US20130302291A1 - Compositions and methods for cell transplantation - Google Patents

Compositions and methods for cell transplantation Download PDF

Info

Publication number
US20130302291A1
US20130302291A1 US13/981,720 US201213981720A US2013302291A1 US 20130302291 A1 US20130302291 A1 US 20130302291A1 US 201213981720 A US201213981720 A US 201213981720A US 2013302291 A1 US2013302291 A1 US 2013302291A1
Authority
US
United States
Prior art keywords
cells
thrombin inhibitor
liver
heparin
cell
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
US13/981,720
Other languages
English (en)
Inventor
Xavier Stephenne
Etienne Sokal
Mustapha Najimi
Stephane Eeckhoudt
Cedric Hermans
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.)
Universite Catholique de Louvain UCL
Original Assignee
Universite Catholique de Louvain UCL
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 Universite Catholique de Louvain UCL filed Critical Universite Catholique de Louvain UCL
Assigned to UNIVERSITE CATHOLIQUE DE LOUVAIN reassignment UNIVERSITE CATHOLIQUE DE LOUVAIN ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HERMANS, CEDRIC, SOKAL, ETIENNE, STEPHENNE, Xavier, NAJIMI, MUSTAPHA, EECKHOUDT, STEPHANE
Publication of US20130302291A1 publication Critical patent/US20130302291A1/en
Assigned to PROMETHERA BIOSCIENCES S.A. reassignment PROMETHERA BIOSCIENCES S.A. LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: SOPARTEC SA, UNIVERSITE CATHOLIQUE DE LOUVAIN
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
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/726Glycosaminoglycans, i.e. mucopolysaccharides
    • A61K31/727Heparin; Heparan
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/737Sulfated polysaccharides, e.g. chondroitin sulfate, dermatan sulfate
    • 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
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • A61K38/58Protease inhibitors from animals; from humans from leeches, e.g. hirudin, eglin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • 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

Definitions

  • the present invention relates to the field of tissue regeneration in general and cell transplantation in particular.
  • the present invention is directed at compositions and methods for improving cell transplantation and particularly for inhibiting procoagulant activity associated with cell transplantation.
  • organ transplantations Various conditions caused by diseased or otherwise damaged or functionally impaired organs may be treated by organ transplantations.
  • transplantation of heart, kidneys, liver, lungs, pancreas, intestine, and thymus can routinely be performed with a reasonable rate of success.
  • a major drawback in organ transplantation however remains the need to find a compatible donor for each recipient patient, since incompatibility between the donor and recipient may result in rejection of the transplanted organ.
  • Transplant rejection can be reduced through serotyping to determine the most appropriate donor-recipient match and through the use of immunosuppressant drugs, although the suitability of these approaches may be diminished due to the medical urgency in some cases.
  • life-long use of immunosuppressant drugs places a burden on the recipient patient in terms of side effects and compliance.
  • the cells or cell cultures are selected such as to maximise the tissue compatibility between the patient and the administered cells, thereby reducing the chance of rejection of the administered cells by patient's immune system (graft vs. host rejection).
  • the cells may be typically selected which have either identical HLA haplotypes (including one or preferably more HLA-A, HLA-B, HLA-C, HLA-D, HLA-DR, HLA-DP and HLA-DQ; preferably one or preferably all HLA-A, HLA-B and HLA-C) to the patient, or which have the most HLA antigen alleles common to the patient and none or the least of HLA antigens to which the patient contains pre-existing anti-HLA antibodies.
  • HLA haplotypes including one or preferably more HLA-A, HLA-B, HLA-C, HLA-D, HLA-DR, HLA-DP and HLA-DQ; preferably one or preferably all HLA-A, HLA-B and HLA-C
  • Tissue regeneration procedures by means of cell transplantation may be executed using a large variety of cell sources, and commonly using cells having proliferative capacity. For instance, in various human inborn metabolic diseases liver cell transplantation can restore at least some degree of metabolic control.
  • intraportal transplantation of pancreatic islets offers improved glycaemic control and insulin independence in type 1 diabetes mellitus.
  • pluripotent stem cells capable of differentiating into a plethora of cell lineages, or progenitor cells committed to one or a few cell lineages (multipotent) and displaying varying degrees of differentiation may be used as a cell source for cell transplantation.
  • procoagulant activity of certain transplanted cells has been reported to cause graft loss and intraportal thrombotic events (Beuneu et al. Diabetes, 2004, vol. 53, 1407-11; Moberg et al. Lancet, 2002, vol. 360, 2039-45).
  • Procoagulant activity has been also observed in isolated primary hepatocytes (Stéphenne et al. Liver Transpl., 2007, vol. 13, 599-606).
  • procoagulant activity previously reported for isolated primary cells such as hepatocytes and islet cells is also observed for stem cells and progenitor cells such as mesenchymal stem cells.
  • the procoagulant activity of stem and progenitor cells may be of concern for transplantation of these cells, in particular may cause undesired bloodstream modifications, loss of the transplanted cells, reduction of the cell engraftment potential and/or thrombotic events.
  • this procoagulant activity cannot be controlled by unfractionated heparin, the conventional anticoagulant for hepatocyte transplantation.
  • the inventors therefore investigated manners to counteract the procoagulant activity of transplanted cells and found that concomitant or associated administration of cells having procoagulant activity with an antithrombin activator and a thrombin inhibitor provides a particularly effective and safe combination for preventing deleterious procoagulant effects.
  • the combination of the antithrombin activator together with the thrombin inhibitor can advantageously prevent cell therapy-induced thrombosis and thrombosis associated complications (e.g., local thrombosis and induction of local inflammation). Accordingly, the inventors realised a particularly advantageous and even synergistic combination therapy and clinical protocols useful for reducing procoagulant activity of transplanted cells, in particular stem and progenitor cells.
  • an aspect relates to a combination comprising cells having procoagulant activity, at least one antithrombin activator, and at least one thrombin inhibitor.
  • Another aspect relates to a combination comprising at least one antithrombin activator, at least one thrombin inhibitor, and cells selected from the group comprising or consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, and liver myofibroblasts, more preferably selected from adult liver progenitor cells and liver myofibroblasts.
  • cells selected from the group comprising or consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, and liver myofibroblasts, more preferably selected from adult liver progenitor cells and liver myofibroblasts.
  • the combination may be configured for separate, simultaneous or sequential in any order administration of the cells, at least one antithrombin activator, and at least one thrombin inhibitor.
  • the cells, at least one antithrombin activator, and/or at least one thrombin inhibitor in said combination may be admixed or may be separate.
  • a method for producing said combination comprising combining the cells, at least one antithrombin activator, and at least one thrombin inhibitor.
  • the individual constituents of the combination may be configured for separate, simultaneous or sequential in any order administration to a subject, or may be administered to a subject separately, simultaneously or sequentially in any order.
  • the cells, the at least one antithrombin activator and the at least one thrombin inhibitor may all be included in the cell suspension to be administered.
  • the cells and the at least one antithrombin activator may be included in the cell suspension to be administered, whereas the at least one thrombin inhibitor may be held separate from said cell suspension and to be administered to the subject simultaneously or sequentially with said cell suspension.
  • the cells and the at least one thrombin inhibitor may be included in the cell suspension to be administered, whereas the at least one antithrombin activator may be held separate from said cell suspension and to be administered to the subject simultaneously or sequentially with said cell suspension.
  • both the at least one antithrombin activator and the at least one thrombin inhibitor may be held separate from the cell suspension and to be administered to the subject simultaneously or sequentially with the cell suspension, and simultaneously or sequentially with one another.
  • composition comprising (a) a combination comprising cells having procoagulant activity, at least one antithrombin activator, at least one thrombin inhibitor and (b) one or more pharmaceutically acceptable excipients.
  • composition comprising (a) a combination comprising at least one antithrombin activator, at least one thrombin inhibitor, and cells selected from the group comprising or consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, and liver myofibroblasts, more preferably selected from adult liver progenitor cells and liver myofibroblasts, and (b) one or more pharmaceutically acceptable excipients.
  • a combination comprising at least one antithrombin activator, at least one thrombin inhibitor, and cells selected from the group comprising or consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, and liver myofibroblasts, more preferably selected from adult liver progenitor cells and liver myofibroblasts, and (b) one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition may be configured for separate, simultaneous or sequential in any order administration of the cells, at least one antithrombin activator, and at least one thrombin inhibitor.
  • the cells, at least one antithrombin activator, and/or at least one thrombin inhibitor in said pharmaceutical composition may be admixed or may be separate.
  • a method for producing said pharmaceutical composition comprising admixing the cells, at least one antithrombin activator, and at least one thrombin inhibitor, each separately or in an admixture, with the one or more pharmaceutically acceptable excipients.
  • kit of parts or an article of manufacture comprising a combination comprising cells having procoagulant activity, at least one antithrombin activator, and at least one thrombin inhibitor, and optionally further comprising one or more pharmaceutically acceptable excipients.
