 sec with adequate rest periods between breath holds (10-15 sec). The imaging protocol first included sagital, axial and oblique scout images to localize the heart.
  • HEP Harmonic Phase
  • Each MRI session is estimated to last 45-60 min.
  • tissue tagging protocols were performed as previously described. The protocol was based upon an ECG triggered fast gradient echo pulse sequence that resulted in 6 mm-tagged separation of the myocardium. This method, yielded quantitative motion and strain parameters on a regional basis that can be used for comparison across subjects at different time points after intervention, thus providing a rapid and repeatable method to assess serial and quantitative LV function.
  • patients received a bolus intravenous injection of 0.2 mmol/kg of gadolinium-DTPA.
  • High-resolution delayed-enhancement images were obtained from eight to ten short-axis cross sections of the LV (ensuring entire cardiac coverage) using an inversion-recovery prepared gated fast gradient echo sequence.
  • the acquired hyper-enhanced regions obtained with this method have been shown to be within 10% of the infarct size measured post-mortem by triphenyl-tetrazolium chloride (TTC) staining, thus signifying this method as a very accurate way to determine infarct size (19).
  • TTC triphenyl-tetrazolium chloride
  • Bone marrow aspirates (25 to 50 ml) were obtained from AllCells LLC (Emeryville, Calif.) under appropriate informed consent and IRB approval.
  • the bone marrow (BM) cells were diluted 1:1 with PBS+1% FCS and layered over ficol to isolate the low density mononuclear cell fraction (MNC).
  • MNC low density mononuclear cell fraction
  • T he MNC were then labeled with CD271 microbeads (Miltenyi Biotech, Cologne, Germany) and the CD271 + cells isolated using a Miltenyi MACS cell selection device (VarioMACS) according to the manufacturers recommended procedures.
  • the CD271 + cells were counted and cytospins prepared for morphological analysis.
  • MSCs Mesenchymal Stem Cells
  • CFU-F Assay The clonegenic potential of BM MNCs, CD271 + and CD271 ⁇ cells were assessed using the colony forming unit-fibroblast (CFU-F) assay.
  • Cells were plated in 35 mm dishes in 2 ml of Mesenchymal Stem Cell Stimulatory Media plus supplements (Stem Cell technologies, Vancouver, Canada). Cells were plated at different cell densities ranging from 10,000 cells per plate up to 1 million cells per plate. Cultures were incubated at 37° C. in 5% CO 2 for 10 days and then stained with Giemsa stain and scored using a dissecting microscope. Typically CFU-F colonies are between 1 and 8 mm in diameter and can be scored macroscopically.
  • CD271 + cells were cultured under non adherent conditions in Teflon bags and the cells harvested and plated in 6-well cell culture dishes (Nunc, Roskilde, Denmark) for differentiation assays.
  • Adipocytic differentiation was induced by culturing these cells in NH AdipoDiff Medium (Miltenyi Biotec Inc., Auburn, Calif., USA) at a concentration of 5 ⁇ 10 4 cells/ml for 2 weeks. Then cells were used for lipid droplet staining using Oil Red O (Sigma-Aldrich, St. Louis, Mo.).
  • Osteogenic differentiation was induced by culturing these cells in NH OsteoDiff Medium (Miltenyi Biotec Inc.) at a concentration of 3 ⁇ 10 4 cells/ml for 3 weeks. Then the cells were stained with SIGMA FAST BCIP/NBT Buffered Substrate Tablet (Sigma) to detect their expression of alkaline phosphatase (AP), an enzyme that is involved in the bone matrix mineralization. Cells that were cultured in alpha-MEM during this period were used as controls.
  • AP alkaline phosphatase
  • ECG electrocardiogram
  • Echocardiography Cardiac function was monitored by Vevo 770 imaging system (VisualSonic Inc., Toronto, Canada) at baseline before surgery, 48 hours, 1, 2, 4 and 8 weeks 8 after infarction and cell injection. Images were recorded under anesthesia with Isoflurane inhalation (1-2%) at heart rates above 400 bpm and body temperatures of (37 ⁇ 10 C). Sonographic parameters of heart structure and anatomy including end diastolic volume (EDV), end systolic volume (ESV) and ejection fraction (EF) were calculated using two dimensional images.
  • EDV end diastolic volume
  • ESV end systolic volume
  • EF ejection fraction
  • Pressure volume loop analysis Eight weeks after cell injection and using the right carotid approach a Millar conductance manometry catheter (SPR 839) (Millar Instruments, TX) progressed to the left ventricle. During the procedure, a diluted 6.25% albumin solution was infused into jugular vein at a rate of 5 ⁇ l/min and anesthesia was achieved with 1-2% Isoflurane inhalation through endotracheal intubation. Pressure volume data were obtained using MPVS Ultra system (Millar Instruments, TX) at baseline and after inferior vena cava (IVC) occlusion. The volume calibration was done using cuvette method and hypertonic saline infusion.
  • Tissue preparation and histopathology After finishing PV loop recordings, hearts were harvested and perfused for 10 minutes with potassium chloride 20 ⁇ M solution and formalin 10% at lml/min through aortic cannulation to fix the hearts in diastole. The hearts were sliced and histology cuts were stained with Mason Trichrome (TM) and H&E or used for immunohistochemistry.
  • TM Mason Trichrome
  • Human specific DNA probes were used (Human Alu Probe, BioGenex, CA) and Fluorescence in Situ Hybridization (FISH) method to trace injected human cells in the mouse tissue (AntiFluorescein-HRP Conjugate, Perkin Elmer, Boston, Mass.). Samples from human fetal heart and PBS injected mice were used as positive and negative controls respectively. The Alu stained slides were scanned with a Zeiss fluorescent laser scanner and the images were analyzed using Mirax viewer software at 20 ⁇ magnification. Images were taken separately and merged using CS3 Adobe Photoshop and positive cells counted in two separate areas of the same cut.
  • FISH Fluorescence in Situ Hybridization
  • Immunohistochemistry The samples were stained with cardiac specific antibodies, alpha sarcomeric actin (a-SA) (Sigma, St. Louis, Mo.), Troponin I (TnI) (Abcam, Cambridge, Mass.), and Connexin 43 (Cx43) (Santa Cruz, Calif.). Laminin (Abcam) staining was used to delineate cells. The stained samples were studied first by immunofluorescent microscopy and then confocal microscope Zeiss LSM710 to prepare images.
  • a-SA alpha sarcomeric actin
  • TnI Troponin I
  • Cx43 Connexin 43
  • CD271 + cells were isolated from normal donor BM cells using an indirect labeling of cells with anti CD271-APC and anti-APC Microbeads.
  • the isolated CD271 + cells demonstrated a primitive morphology with a low cytoplasm to nuclear ratio ( FIG. 1 ).
  • the CD271 + cells When placed into liquid culture, the CD271 + cells formed MSC within 7 days ( FIG. 2A ), while whole BM MNCs formed fewer MSC ( FIG. 2B ).
  • the generation of MSCs from CD271 + cells were compared to CD271 ⁇ cells and while 170,000 CD271+ cells generated significant MSCs after 7 days of culture in a T162 cm2 flask, one hundred times more CD271 ⁇ cells (1.7 ⁇ 10 7 cells in a T162 cm 2 flask) failed to generate significant MSCs in 7 days ( FIG. 2C ). As shown in FIG. 2C there were some adherent cells in the culture of CD271 ⁇ cells, however, these appeared to be endothelial cells not MSCs.
  • the CD271 ⁇ cells were enriched for CFU-F ( FIG. 3 ), with 1 in 250 CD271 + cells forming CFU-F compare to 1 in 12,500 BM MNC. Also the CD271 negative fraction contained very few CFU-F with less than 1 in 100,000 cells.
  • CD271 + cells were grown in non adherent culture conditions in Teflon bags for 1 to 3 months and the early stages of growth are presented in FIG. 4 .
  • the media for these cultures consisted of alpha-MEM+20% FCS with 20 ng/ml recombinant human basic fibroblast growth factor (rhbFGF).
  • rhbFGF human basic fibroblast growth factor
  • the bags were massaged to detach adherent cells and the media changed weekly.
  • the clusters of cells continued to proliferate and form spheres as shown in FIG. 4 with large spheres developing by day 21.
  • FIG. 5 shows that the majority expressed CD105, a typical MSC marker.
  • CD271 + cells grown under non adherent conditions Adipocytic and osteoblastic differentiation potential of the CD271 + cells grown under non adherent conditions was also evaluated.
  • Cells were harvested from the culture bags and plated in 6-well cell culture dishes (Nunc, Roskilde, Denmark) for differentiation assays. As shown in FIG. 6 , the cultured CD271 ⁇ cells differentiated into both adipocytes and osteoblasts.
  • Cells were also cultured under typical MSC culture conditions, namely culture in tissue culture flasks with alpha MEM media plus 20% FCS.
  • typical MSC culture conditions namely culture in tissue culture flasks with alpha MEM media plus 20% FCS.
  • the spheres shown in FIG. 4 attached to the plastic surface of the flasks and typical MSCs grew from the spheres and formed confluent cultures after 1 to 2 weeks.
  • CD271 + cells cultured under non adherent conditions form spheres of cells which are CD105 + , have adipocyte and osteoblast potential and form plastic adherent MSC like cells consistent with MSC properties.
  • EDV ventricular volume at end diastole
  • ESV-CD 271 treated animals had a significantly lower ESV (81.5 ⁇ 12.4 ⁇ l) compared to control animals (119.7 ⁇ 17.8 ⁇ l, p ⁇ 0.0001) and low dose MSC treated animals (117.1 ⁇ 13.4 ⁇ l, p ⁇ 0.0001) but not the high dose MSC treated animals (91.2 ⁇ 12.6 ⁇ l).
  • Human cells were also detected in the hearts of animals injected with the high dose of MSCs and again these cells were either embedded in the vascular wall or between host cardiomyocytes. There was no detection of Alu staining cardiomyocytes or expression of cardiac markers in Alu positive cells in the hearts of animals injected with the high dose of MSCs.