  • kit of parts or an article of manufacture comprising a combination comprising at least one antithrombin activator, at least one thrombin inhibitor, and cells selected from the group comprising or consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, and liver myofibroblasts, more preferably selected from adult liver progenitor cells and liver myofibroblasts, and optionally further comprising one or more pharmaceutically acceptable excipients.
  • mesenchymal stem cells preferably bone marrow mesenchymal stem cells
  • skin fibroblasts fibroblasts
  • liver myofibroblasts more preferably selected from adult liver progenitor cells and liver myofibroblasts, and optionally further comprising one or more pharmaceutically acceptable excipients.
  • the kit of parts or article of manufacture may be configured for separate, simultaneous or sequential in any order administration of the cells, at least one antithrombin activator, and at least one thrombin inhibitor.
  • the cells, at least one antithrombin activator, and/or at least one thrombin inhibitor in said kit of parts or article of manufacture may be admixed or may be separate, particularly may be separate such as for example contained in separate containers.
  • a method for producing said kit of parts or article of manufacture comprising including the cells, at least one antithrombin activator, and at least one thrombin inhibitor, and optionally one or more pharmaceutically acceptable excipients, in a kit of parts or an article of manufacture.
  • the kit of parts or article of manufacture for use in any one and each of the herein-described indications.
  • any cells as described herein such as particularly cells having procoagulant activity, at least one antithrombin activator and at least one thrombin inhibitor, in any one and each of the above-described indications.
  • a further aspect relates to a combination comprising at least one antithrombin activator and at least one thrombin inhibitor.
  • the combination may be configured for separate, simultaneous or sequential in any order administration of the at least one antithrombin activator and at least one thrombin inhibitor.
  • the at least one antithrombin activator and at least one thrombin inhibitor in said combination may be admixed or may be separate.
  • a method for producing said combination comprising combining the at least one antithrombin activator and at least one thrombin inhibitor.
  • a pharmaceutical composition comprising a combination comprising at least one antithrombin activator, at least one thrombin inhibitor and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition may be configured for separate, simultaneous or sequential in any order administration of the at least one antithrombin activator and at least one thrombin inhibitor.
  • the at least one antithrombin activator and at least one thrombin inhibitor in said pharmaceutical composition may be admixed or may be separate.
  • a method for producing said pharmaceutical composition comprising admixing the at least one antithrombin activator and at least one thrombin inhibitor, each separately or in an admixture, with the one or more pharmaceutically acceptable excipients.
  • kit of parts or an article of manufacture comprising a combination comprising at least one antithrombin activator and at least one thrombin inhibitor, and optionally further comprising one or more pharmaceutically acceptable excipients.
  • the kit of parts or article of manufacture may be configured for separate, simultaneous or sequential in any order administration of the at least one antithrombin activator and at least one thrombin inhibitor.
  • the at least one antithrombin activator and at least one thrombin inhibitor in said kit of parts or article of manufacture may be admixed or may be separate, particularly may be separate such as for example contained in separate containers.
  • kits of parts or article of manufacture comprising including the at least one antithrombin activator and at least one thrombin inhibitor, and optionally one or more pharmaceutically acceptable excipients, in a kit of parts or an article of manufacture. Also provided is the kit of parts or article of manufacture for use in any one and each of the herein-described indications.
  • any of the above methods may comprise the steps of: (a) preparing a composition comprising a cell suspension of the cells as described herein such as particularly cells having procoagulant activity in an aqueous solution containing the at least one antithrombin activator; (b) preparing an aqueous solution containing the at least one thrombin inhibitor (i.e., distinct from or separate from composition (a)); and (c) administering the composition as defined in (a) and the solution as defined in (b) simultaneously, separately or sequentially to the subject.
  • composition comprising a cell suspension of the cells as described herein such as particularly cells having procoagulant activity in an aqueous solution containing the at least one antithrombin activator is to be prepared;
  • an aqueous solution containing the at least one thrombin inhibitor is to be prepared (i.e., distinct from or separate from composition (a)); and
  • the composition as defined in (a) and the solution as defined in (b) is to be administered simultaneously, separately or sequentially to a subject.
  • an arrangement comprising a surgical instrument or device for administration of a composition to a subject, such as for example systemically, topically, within an organ or tissue (e.g., portal vein of the liver, spleen, pancreas, liver, kidney capsule, peritoneum and omental pouch), and further comprising the combination or pharmaceutical composition comprising cells as described herein such as particularly procoagulant cells as taught herein, wherein the arrangement is adapted for administration of said combination or pharmaceutical composition for example systemically, topically, within an organ or tissue.
  • a suitable surgical instrument may be capable of injecting a liquid composition comprising the combination or pharmaceutical composition taught herein, such as systemically, topically, within an organ or tissue.
  • Cells having procoagulant activity as intended throughout this specification encompass any cells which are capable of activating the coagulation cascade and to induce coagulation or clot formation.
  • Procoagulant activity may be conveniently determined using any known coagulation test, such as without limitation thromboelastometry.
  • cells may be denoted as having procoagulant activity in the sense of the present invention when, in a standard thromboelastometry test, the cells display clotting time (CT) significantly shorter (p ⁇ 0.05 applying a suitable test of statistical significance) than a negative control without addition of cells.
  • CT clotting time
  • thromboelastometry represents a standard laboratory technique, for reasons of further guidance suitable thromboelastometry for testing the procoagulant nature of the cells as intended herein may be as follows:
  • ROTEM® delta analyser (Pentapharm, Kunststoff, Germany).
  • ROTEM® assesses the kinetics and quality of clot formation and clot lysis in real-time.
  • the clotting time (CT) is defined as the period of time from the start of the analysis until the start of clot formation, until the 2 mm amplitude is reached.
  • CT clotting time
  • 300 ⁇ l of whole blood is pipetted into a cup pre-warmed at 37° C. Suspended cells (5 ⁇ 10exp5) are subsequently added to whole blood (negative control: equal volume of suspension medium without any suspended cells).
  • TF tissue factor
  • Owren buffer such as obtainable from Clin-Tech Ltd, UK
  • cells as intended herein such as particularly cells having procoagulant activity may be of any origin and/or differentiation state.
  • the cells as intended herein such as cells having procoagulant activity are selected from the group consisting of stem cells and progenitor cells. More preferably, the cells as intended herein such as cells having procoagulant activity are mesenchymal stem cells. Also preferably, the cells as intended herein such as cells having procoagulant activity are adult liver-derived progenitor or stem cells.
  • the cells as intended herein such as cells having procoagulant activity are adult-derived human liver stem cells as generally described in WO 2007/071339; more particularly, human progenitor or stem cells originated from adult liver which express alpha-smooth muscle actin (ASMA) and albumin (ALB) and do not express cytokeratin-19 (CK-19) as described therein; even more particularly, human progenitor or stem cells originated from adult liver which express CD90, CD73, CD44, vimentin, ASMA and ALB and optionally express CYP3A4 and do not express CK-19 as described therein; yet more particularly, adult-derived human liver stem cells (ADHLSC) as described by Najimi et al., Cell Transplant, 2007, vol.
  • ADHLSC adult-derived human liver stem cells
  • the cells as intended herein such as cells having procoagulant activity are non-oval adult human liver-derived pluripotent progenitor cells as generally described in WO 2006/126236; more particularly, a non-oval human liver pluripotent progenitor cell line isolated from adult tissue which expresses hepatic cell markers and which is capable of differentiating into mature liver cells, insulin-producing cells, osteogenic cells and epithelial cells, or also particularly, a non-oval human liver pluripotent progenitor cell line isolated from adult tissue which expresses hepatic cell markers and which is capable of differentiating into mature liver cells, insulin-producing cells, osteogenic cells and endothelial cells, as described therein; even more particularly human liver stem cells (HLSC) as described by Herrera et al. Stem Cells, 2006, vol. 24, 2840-50.
  • HLSC human liver stem cells
  • tissue factor also known as platelet tissue factor, factor III, thrombokinase or CD142
  • the cells having procoagulant activity express tissue factor.
  • procoagulant cells as used herein may comprise a procoagulant activity component independent of the expression of tissue factor (TF) by the cells. More specifically, such procoagulant cells will at least partly (e.g., only partly or wholly) retain their procoagulant activity as measured by thromboelastometry in Factor VII deficient plasma, or as measured by thromboelastometry in blood or normal plasma when TF activity is blocked, such as by pre-incubation of cells with anti-TF antibody.
  • the measurable procoagulant activity of cells in factor VII deficient plasma may at least in part also be related to residual small amounts of factor VII.
  • procoagulant cells as used herein may comprise a procoagulant activity component resistant to heparin. More specifically, such procoagulant cells will at least partly (e.g., only partly or wholly) retain their procoagulant activity as measured by thromboelastometry in the presence of heparin alone (i.e., in the absence of thrombin activator), such as particularly in the presence of unfractionated heparin, enoxaparin or fondaparinux, particularly at concentrations of 10 Ul/ml, 1 Ul/ml and 0.34 mg/l, respectively. Hence, in further preferred embodiments, procoagulant cells as used herein may comprise a procoagulant activity component independent of the expression of tissue factor (TF) by the cells and may comprise a procoagulant activity component resistant to heparin.