  • CD271 + cells were present at a low frequency in BM cells consisting of approximately 0.3% of the BM MNC population. Using Miltenyi MACS selection reagents and devices, a highly purified population of CD271 + cells were isolated having greater than 90% purity with an average recovery of 4.2 ⁇ 10 5 cells from a 25 cc BM aspirate. The morphology of the CD271 + cells demonstrated a primitive phenotype of uniform blast cells consisting of a low cytoplasm to nuclear ratio. The data confirmed the enrichment of MSC forming cells in the CD271 + population with a 60 fold higher level of CFU-F in CD271 ⁇ cells compared to BM MNCs.
  • CD271 negative population was greatly reduced at 1 in 100,000 cells compared to 1 in 12,500 in the BM MNC population and 500 fold lower than the CD271 + population.
  • MSCs generated from CD271 + cells by adherence to plastic flasks, presented identical in vitro properties as MSCs generated from BM MNCs with expression of CD105 + and capable of forming adipocytes and osteoblasts in vitro.
  • Previous studies have demonstrated that MSC potential resides in the CD271 + CD45 ⁇ cell population.
  • phenotypic analysis demonstrated that the CD271 + cells were negative for typical MSC markers including CD105 and CD90, however, after culture the cells expressed classic MSC markers, CD105, CD90 and CD73.
  • CD271 expression inhibits differentiation of MSCs to osteogenic, adipogenic, chondrogenic and myogenic lineages.
  • the data herein, are consistent with the literature and support the proposal that CD271 + cells are a stem cell population which is a precursor of MSCs and that differentiation of CD271 + cells to plastic adherent MSCs results in commitment to adipogenic, chondrogenic and osteogenic lineages.
  • MSCs have the bone surface to act as a substrate for adherent cell growth, however, no equivalent substrate exists in the heart tissue. Therefore culture conditions were developed for expansion of MSCs under non adherent conditions (NA-MSCs).
  • CD271 + cells were cultured in teflon bags which minimized the adherence of cells and regular massaging of the bags maintained the cells in suspension.
  • MSCs proliferated in the bags forming spheres of cells with MSC properties, forming classic adherent MSC like cells when placed in standard plastic flasks and expressing CD105.
  • the NA-MSCs also differentiated into adipocytes and chondrocytes under appropriate culture conditions.
  • the osteogenic capacity of the NA-MSCs is currently being evaluated.
  • the CD271 ⁇ cells can be induced to express cardiac specific genes when cultured in the presence of media conditioned by murine heart derived stromal cells. Media conditioned by stromal cells derived from human fetal heart tissue also stimulates cardiac gene expression.
  • a number of groups have evaluated the potential of several bone marrow derived populations including mononuclear cells, CD34 ⁇ cells, CD133 + cells and MSCs.
  • the potential of BM derived MSCs was evaluated in large animal preclinical studies and it was demonstrated that MSCs can engraft in and adjacent to chronic infarct scars and stimulate significant myocardial recovery including substantial reductions in infarct size, reverse remodeling of injured hearts, and increases in both myocardial contractility and tissue perfusion.
  • BM derived CD271 + cells represent an ideal autologous cell source for these patients.
  • the CD271 + cells can be isolated from a BM aspirate within 4 to 5 hours providing a readily available cell population for treatment of CABG patients who require urgent surgery.
  • the CD271 + cells Compared to MSCs isolated by plastic adherence, the CD271 + cells have a greater in vitro cardiac differentiation potential and treatment of mice following an MI result in greater improvement in cardiac function.
  • the CD271 + cells can be isolated rapidly using magnetic cell selection, therefore could be a potential cell source for patients requiring bypass surgery.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cardiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Hospice & Palliative Care (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Materials For Medical Uses (AREA)
US13/819,154 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair Abandoned US20140023621A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/819,154 US20140023621A1 (en) 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US37766110P 2010-08-27 2010-08-27
US13/819,154 US20140023621A1 (en) 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair
PCT/US2011/049542 WO2012027740A1 (en) 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/049542 A-371-Of-International WO2012027740A1 (en) 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/720,373 Continuation US20180214486A1 (en) 2010-08-27 2017-09-29 Bone marrow derived cd271 precursor cells for cardiac repair