  • tissue factor TF
  • An antithrombin activator as intended throughout this specification encompasses any agent capable of increasing the binding of antithrombin to any one or more of its targets.
  • the antithrombin activator is selected from the group consisting of unfractionated heparin and low molecular weight heparin, preferably unfractionated heparin.
  • a thrombin inhibitor as intended throughout this specification is an agent capable of directly binding to thrombin and inhibiting or preventing thrombin-mediated fibrinogen activation.
  • the thrombin inhibitor is selected from the group consisting of bivalirudin and hirudin, preferably bivalirudin.
  • bivalirudin has a comparably short half live of about 35 to about 40 minutes, thereby allowing for a prompt return of a subject to a normal haemostasis status.
  • FIG. 1 Clotting time (CT) essayed by ROTEM after recalcification, with and without added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of cells suspended in human albumin 5%.
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells (black)
  • A human albumin 5%.
  • ALDSC vs hepatocytes Mann-Whitney test *p ⁇ 0.05
  • ALDSC vs hepatocytes vs control Kruskal-Wallis test ***p ⁇ 0.001.
  • FIG. 2 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of cells suspended in human albumin 5% and heparin 10 Ul/ml (hepar).
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells (black)
  • A human albumin 5%.
  • ALDSC vs hepatocytes Mann-Whitney test **p ⁇ 0.01;
  • ALDSC vs hepatocytes vs control Kruskal-Wallis test ***p ⁇ 0.001.
  • FIG. 3 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ l), of plasma (300 ⁇ l) obtained after 30 min incubation of citrated blood in presence or not of cells suspended in human albumin 5% and heparin (hepar) (10-50-100 Ul/ml).
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells (black)
  • A human albumin 5%.
  • FIG. 4 Clotting time (CT) essayed by ROTEM, after recalcification, with and without added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of cells.
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells (black)
  • A human albumin 5%
  • hir, hir2, hir5 hirudin at 1, 2, 5-fold the bolus concentration in clinical use (0.4 mg/kg)
  • hepar heparin 10 Ul/ml in cell suspension
  • Control whole blood.
  • FIG. 5 Clotting time (CT) essayed by ROTEM after recalcification, with and without added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of cells.
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells s (black)
  • A human albumin 5%
  • biv, biv2 bivalirudin at 1.2-fold the bolus concentration in clinical use (0.75 mg/kg);
  • hepar heparin 10 Ul/ml in cell suspension; Control: whole blood.
  • ALDSC vs ALDSC biva Mann-Whitney test **p ⁇ 0.01; A vs ALDSC biva: Mann-Whitney test **p ⁇ 0.01; ALDSC biva vs ALDSC biva hepar: Mann-Whitney test **p ⁇ 0.01; ALDSC hepar vs ALDSC biva hepar: Mann-Whitney test ***p ⁇ 0.001; ALDSC vs ALDSC biva vs ALDSC biva hepar: Kruskal-Wallis test ***p ⁇ 0.001.
  • FIG. 6 Clotting time (CT) essayed by ROTEM after recalcification, without added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of cells.
  • Hep Hepatocytes (white)
  • ALDSC Adult-derived human liver mesenchymal stem cells (black)
  • BMMC bone marrow mesenchymal stem cells
  • BMHC bone marrow haematopoietic stem cells
  • A Albumin.
  • ALDSC vs hepatocytes Mann-Whitney test *p ⁇ 0.05;
  • a vs ALDSC Mann-Whitney test ***p ⁇ 0.001
  • FIG. 7 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ l), of citrated whole blood (300 ⁇ l) in presence or not of bone marrow haematopoietic stem cells suspended in human albumin 5% and heparin 10 Ul/ml (hepar).
  • FIG. 8 Tissue factor and tissue factor pathway inhibitor (TFPI) mRNA expression in ALDSC evaluated by conventional RT-PCR.
  • TF Tissue factor (first set of primers);
  • TF' Tissue factor (second set of primers),
  • TFPI Tissue factor pathway inhibitor,
  • GAPDH Glyceraldehyde 3-Phosphate Dehydrogenase (technique control).
  • FIG. 9 Tissue factor mRNA (TF and as-TF) and TFPI expression of ALDSC evaluated by Real Time-PCR.
  • CAPAN-2 cells and HUVEC are positive control for TF, asTF and TFPI, respectively.
  • FIG. 10 (A) Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of cells suspended in human albumin 5%. No coagulation is induced if absence of recalcification. Hepatocytes (white), hALPCs (black), Control (albumin) (grey). Hepatocytes vs. hALPCs p ⁇ 0.001; Hepatocytes vs control p ⁇ 0.001; hALPCs vs control p ⁇ 0.01; hALPCs vs hepatocytes vs. control: Kruskal-Wallis test ***p ⁇ 0.001.
  • FIG. 11 Clotting time (CT) essayed by ROTEM after recalcification, without added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of cells suspended in human albumin 5%. No coagulation is induced if absence of recalcification. Hepatocytes (white), hALPCs (black)
  • FIG. 12 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence of supernatant of hALPCs culture. No coagulation is induced if absence of recalcification.
  • FIG. 13 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs, hepatocytes, skin fibroblasts, bone marrow mesenchymal stem cells (BMMSC), bone marrow haematopoietic stem cells (BMHSC), liver myofibroblasts suspended in human albumin 5%. Fibroblasts vs. control p ⁇ 0.01; BMMSC vs. control p ⁇ 0.01; Liver myofibroblasts vs. control p ⁇ 0.01.
  • CT Clotting time
  • FIG. 14 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of plasma (300 ⁇ l) deficient in coagulation factor VII, V, X and II (7d Pl, 5d Pl, 10d Pl, 2d Pl) in presence of cells suspended in human albumin 5%.
  • hALPCs black
  • Control albumin
  • Nl pl normal plasma
  • FIG. 15 (A) Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs suspended in human albumin 5% with heparin (Hepar). At contrario, enoxaparin (Eno) or Fondaparinux (Fond) was extemporaneously added to blood in contact with cells suspended in albumin.
  • hALPCs black
  • Control albumin
  • f as compared to control.
  • C Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hepatocytes suspended in human albumin 5% with heparin (Hepar).
  • Tissue Factor ExTem 20 ⁇ L
  • Hepar heparin
  • enoxaparin Eno
  • Fondaparinux Fondaparinux
  • Hepatocytes (white), Control (albumin) (grey). * as compared to hepatocytes. f as compared to control.
  • CT Clotting time
  • ExTem 20 ⁇ L Tissue Factor
  • Bivalirudin Bovalirudin
  • Hirudin Hirudin
  • FIG. 16 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs suspended in human albumin 5% with heparin (Hepar) or with Enoxaparin (Eno) or Fondaparinux (Fond) extemporaneously added to blood. Combination of anticoagulant drugs was obtained when bivalirudin (Biva) was extemporaneously added to blood.
  • hALPCs black
  • Control albumin
  • * as compared to hALPCs.
  • f as compared to control. $ as compared to bivalirudin. (B).
  • Combination of anticoagulant drugs was obtained when bivalirudin (Biva) was extemporaneously added to blood.
  • FIG. 17 Immunofluorescence for TF was performed on hALPCs (A) placed on cover slips and fixed by paraformaldehyde (magnification 20 ⁇ ). The nuclei were revealed by DAPI (blue staining). (B) Negative control (without primary antibody).
  • FIG. 18 Tissue factor and tissue factor pathway inhibitor (TFPI) mRNA expression in hALPCs and hepatocytes evaluated by conventional RT-PCR.
  • Tissue factor TF
  • alternatively spliced Tissue Factor asTF
  • Tissue factor pathway inhibitor TFPI
  • GPDH Glyceraldehyde 3-Phosphate Dehydrogenase
  • FIG. 19 Tissue factor mRNA (TF and as-TF) and TFPI expression of hALPCs and hepatocytes evaluated by Real Time-PCR.
  • CAPAN-2 cells and HUVEC are positive control for TF, asTF and TFPI, respectively.
  • FIG. 20 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of cells suspended in human albumin 5% after incubation of cells with TF antibody (TF+) or not (TF ⁇ ).
  • Hepatocytes white
  • hALPCs black
  • Control albumin
  • FIG. 21 After 30 min incubation of cells suspended in albumin supplemented or not with heparin (Hepar) (10 Ul/ml, 50 Ul/ml, and 100 Ul/ml) in blood, anti-Xa activity (Ul/ml) was measured in plasma obtained after blood centrifugation.
  • Hepar heparin
  • Ul/ml anti-Xa activity
  • FIG. 22 During cell infusion, the patients receive bivalirudin (1.75 mg/kg). Between consecutive cell infusions, the dose was decreased to 0.25 mg/kg for 2 to 4 hours.