Publications (1)

Publication Number Publication Date
US20140023621A1 true US20140023621A1 (en) 2014-01-23

Family

ID=45723836

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/819,154 Abandoned US20140023621A1 (en) 2010-08-27 2011-08-29 Bone marrow derived cd271 precursor cells for cardiac repair
US15/720,373 Abandoned US20180214486A1 (en) 2010-08-27 2017-09-29 Bone marrow derived cd271 precursor cells for cardiac repair
US17/811,418 Pending US20230071551A1 (en) 2010-08-27 2022-07-08 Bone Marrow Derived CD271 Precursor Cells for Cardiac Repair

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/720,373 Abandoned US20180214486A1 (en) 2010-08-27 2017-09-29 Bone marrow derived cd271 precursor cells for cardiac repair
US17/811,418 Pending US20230071551A1 (en) 2010-08-27 2022-07-08 Bone Marrow Derived CD271 Precursor Cells for Cardiac Repair

Country Status (14)

Country Link
US (3) US20140023621A1 (es)
EP (1) EP2608797B1 (es)
JP (1) JP6061345B2 (es)
KR (1) KR101920891B1 (es)
CN (1) CN103221058B (es)
AU (1) AU2011293144B2 (es)
BR (1) BR112013004700B1 (es)
CA (1) CA2842181C (es)
ES (1) ES2556961T3 (es)
IL (1) IL224917A (es)
MX (1) MX340185B (es)
NZ (1) NZ608734A (es)
SG (1) SG188305A1 (es)
WO (1) WO2012027740A1 (es)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160158953A1 (en) * 2014-12-03 2016-06-09 Urschel Laboratories, Inc. Machines and methods for cutting products and impellers therefor
CN110302212A (zh) * 2019-07-17 2019-10-08 陶正博 一种用于治疗急性心肌梗死的细胞制剂及其制备方法
WO2020069215A1 (en) * 2018-09-27 2020-04-02 Biocardia, Inc. Bone marrow derived neurokinin-1 receptor positive (nk1r+) precursor cells for therapeutic applications

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2012279995C1 (en) 2011-07-06 2019-10-24 Cell Therapy Limited Progenitor cells of mesodermal lineage
JP6090735B2 (ja) * 2012-03-02 2017-03-08 国立大学法人山口大学 消化器系がん幹細胞を培養するための無血清培地、及びそれを用いた消化器系がん幹細胞の増殖方法
WO2013163539A1 (en) * 2012-04-26 2013-10-31 The Cohen Mcniece Foundation Human fetal heart derived stromal cells for treatment of patients following a myocardial infarction
WO2014022376A2 (en) 2012-08-01 2014-02-06 United Therapeutics Corporation Treatment of pulmonary arterial hypertension with prostacyclin-treated endothelial progenitor cells
JP6901823B2 (ja) 2012-08-01 2021-07-14 ユナイテッド セラピューティクス コーポレイション 間葉系幹細胞による肺動脈性高血圧症の処置
EP3878452B1 (en) * 2013-01-09 2023-09-20 United Therapeutics Corporation Treatment of vasculopathy with prostacyclin and mesenchymal stem cells
BR112017012911B1 (pt) * 2014-12-23 2021-10-05 Mesoblast International Sàrl Método para tratamento de insuficiência cardíaca
JP6777840B2 (ja) * 2015-12-25 2020-10-28 北海道公立大学法人 札幌医科大学 脳梗塞の治療用医薬
CN106474456A (zh) * 2016-10-18 2017-03-08 广州赛莱拉干细胞科技股份有限公司 一种细胞制剂及其制备方法和应用
IL266175B2 (en) 2016-10-24 2024-01-01 United Therapeutics Corp Enhancement of immunomodulatory properties of MSC using treprostinil