  • Coagulation tests including thromboelastometry (CT in portal vein (port) or via the central line (centr)), platelets (PLT) (normal values: 150-350 10exp3/ ⁇ l) and D-Dimers levels (normal values: ⁇ 500 ng/ml), Thrombin time (TT, normal values: 15-24 sec), Prothrombin time (PT, normal values: 9-14 sec) and Partial thromboplastin time (PTT, normal values: 20-33 sec) were repetitively performed before, 20 min after beginning and at the end of each infusion.
  • CT thromboelastometry
  • PKT platelets
  • TT Thrombin time
  • PT Prothrombin time
  • Partial thromboplastin time PTT, normal values: 20-33 sec
  • FIG. 23 (A) Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs (Black) suspended in human albumin 5% with or without heparin (Hepar) at several concentrations (Hepar-10 Ul/ml, Hepar 5 ⁇ -50 Ul/ml, Hepar10 ⁇ -100 Ul/ml). Control (albumin) (grey). Control vs. hALPCs Hepar 5 ⁇ p ⁇ 0.01.
  • FIG. 24 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs suspended in human albumin 5%. Increased concentration of hirudin (Hir) (2 ⁇ (Hir 2 ⁇ ) or 5 ⁇ (Hir 5 ⁇ )) was extemporaneously added to blood.
  • hALPCs black
  • Control albumin
  • Control vs. hALPCs Hir 2 ⁇ , p ⁇ 0.01; hALPCs vs. hALPCs Hir 2 ⁇ , p ⁇ 0.01; hALPCs Hir vs. hALPCs Hir 2 ⁇ , n.s.
  • FIG. 25 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs suspended in human albumin 5%. Increased concentration of bivalirudin (Biva) (2 ⁇ (Biva 2 ⁇ )) was extemporaneously added to blood.
  • hALPCs black
  • Control albumin
  • Control vs. hALPCs Biva 2 ⁇ , p ⁇ 0.01
  • FIG. 26 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs suspended in human albumin 5% with or without heparin (Hepar). Enoxaparin (Eno) or fondaparinux (Fond) was extemporaneously added to blood with cells suspended or not in heparin hALPCs (black), Control (albumin) (grey). Control vs hALPCs Hepar+Eno, p ⁇ 0.01; Control vs. hALPCs Hepar+Fond, p ⁇ 0.01
  • FIG. 27 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not of hALPCs, hepatocytes, skin fibroblasts, bone marrow mesenchymal stem cells (BMMSC), bone marrow haematopoietic stem cells (BMHSC), liver myofibroblasts suspended in human albumin 5% with or without heparin (10 Ul/ml) (Hepar). Fibroblasts Hepar vs. control n.s.; Liver myofibroblasts Hepar vs. control p ⁇ 0.01
  • FIG. 28 Clotting time (CT) essayed by ROTEM after recalcification, with added Tissue Factor (ExTem 20 ⁇ L), of citrated whole blood (300 ⁇ l) in presence or not liver myofibroblasts suspended in human albumin 5% with or without heparin (10 Ul/ml) (Hepar). Combination of anticoagulant drugs was obtained when bivalirudin (Biva) was extemporaneously added to blood in contact with cells suspended in heparin.
  • CT Clotting time
  • the term “one or more”, such as one or more members of a group of members, is clear per se, by means of further exemplification, the term encompasses inter alia a reference to any one of said members, or to any two or more of said members, such as, e.g., any ⁇ 3, ⁇ 4, ⁇ 5, ⁇ 6 or ⁇ 7 or etc. of said members, and up to all said members.
  • Preferred cells in the combinations, compositions, kits, methods and uses as described herein are adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, or liver myofibroblasts, more preferably adult liver progenitor cells or liver myofibroblasts, most preferably adult liver progenitor cells.
  • cells having procoagulant activity encompasses cells which are capable of or have the propensity to activate the coagulation cascade and induce coagulation or clot formation.
  • Cells having procoagulant activity as intended herein may trigger the coagulation cascade at any stage, whereby ultimately fibrinogen is converted to fibrin, which cross-links into a clot.
  • cells having procoagulant activity may express tissue factor, the expression of which may trigger the activation of factor X to factor Xa, which in its turn, via cleavage of prothrombin to thrombin, leads to clot formation via thrombin-mediated fibrinogen conversion to fibrin.
  • tissue factor the expression of which may trigger the activation of factor X to factor Xa, which in its turn, via cleavage of prothrombin to thrombin, leads to clot formation via thrombin-mediated fibrinogen conversion to fibrin.
  • procoagulant activity can be used interchangeably with “procoagulant cells”.
  • procoagulant activity can be used interchangeably with “prothrombotic activity”.
  • procoagulant activity of cells may be determined by the presence (or absence) of specific cell characteristics, such as for instance, and without limitation, the expression of specific markers, such as tissue factor
  • procoagulant activity of cells may equally be determined by techniques such as for instance, and without limitation, thromboelastometry.
  • thromboelastometry is an established viscoelastic method for haemostasis testing in blood (or by extension any sample containing the components of the coagulation cascade, such as plasma), whereby elasticity changes in a sample are correlated with clot formation.
  • thromboelastometry measurements can be performed on a ROTEM® delta analyser (Pentapharm, Kunststoff, Germany).
  • procoagulant activity may be measured by the tubing loop method, as described in Johansson et al. (Diabetes, 2005, 54:1755-1762). Procoagulant activity may also for instance be apparent from and determined by specific cytokine profiles (reviewed for instance in van der Poll et al. Regulatory role of cytokines in disseminated intravascular coagulation. Semin Thromb Hemost. 2001, 27:639-51).
  • the cells having procoagulant activity as intended herein may be particularly suited or configured for transplantation thereof.
  • the cells may be allogeneic cells (i.e., isolated from a different subject, but of the same species, as the subject to which the cells are to be transplanted) or alternatively may be autologous cells (i.e. isolated from the same subject as the subject to which the cells are to be transplanted), or may even be xenogeneic cells (i.e., isolated from a subject of a different species than the subject to which the cells are to be transplanted).
  • the procoagulant cells may be primary cells or alternatively may be cells that have been subject to manipulation in vitro.
  • the term “manipulation in vitro” refers to any kind of manipulation of the cells outside the body. Examples of such manipulations are, without limitation, administration of drugs or other compounds which elicit an effect in the cells; depletion of specific cell constituents; genetic manipulation; gene therapy; stable or transient transfection, (pseudo)viral infection, or transformation; differentiation; dedifferentiation; subcloning etc. It may be clear that regardless of the cell origin, the cells may be subjected to storage (e.g. cryopreservation) and/or proliferation or passaging before transplantation.
  • storage e.g. cryopreservation
  • Cells may be induced to express one or more specific proteins (whether or not own to the cell, i.e., autologous) or to increase or decrease (or completely or substantially completely block) the expression thereof.
  • the cells to be transplanted may be subject to manipulation prior to isolation from the donor (e.g., drug treatment, gene therapy, etc.).
  • the cells having procoagulant activity may be a cell line.
  • procoagulant cells as intended herein may be non-haematopoietic (stem) cells, as said cells tend to not display procoagulant activity.
  • the cell product to be transplanted may include without limitation cancer cells (e.g., for study of cancer in animal models), cell-based vaccines or immunotolerance agents, etc.
  • the cells may be stably or transiently transformed with nucleic acids of interest prior to introduction to the subject.
  • Nucleic acid sequences of interest include, but are not limited to those encoding gene products that enhance the growth, differentiation and/or functioning of said cells.
  • an expression system for a protein normally expressed by liver cells can be introduced in a stable or transient fashion for the purpose of treating diseases or conditions benefiting from expression of such a protein using so-transformed (preferably liver) cells, e.g., inborn errors of liver metabolism. Methods of cell transformation are known to those skilled in the art.
  • Cells as intended herein such as in particular procoagulant cells as intended herein may be preferably of animal origin, more preferably of warm-blooded animal, even more preferably of vertebrate, yet more preferably of mammalian, and still more preferably of primate origin, and specifically including cells of human or non-human mammal or primate origin.
  • Preferred cells such as procoagulant cells are of human origin.
  • mammal as used throughout this specification includes any animal classified as such, including, but not limited to, humans, domestic and farm animals, zoo animals, sport animals, pet animals, companion animals and experimental animals, such as, for example, mice, rats, hamsters, rabbits, dogs, cats, guinea pigs, cattle, cows, sheep, horses, pigs and primates, e.g., monkeys and apes.
  • procoagulant cells may encompass without limitation progenitor cells, stem cells or partly or fully differentiated cells, such as terminally differentiated cells (i.e., fully specialised cells that may be post-mitotic).
  • cells such as procoagulant cells, and particularly progenitor or stem cells, may be of adult origin (e.g., adult progenitor or stem cells) i.e., present in or obtained from (such as removed or isolated from) an organism at the foetal stage or more preferably after birth (postpartum).
  • adult progenitor or stem cells i.e., present in or obtained from (such as removed or isolated from) an organism at the foetal stage or more preferably after birth (postpartum).
  • adult origin of cells as intended herein may refer to origin from neonatal tissue or from tissue at any subsequent developmental stage such as inter alia stages conventionally denoted in human development as infant, child, youth, adolescent or adult.
  • adult origin may refer to origin from a tissue (such as liver tissue) at any time after birth, preferably full term, and may be, e.g., at least one month of age after birth, e.g., at least 2 months, at least 3 months, e.g., at least 4 months, at least 5 months, e.g., at least 6 months age after birth, such as, for example, 1 year or more, 5 years or more, at least 10 years or more, 15 years or more, 20 years or more, or 25 years or more of age after birth.
  • progenitor or “progenitor cell” are synonymous and generally refer to an unspecialised or relatively less specialised and proliferation-competent cell which can under appropriate conditions give rise to at least one relatively more specialised cell type, such as inter alia to relatively more specialised progenitor cells or eventually to terminally differentiated cells.
  • a progenitor cell may “give rise” to another, relatively more specialised cell when, for example, the progenitor cell differentiates to become said other cell without previously undergoing cell division, or if said other cell is produced after one or more rounds of cell division and/or differentiation of the progenitor cell.
  • stem cell generally refers to a progenitor cell capable of self-renewal, i.e., which can under appropriate conditions proliferate without differentiation.
  • the term encompasses stem cells capable of substantially unlimited self-renewal, i.e., wherein at least a portion of the stem cell's progeny substantially retains the unspecialised or relatively less specialised phenotype, the differentiation potential, and the proliferation capacity of the mother stem cell; as well as stem cells which display limited self-renewal, i.e., wherein the capacity of the stem cell's progeny for further proliferation and/or differentiation is demonstrably reduced compared to the mother cell.
  • Progenitor or stem cells as intended herein may be pluripotent (i.e., capable under appropriate conditions of producing progeny of different cell types that are derivatives of all three germ layers, i.e., endoderm, mesoderm, and ectoderm, according to a standard art-accepted test, such as inter alia the ability to form a teratoma in SCID mice, or the ability to form identifiable cells of all three germ layers in tissue culture), multipotent (i.e., capable under appropriate conditions of producing progeny of at least three cell types from each of two or more different organs or tissues of an organism, wherein said cell types may originate from the same or from different germ layers, but not capable of giving rise to all of the cell types of an organism), or committed to only one or a few (e.g., one, two or three) cell lineages.
  • pluripotent i.e., capable under appropriate conditions of producing progeny of different cell types that are derivatives of all three germ layers, i.e.,
  • Prototype mammalian pluripotent stem cells may be derived from any kind of mammalian embryonic tissue, e.g., embryonic, foetal or pre-foetal tissue. Included in the definition of mPS cells are embryonic stem cells of various types, exemplified without limitation by murine embryonic stem cells, e.g., as described by Evans & Kaufman 1981 (Nature 292: 154-6) and Martin 1981 (PNAS 78: 7634-8); rat pluripotent stem cells, e.g., as described by lannaccone et al.
  • bovine embryonic stem cells e.g., as described by Roach et al. 2006 (Methods Enzymol 418: 21-37); human embryonic stem (hES) cells, e.g., as described by Thomson et al. 1998 (Science 282: 1145-1147); human embryonic germ (hEG) cells, e.g., as described by Shamblott et al. 1998 (PNAS 95: 13726); embryonic stem cells from other primates such as Rhesus stem cells, e.g., as described by Thomson et al. 1995 (PNAS 92:7844-7848) or marmoset stem cells, e.g., as described by Thomson et al. 1996 (Biol Reprod 55: 254-259).
  • ES cells As noted, prototype “human ES cells” are described by Thomson et al. 1998 (supra) and in U.S. Pat. No. 6,200,806. The scope of the term covers pluripotent stem cells that are derived from a human embryo at the blastocyst stage, or before substantial differentiation of the cells into the three germ layers.
  • ES cells in particular hES cells, are typically derived from the inner cell mass of blastocysts or from whole blastocysts. Derivation of hES cell lines from the morula stage has been documented and ES cells so obtained can also be used in the invention (Strelchenko et al. 2004. Reproductive BioMedicine Online 9: 623-629).
  • EG cells are described by Shamblott et al. 1998 (supra). Such cells may be derived, e.g., from gonadal ridges and mesenteries containing primordial germ cells from foetuses. In humans, the foetuses may be typically 5-11 weeks post-fertilisation.
  • mPS cells may include primary tissue cells and established lines that bear phenotypic characteristics of the respective cells, and derivatives of such primary cells or cell lines that still have the capacity of producing progeny of each of the three germ layers.
  • Exemplary but non-limiting established lines of human ES cells include lines which are listed in the NIH Human Embryonic Stem Cell Registry (http://stemcells.nih.gov/research/registry), and sub-lines thereof, such as, lines hESBGN-01, hESBGN-02, hESBGN-03 and hESBGN-04 from Bresagen Inc.
  • ES cell lines 22: 790-7 Further exemplary ES cell lines include lines FC018, AS034, AS034.1, AS038, SA111, SA121, SA142, SA167, SA181, SA191, SA196, SA203 and SA204, and sub-lines thereof, from Cellartis AB (Göteborg, Sweden).
  • mammalian pluripotent stem cells are such mPS cells obtainable by manipulation, such as inter alia genetic and/or growth factor and/or small molecule mediated manipulation, of non-pluripotent mammalian cells, such as somatic and particularly adult somatic mammalian cells, including the use of induced pluripotent stem (iPS) cells, as taught inter alia by Yamanaka et al. 2006 (Cell 126: 663-676), Yamanaka et al. 2007 (Cell 131: 861-872) and Lin et al. 2009 (Nature Methods 6: 805-808).
  • iPS induced pluripotent stem
  • Preferred cells as intended herein such as procoagulant cells as intended herein may include mesenchymal stem cells.
  • mesenchymal stem cell or “MSC” as used herein refers to an adult, mesoderm-derived stem cell that is capable of generating cells of mesenchymal lineages, typically cells of three or more mesenchymal lineages, e.g., osteocytic (bone), chondrocytic (cartilage), myocytic (muscle), tendonocytic (tendon), fibroblastic (connective tissue), adipocytic (fat), stromogenic (marrow stroma) lineage.
  • a cell may be considered MSC if it is capable of forming cells of each of the adipocytic, chondrocytic and osteocytic lineages, using standard, art-accepted differentiation conditions and cellular phenotype evaluation methods, e.g., as described in Pittenger et al. 1999 (Science 284: 143-7) or Barberi et al. (PLoS Med 2: e161, 2005).
  • MSC cells may be isolated from, e.g., bone marrow, blood, umbilical cord, placenta, foetal yolk sac, dermis especially foetal and adolescent skin (Young et al. 2001.
  • MSC obtained from bone marrow
  • bone marrow mesenchymal stem cells which are commonly referred to as “bone marrow mesenchymal stem cells”, “bone marrow stromal cells” or “BMSC”.
  • a sample of bone marrow for isolation of BMSC may be acquired, e.g., from iliac crest, femora, tibiae, spine, rib or other medullar spaces.
  • MSC or MSC populations as used herein may originate from bone marrow, e.g., may be isolated and optionally expanded from a bone marrow sample.
  • MSC and MSC populations originated from bone-marrow can have characteristics (e.g., marker profile, function, expansion, differentiation, etc.) different from and/or favourable over MSC originated from other tissues, such as without limitation may more efficiently and/or more controllably differentiate into certain cell lineages.
  • the terms MSC and BMSC also encompass the progeny of MSC or BMSC, e.g., progeny obtained by in vitro or ex vivo propagation of MSC or BMSC obtained from a biological sample of a subject.
  • procoagulant cells may include without limitation adult progenitor or stem cells obtained or derived from (e.g., removed or isolated from) tissues including muscle tissue (e.g., satellite cells), endocrine tissue (e.g., pancreas, gonads, adrenal gland, pineal gland, pituitary gland, thyroid and parathyroid glands), nervous tissue (e.g., neuronal or glial tissue), blood and immune system tissues, epithelial, liver, bone, cartilage, adipose or endothelial tissues.
  • muscle tissue e.g., satellite cells
  • endocrine tissue e.g., pancreas, gonads, adrenal gland, pineal gland, pituitary gland, thyroid and parathyroid glands
  • nervous tissue e.g., neuronal or glial tissue
  • blood and immune system tissues epithelial, liver, bone, cartilage, adipose or endothelial tissues.
  • Particularly preferred cells as described herein such as particularly cells having procoagulant activity are adult liver-derived progenitor or stem cells, more specifically such cells as detailed in the summary section.
  • adult liver-derived progenitor or stem cells or adult liver progenitor cells or similar may generally denote liver-originating cells having progenitor or stem cell characteristics and capable of differentiating towards one or more liver cell types, such as for example capable of at least or only hepatic differentiation (i.e., differentiating towards hepatocytes or hepatocyte-like cells).