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009060840A (ja) * 2007-09-06 2009-03-26 Keio Gijuku ヒト間葉系幹細胞濃縮方法
WO2009140452A2 (en) * 2008-05-15 2009-11-19 University Of Miami Isolation of stem cell precursors and expansion in non-adherent conditions
US20110305673A1 (en) * 2008-11-12 2011-12-15 The University Of Vermont And State Agriculture College Compositions and methods for tissue repair

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Herrmann et al., Postinfarct intramyocardial injection of mesenchymal stem cells pretreated with TGF-a improves acute myocardial function, Am. J. Physiol Regul. Intergr. Comp. Physiol. 299: R371-R378, 2010 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160158953A1 (en) * 2014-12-03 2016-06-09 Urschel Laboratories, Inc. Machines and methods for cutting products and impellers therefor
WO2020069215A1 (en) * 2018-09-27 2020-04-02 Biocardia, Inc. Bone marrow derived neurokinin-1 receptor positive (nk1r+) precursor cells for therapeutic applications
US11642377B2 (en) 2018-09-27 2023-05-09 Biocardia, Inc. Bone marrow derived neurokinin-1 receptor positive (NK1R+) mesenchymal stem cells for therapeutic applications
CN110302212A (zh) * 2019-07-17 2019-10-08 陶正博 一种用于治疗急性心肌梗死的细胞制剂及其制备方法

Also Published As

Publication number Publication date
US20180214486A1 (en) 2018-08-02
CN103221058B (zh) 2015-07-29
JP6061345B2 (ja) 2017-01-18
KR101920891B1 (ko) 2018-11-21
JP2013542178A (ja) 2013-11-21
AU2011293144B2 (en) 2015-04-23
KR20130106381A (ko) 2013-09-27
EP2608797B1 (en) 2015-09-23
BR112013004700B1 (pt) 2020-12-08
MX2013002381A (es) 2013-04-29
CN103221058A (zh) 2013-07-24
NZ608734A (en) 2014-09-26
WO2012027740A1 (en) 2012-03-01
AU2011293144A1 (en) 2013-04-18
CA2842181C (en) 2021-01-05
ES2556961T3 (es) 2016-01-21
EP2608797A4 (en) 2014-01-22
IL224917A (en) 2017-02-28
US20230071551A1 (en) 2023-03-09
EP2608797A1 (en) 2013-07-03
MX340185B (es) 2016-06-29
CA2842181A1 (en) 2012-03-01
BR112013004700A2 (pt) 2017-07-04
SG188305A1 (en) 2013-04-30

Similar Documents

Publication Publication Date Title
US20230071551A1 (en) Bone Marrow Derived CD271 Precursor Cells for Cardiac Repair
López et al. Wharton's jelly or bone marrow mesenchymal stromal cells improve cardiac function following myocardial infarction for more than 32 weeks in a rat model: a preliminary report
US8343479B2 (en) Methods and compositions for the repair and/or regeneration of damaged myocardium
US20200016210A1 (en) Methods of Reducing Teratoma Formation During Allogeneic Stem Cell Therapy
JP7419490B2 (ja) 標的臓器の細胞外構成成分を使用する治療用細胞の作製
US20210145893A1 (en) Compositions to amplify cardiac stem cells in vitro and in viv
JPWO2014027684A1 (ja) 心筋梗塞の修復再生を誘導する多能性幹細胞
JP7082372B2 (ja) 肺線維症治療剤、ptprr発現促進剤及び肺線維症治療用キット
JP2023103416A (ja) 拡張型心筋症治療剤
JP7108541B2 (ja) 疾患治療剤調製用キット、疾患治療剤及び疾患治療剤の調製方法
US20240082310A1 (en) Bone marrow derived neurokinin-1 receptor positive (nk1r+) mesenchymal stem cells for therapeutic applications
JP5894071B2 (ja) 心臓組織由来細胞
EP2205251B1 (en) A method to amplify cardiac stem cells in vitro and in vivo
US11963983B2 (en) Methods of cardiac repair

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF MIAMI, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HARE, JOSHUA M.;MCNIECE, IAN K.;SIGNING DATES FROM 20140113 TO 20140114;REEL/FRAME:031987/0023

AS Assignment

Owner name: HARE, JOSHUA M, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UNIVERSITY OF MIAMI;REEL/FRAME:039235/0554

Effective date: 20160613

STCB Information on status: application discontinuation

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