  • Cells as intended herein such as procoagulant cells which are partly or fully differentiated or mature, such as terminally differentiated cells (i.e., fully specialised cells that may be post-mitotic), may include without limitation muscle cells (e.g., cardiomyocytes, myocytes, myotubes, myoblasts, vascular smooth muscle cells), pancreatic endocrine cells (e.g., beta cells, alpha cells, delta cells, PP-producing cells or epsilon cells), nervous cells (e.g., neurons, glial cells such as astrocytes, oligodendrocytes, Schwann cells), cells of blood and immune systems (e.g., B- or T-lymphocytes, dendritic cells, granulocytes, macrophages, etc.), epithelial cells (e.g., keratinocytes, melanocytes, kidney cells, lung cells), liver cells (e.g., hepatocytes, oval cells), bone cells (osteoblasts,
  • an antithrombin activator refers to an agent (e.g., a compound, substance or molecule) which directly activates antithrombin.
  • an antithrombin activator increases the catalytic (antagonistic) activity (i.e., increased rate constant) of antithrombin towards its target(s), such as for example thrombin, factor Xa and/or factor IXa.
  • an antithrombin activator increases the catalytic (antagonistic) activity of antithrombin towards at least factor Xa.
  • an antithrombin activator may increase the catalytic (antagonistic) activity of antithrombin specifically towards factor Xa, such as for example but without limitation fondaparinux.
  • Antithrombin activation by an antithrombin activator may be accomplished by any means, for example, and without limitation, by direct binding and induction of conformational changes in antithrombin, leading to increased accessibility and/or activity of the catalytic (target-binding) site.
  • antithrombin refers to any of the known antithrombins, preferably antithrombin III (gene symbol SERPINC1). Accordingly, as used herein, “antithrombin activator” preferably refers to an activator of antithrombin III.
  • the antithrombin activator according to the invention is heparin. In another embodiment, the antithrombin activator according to the invention is selected from the group consisting of unfractionated heparin and low molecular weight heparin. In a further embodiment, the antithrombin activator according to the invention is fondaparinux, that may be represented as 2-deoxy-6-O-sulfo-2-(sulfoamino)- ⁇ -D-glucopyranosyl-(1 ⁇ 4)-O- ⁇ -D-glucopyranuronosyl-(1 ⁇ 4)—O-2-deoxy-3,6-di-O-sulfo-2-(sulfoamino)- ⁇ -D-glucopyranosyl-(1 ⁇ 4)-O-2-O-sulfo- ⁇ -L-idopyranouronosyl-(1 ⁇ 4)-O-methyl-2-deoxy-6-O-sulfo-2-(sulfoamino)-
  • the antithrombin activator according to the invention is unfractionated heparin.
  • unfractionated heparin particularly refers to natural heparin, which is polydisperse consisting of molecular chains of varying length (usually ranging between about 5 and about 40 kDa).
  • any type of heparin may be used.
  • pharmaceutical grade heparin is derived from mucosal tissues of slaughtered meat animals such as porcine intestine or bovine lung.
  • low molecular weight heparin refers to heparin having typically an average molecular weight of less than about 8 kDa and for which at least about 60% of all chains have a molecular weight less than about 8 kDa.
  • LMWH is obtained by various methods of fractionation or depolymerisation of polymeric heparin. Examples of LMWH include, without limitation, ardeparin, certoparin, enoxaparin, parnaparin, tinzaparin, dalteparin, reviparin and nadroparin.
  • certain antithrombin activators may act through increasing the catalytic (antagonistic) activity of antithrombin (specifically) towards factor Xa. Accordingly, in some aspects and embodiments, what is contemplated are combinations, compositions, kits, methods and uses as described herein that employ, as an alternative to the antithrombin activator or in addition to the antithrombin activator, a Factor Xa inhibitor, particularly a Factor Xa inhibitor other than an antithrombin activator. Such Factor Xa inhibitor may be indirect or alternatively a direct Factor Xa inhibitor. Indirect Factor Xa inhibitors include for instance substances that inhibit the conversion of Factor X into Factor Xa.
  • a direct Factor Xa inhibitor may act directly upon Factor Xa in the coagulation cascade, without using antithrombin as a mediator, thereby preventing Factor Xa mediated conversion of prothrombin into thrombin.
  • direct Factor Xa inhibitors include Apixaban, Edoxaban, Otamixaban, Rivaroxaban, DX9065a and YM466.
  • thrombin inhibitor refers to a compound which directly binds to and inactivates thrombin. As such, a thrombin inhibitor significantly decreases or ideally completely or substantially completely blocks the catalytic activity of thrombin as conveniently measured by a decreased rate constant for its catalytic target(s), most notably fibrinogen.
  • Thrombin inhibition by a thrombin inhibitor may be reversible or irreversible, preferably reversible. Thrombin inhibition by a thrombin inhibitor may be accomplished by any means, for example, and without limitation, by direct binding to the catalytic site of thrombin.
  • the thrombin inhibitor according to the invention is selected from the group consisting of bivalirudin and hirudin.
  • bivalirudin (CAS No. 128270-60-0) is a synthetic congener of the naturally occurring drug hirudin.
  • Naturally occurring hirudin typically contains a mixture of various isoforms of this protein.
  • the term “hirudin” as used in herein particularly includes any protein having the primary amino acid sequence of a naturally occurring hirudin isoform, such as inter alia HV1, HV2, HV3, P1 or P2.
  • Recombinant hirudin can be made to produce homogeneous preparations of hirudin such as for example, and without limitation, lepirudin and desirudin.
  • Hirudin as intended herein also encompasses suitable derivatives or analogues of hirudin, e.g., by way of amino acid substitution, deletion, insertion, extension, functionalisation or chemical modification, said derivative having thrombin inhibitor activity; and further encompasses hybrids of more than one hirudin, which may be produced by genetic engineering.
  • suitable derivatives or analogues of hirudin e.g., by way of amino acid substitution, deletion, insertion, extension, functionalisation or chemical modification, said derivative having thrombin inhibitor activity; and further encompasses hybrids of more than one hirudin, which may be produced by genetic engineering.
  • WO 91/17250 describes a hirudin composed of the first 46 residues of HV1 followed by amino acids 47 to 65 of HV2.
  • the thrombin inhibitor is bivalirudin (for instance manufactured by The Medicines Company as Angiomax® or Angiox®). Whereas (natural or recombinant) hirudin and hirudin derivatives as well as bivalirudin are known to a skilled person, for further guidance consult inter alia Fenton et al. Semin Thromb Hemost., 1998, vol. 24, 87-91.
  • compositions embodying the principles of the invention may comprise, consist essentially of or consist of an anti-thrombin activator selected from the group consisting of heparin, unfractionated heparin, low molecular weight heparin and fondaparinux, preferably unfractionated heparin, a thrombin inhibitor selected from the group consisting of bivalirudin and hirudin, preferably bivalirudin, and optionally cells selected from the group consisting of adult liver progenitor cells, mesenchymal stem cells (preferably bone marrow mesenchymal stem cells), skin fibroblasts, or liver myofibroblasts, preferably adult liver progenitor cells or liver myofibroblasts, most preferably adult liver progenitor cells.
  • an anti-thrombin activator selected from the group consisting of heparin, unfractionated heparin, low molecular weight heparin and fondaparinux, preferably unfractionated heparin
  • a thrombin inhibitor selected from
  • cell transplantation carries its normal meaning and particularly refers to the administration of cells to a subject.
  • the term “cell transplantation” can be used interchangeably with “cell therapy”.
  • Cell transplantation may be performed by any technique known in the art.
  • cells may be transplanted by infusion into a subject.
  • cell infusion may be performed parenterally, e.g., intravascularly, subcutaneously, intradermally, or intramuscularly, preferably intravascularly.
  • Cells may be administered for instance, and without limitation, systemically, topically or at the site of a lesion. It may be clear that, depending on the specific application, targeted tissues, therapeutic purpose or cell type, adjustment may be made accordingly in respect of routes of administration, as well as formulations, concentrations, etc.
  • thrombotic complications or “procoagulant complications” may particularly refer to deleterious effects or complications associated with transplantation of cells having procoagulant activity, apart from clot formation per se.
  • Such effects can be for instance, and without limitation, cell loss, cell rejection or inflammation.
  • cell loss or cell rejection is meant loss or rejection of transplanted cells.
  • the result of these effects is a decrease of cell transplantation efficiency or cell engraftment potential, as less than, or in extreme cases none of, the administered total amount of cells is available to perform their intended function after transplantation.
  • Cell loss can for instance occur due to inclusion of transplanted cells in clots.
  • Cell rejection can for instance occur due to an immunological response of the host.
  • An inflammatory response can for instance be associated, or result from, the activation of the coagulation cascade.
  • inflammation can be associated with, result from, or cause cell rejection.
  • compositions comprising the herein taught combinations and further comprising one or more other components.
  • components may be included that can maintain or enhance the viability of cells.
  • such components may include salts to ensure substantially isotonic conditions, pH stabilisers such as buffer system(s) (e.g., to ensure substantially neutral pH, such as phosphate or carbonate buffer system), carrier proteins such as for example albumin, media including basal media and/or media supplements, serum or plasma, nutrients, carbohydrate sources, preservatives, stabilisers, anti-oxidants or other materials well known to those skilled in the art.
  • methods of producing said compositions by admixing the respective components of the herein taught combinations with said one or more additional components as above.
  • compositions may be for example liquid or may be semi-solid or solid (e.g., may be frozen compositions or may exist as gel or may exist on solid support or scaffold, etc.).
  • Cryopreservatives such as inter alia DMSO are well known in the art.
  • compositions as taught herein comprise one or more pharmaceutically acceptable excipient.
  • pharmaceutically acceptable as used herein is consistent with the art and means compatible with the other ingredients of a pharmaceutical composition and not deleterious to the recipient thereof.
  • carrier or “excipient” includes any and all solvents, diluents, buffers (such as, e.g., neutral buffered saline or phosphate buffered saline), solubilisers, colloids, dispersion media, vehicles, fillers, chelating agents (such as, e.g., EDTA or glutathione), amino acids (such as, e.g., glycine), proteins, disintegrants, binders, lubricants, wetting agents, emulsifiers, sweeteners, colorants, flavourings, aromatisers, thickeners, agents for achieving a depot effect, coatings, antifungal agents, preservatives, stabilisers, antioxidants, tonicity controlling agents, absorption delaying agents, and the like.
  • buffers such as, e.g., neutral buffered saline or phosphate buffered saline
  • solubilisers such as, e.g., EDTA
  • the composition may be in the form of a parenterally acceptable aqueous solution, which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • the reader is referred to Cell Therapy: Stem Cell Transplantation, Gene Therapy, and Cellular Immunotherapy, by G. Morstyn & W. Sheridan eds., Cambridge University Press, 1996; and Hematopoietic Stem Cell Therapy, E. D. Ball, J. Lister & P. Law, Churchill Livingstone, 2000.
  • Liquid pharmaceutical compositions may generally include a liquid carrier such as water or a pharmaceutically acceptable aqueous solution.
  • a liquid carrier such as water or a pharmaceutically acceptable aqueous solution.
  • physiological saline solution, tissue or cell culture media, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • the composition may include one or more cell protective molecules, cell regenerative molecules, growth factors, anti-apoptotic factors or factors that regulate gene expression in the cells. Such substances may render the cells independent of its environment.
  • compositions may contain further components ensuring the viability of the cells therein.
  • the compositions may comprise a suitable buffer system (e.g., phosphate or carbonate buffer system) to achieve desirable pH, more usually near neutral pH, and may comprise sufficient salt to ensure isoosmotic conditions for the cells to prevent osmotic stress.
  • suitable solution for these purposes may be phosphate-buffered saline (PBS), sodium chloride solution, Ringer's Injection or Lactated Ringer's Injection, as known in the art.
  • the composition may comprise a carrier protein, e.g., albumin (e.g., bovine or human albumin), which may increase the viability of the cells.
  • albumin e.g., bovine or human albumin
  • suitably pharmaceutically acceptable carriers or additives are well known to those skilled in the art and for instance may be selected from proteins such as collagen or gelatine, carbohydrates such as starch, polysaccharides, sugars (dextrose, glucose and sucrose), cellulose derivatives like sodium or calcium carboxymethylcellulose, hydroxypropyl cellulose or hydroxypropylmethyl cellulose, pregeletanized starches, pectin agar, carrageenan, clays, hydrophilic gums (acacia gum, guar gum, arabic gum and xanthan gum), alginic acid, alginates, hyaluronic acid, polyglycolic and polylactic acid, dextran, pectins, synthetic polymers such as water-soluble acrylic polymer or polyvinylpyrrolidone, proteoglycans, calcium phosphate and the like.
  • proteins such as collagen or gelatine
  • carbohydrates such as starch, polysaccharides, sugars (dextrose, glucose and sucrose), cellulose derivatives like
  • cell preparation can be administered on a support, scaffold, matrix or material to provide improved tissue regeneration.
  • the material can be a granular ceramic, or a biopolymer such as gelatine, collagen, or fibrinogen.
  • Porous matrices can be synthesized according to standard techniques (e.g., Mikos et al., Biomaterials 14: 323, 1993; Mikos et al., Polymer 35:1068, 1994; Cook et al., J. Biomed. Mater. Res. 35:513, 1997).
  • Such support, scaffold, matrix or material may be biodegradable or non-biodegradable.
  • the cells may be transferred to and/or cultured on suitable substrate, such as porous or non-porous substrate, to provide for implants.
  • cells that have proliferated, or that are being differentiated in culture dishes can be transferred onto three-dimensional solid supports in order to cause them to multiply and/or continue the differentiation process by incubating the solid support in a liquid nutrient medium of the invention, if necessary.
  • Cells can be transferred onto a three-dimensional solid support, e.g. by impregnating said support with a liquid suspension containing said cells.
  • the impregnated supports obtained in this way can be implanted in a human subject.
  • Such impregnated supports can also be re-cultured by immersing them in a liquid culture medium, prior to being finally implanted.
  • the three-dimensional solid support needs to be biocompatible so as to enable it to be implanted in a human. It may be biodegradable or non-biodegradable.
  • the cells or cell populations can be administered in a manner that permits them to survive, grow, propagate and/or differentiate towards desired cell types such as, e.g., hepatocytes.
  • the cells or cell populations may be grafted to or may migrate to and engraft within the intended organ, such as, e.g., liver. Engraftment of the cells or cell populations in other places, tissues or organs such as liver, spleen, pancreas, kidney capsule, peritoneum or omentum may be envisaged.
  • the pharmaceutical cell preparation as defined above may be administered in a form of liquid composition.
  • the cells or pharmaceutical composition comprising such can be administered systemically, topically, within an organ or at a site of organ dysfunction or lesion.
  • the pharmaceutical compositions may comprise a therapeutically effective amount of the desired cells.
  • therapeutically effective amount refers to an amount which can elicit a biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, and in particular can prevent or alleviate one or more of the local or systemic symptoms or features of a disease or condition being treated.
  • the present combinations, pharmaceutical compositions and other related aspects are particularly useful for transplantation of cells as described herein such as particularly procoagulant cells, even more particularly for the treatment of diseases or conditions which can benefit from transplantation of said cells in subjects.
  • subject or “patient” are used interchangeably and refer to animals, preferably vertebrates, more preferably mammals, and specifically includes human patients and non-human mammals. Accordingly, “subject” or “patient” as used herein means any animal, mammalian or human patient or subject to which the combinations or compositions as taught herein can be administered. Preferred patients are human subjects.
  • the terms “treat” or “treatment” refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilised (i.e., not worsening) state of disease, delay or slowing of disease progression and occurrence of complications, amelioration or palliation of the disease state. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • a phrase such as “a subject in need of treatment” includes subjects, such as mammalian or human subjects, that would benefit from treatment of a given condition, preferably a condition or disease as above. Such subjects will typically include, without limitation, those that have been diagnosed with the condition, those prone to have or develop the said condition and/or those in whom the condition is to be prevented.
  • compositions described herein may be used alone or in combination with any of the known therapies or active compounds for the respective disorders.
  • the administration may be simultaneous or sequential in any order.
  • concomitant immunosuppression therapy may be typically administered, e.g., using immunosuppressive agents, such as cyclosporine or tacrolimus (FK506).
  • immunosuppressive agents such as cyclosporine or tacrolimus (FK506).
  • liver cells such as liver progenitor or stem cells (e.g., ADHLSC cells) or hepatocytes
  • they may be employed inter alia for the treatment of liver-associated diseases including but not limited to liver dysfunction or failure, hepatitis and inborn errors of metabolism.
  • Non exhaustive examples of inborn metabolic deficiencies of liver include phenylketonuria and other aminoacidopathies, haemophilia and other clotting factor deficiencies, familial hypercholesterolemia and other lipid metabolism disorders, urea cycle disorders, glycogenosis, galactosemia, fructosemia, tyrosinemia, protein and carbohydrate metabolism deficiencies, organic aciduria, mitochondrial diseases, peroxysomal and lysosomal disorders, protein synthesis abnormalities, defects of liver cell transporters, defect of glycosylation and the like.
  • liver-associated diseases or conditions include, without limitation, acquired progressive liver degenerative diseases, fulminant liver failure and acute or chronic liver failure, human hepatotropic virus infections (HBV, HAV, HCV, HEV, HDV, . . . ).
  • liver cells include but are not limited to Alagille syndrome, alcoholic liver disease (alcohol-induced cirrhosis), a1-antitrypsin deficiency (all phenotypes), hyperlipidemias and other lipid metabolism disorders, autoimmune hepatitis, Budd-Chiari syndrome, biliary atresia, progressive familial cholestasis type I, II and III, cancer of the liver, Caroli Disease, Crigler-Najjar syndrome, fructosemia, galactosemia, carbohydrate deficient glycosylation defects, other carbohydrate metabolism disorders, Refsum disease and other peroxysomal diseases, Niemann Pick disease, Wolman disease and other lysosomal disorders, tyrosinemia, triple H, and other amino acid metabolic disorders, Dubin-Johnson syndrome, fatty liver (non alcoholi steato hepatit)
  • combinations or pharmaceutical compositions may be advantageously administered via injection (encompassing also catheter administration) or implantation, e.g. localised injection, systemic injection, intrasplenic injection (see also Gupta et al., Seminars in Liver Disease 12: 321, 1992), injection to a portal vein, injection to liver pulp, e.g., beneath the liver capsule, parenteral administration, or intrauterine injection into an embryo or foetus.
  • injection encompassing also catheter administration
  • implantation e.g. localised injection, systemic injection, intrasplenic injection (see also Gupta et al., Seminars in Liver Disease 12: 321, 1992)
  • injection to a portal vein injection to liver pulp, e.g., beneath the liver capsule, parenteral administration, or intrauterine injection into an embryo or foetus.
  • the combinations or pharmaceutical compositions comprising liver cells or liver derived cells as described herein may be used for tissue engineering and cell therapy via liver cell transplantation (LCT).
  • LCT liver cell transplantation
  • Liver cell transplantation, and liver stem cell transplantation refers to the technique of infusing mature hepatocytes or liver progenitor cells in any way leading to hepatic access and engraftment of the cells, preferably via the portal vein, but also by direct hepatic injection, or by intrasplenic injection.
  • the combinations or pharmaceutical compositions comprising the mesenchymal stem cells as described herein may be used for any solid organ repair (brain, heart, liver, kidney, pancreas, spleen, lung, gut, bladder, gallbladder), to control immune disorder, to control Cröhn disease and other auto-immune diseases, to control graft versus host disease, to control organ rejection following transplantation,
  • the combinations or pharmaceutical compositions comprising the skin fibroblasts as described herein may be used for skin repair or bone matrix formation.
  • the combinations or pharmaceutical compositions comprising the liver myofibrobasts as described herein may be used for treating or repairing damage from connective tissue disease or for creating scaffolds in combination with other cells.
  • between 10 2 to 10 10 or between 10 2 to 10 9 , or between 10 3 to 10 10 or between 10 3 to 10 9 , or between 10 4 to 10 10 or between 10 4 to 10 9 , such as between 10 4 and 10 8 , or between 10 5 and 10 7 , e.g., about 1 ⁇ 10 5 , about 5 ⁇ 10 5 , about 1 ⁇ 10 6 , about 5 ⁇ 10 6 , about 1 ⁇ 10 7 , about 5 ⁇ 10 7 , about 1 ⁇ 10 8 , about 5 ⁇ 10 8 , about 1 ⁇ 10 9 , about 2 ⁇ 10 9 , about 3 ⁇ 10 9 , about 4 ⁇ 10 9 , about 5 ⁇ 10 9 , about 6 ⁇ 10 9 , about 7 ⁇ 10 9 , about 8 ⁇ 10 9 , about 9 ⁇ 10 9 or about 1 ⁇ 10 1 ° cells can be administered to a human subject.
  • between 10 6 to 10 8 cells per kg body weight or between 1 ⁇ 10 7 to 9 ⁇ 10 7 cells per kg body weight, e.g., about 1 ⁇ 10 7 , about 2 ⁇ 10 7 , about 3 ⁇ 10 7 , about 4 ⁇ 10 7 , about 5 ⁇ 10 7 , about 6 ⁇ 10 7 , about 7 ⁇ 10 7 , about 8 ⁇ 10 7 , about 9 ⁇ 10 7 or about 1 ⁇ 10 8 cells per kg body weight can be administered to a human subject.
  • such number of cells or such number of cells per kg body weight may particularly refer to the total number of cells to be administered to a subject, which administration may be suitably distributed over one or more doses (e.g., distributed over 2, 3, 4, 5, 6, 7, 8 9 or 10 or more doses) administered over one or more days (e.g., over 1, 2, 3, 4 or 5 or more days).
  • doses e.g., distributed over 2, 3, 4, 5, 6, 7, 8 9 or 10 or more doses
  • days e.g., over 1, 2, 3, 4 or 5 or more days.
  • the precise determination of a therapeutically effective dose may be based on factors individual to each patient, including their size, age, size tissue damage, and amount of time since the damage occurred, and can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art.
  • cells in a composition to be administered, may be present at a concentration between about 10 4 /ml to about 10 8 /ml, preferably between about 10 5 /ml and about 10 7 /ml, yet more preferably between about 1 ⁇ 10 6 /ml and about 1 ⁇ 10 7 /ml, such as, e.g., about 5 ⁇ 10 6 /ml.
  • the dosage or amount of active substances as disclosed herein used depends on the individual case and is, as is customary, to be adapted to the individual circumstances to achieve an optimum effect.
  • a typical single dosage might range from about 1 ⁇ g/kg to about 250 mg/kg body weight or more, preferably from about 1 ⁇ g/kg to about 100 mg/kg body weight, more preferably from about 0.01 mg/kg to about 50 mg/kg body weight, even more preferably from about 0.01 mg/kg to about 10 mg/kg body weight, and still more preferably from about 0.05 mg/kg to about 10 mg/kg body weight or from about 0.05 mg/kg to about 1 mg/kg body weight, depending on the factors mentioned above.
  • a preferred dosage of the agent may be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered as a single daily dose, divided over one or more daily doses, or essentially continuously, e.g., using a drip infusion, or intermittently, e.g., every week or every three weeks.
  • the antithrombin activator and particularly unfractionated heparin may be administered to a subject via a cell suspension typically comprising about 5-15 Ul/ml, more typically about 8-12 Ul/ml, and even more typically about 10 Ul/ml.
  • typical dose may be about 10-30 Ul/kg/hr, more typically about 15-25 Ul/kg/hr, and even more typically about 20 Ul/kg/hr. Understandably, dose may be adapted according to coagulation tests.
  • the thrombin inhibitor may be administered to a subject at between about 0.05 and about 5 mg/kg body weight, more preferably between about 0.1 and about 3 mg/kg body weight, even more preferably between about 0.2 and about 2 mg/kg body weight; more preferably for bivalirudin between about 0.50 and about 3.00 mg/kg body weight, and yet more preferably between about 0.50 and about 2 mg/kg body weight, and even more preferably between about 0.75 and about 1.75 mg/kg body weight, or also preferably between about 1.75 and about 3.00 mg/kg body weight or more preferably between about 2.25 and about 2.75 mg/kg body weight, such as for example about 2.50 mg/kg body weight; and more preferably for hirudin between about 0.2 and about 0.6 mg/kg body weight, and yet more preferably between about 0.3 and about 0.5 mg/kg body weight, and even more preferably at about 0.4 mg/kg body weight.
  • ADSC adult-derived human liver mesenchymal stem cells
  • Human fibroblasts were collected by skin biopsy (medio-anterior side of the forearm) of 8 years old to 35 years old volunteers after written informed consent as previously described (Lysy et al. Hepatology, 2007, vol. 46, 1574-1585). Bone marrow samples were collected by aspiration of vertebrae or iliac crests of postmortem donors aged 8 to 67 years. Aspirates were collected into heparinized syringes containing 10% Hanks' balanced salt solution (Invitrogen, Merelbeke, Belgium) and were processed within 48 h according to a previously described protocol (Lysy et al. Cell Prolif., 2008, vol. 41, 36-58).
  • RNA was extracted from 0.5 ⁇ 10 6 cells using the Tripure isolation reagent kit (Roche Applied Science, Brussels, Belgium) following the manufacturer's instructions.
  • Tripure isolation reagent kit Roche Applied Science, Brussels, Belgium
  • One-step RT-PCR was performed on a Thermocycler instrument (Applied Biosystems, Lennik, Belgium) with primers synthesized at Invitrogen.
  • RT-PCR for TF or glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was realized with the primers detailed in Table 2.
  • Products were separated by electrophoresis on 1% agarose gel and visualized with ethidium bromide under ultraviolet lamp.
  • a real-time RT-PCR for TF, as-TF, TFPI, and cyclophilin A was also realized on a StepOnePlus real-time PCR system (Applied Biosystems, California, USA) using TaqMan® Gene Expression Assays, listed in Table 3.
  • TF expression two assays were used, one (TF common) amplifying a region present in both membrane and soluble (alternative splicing, asTF) form, and the other (TF membrane) amplifying a region present only in the membrane (classical) form.
  • the parameter Ct was derived for each cDNA sample and primer pair and the Cyclophilin A Ct was subtracted to obtain the ⁇ Ct.
  • the ⁇ Ct was obtained by subtracting the calibrator gene Ct, and the results expressed as fold change of the mRNA amount ( FIG. 9 ).
  • the asTF expression was calculated as difference between the ⁇ Ct of the TF common and TF membrane.
  • the Primers obtained from Applied Biosystems were as detailed in Table 3.
  • the clotting time is defined as the period of time from the start of the analysis until the start of clot formation, normally until the 2 mm amplitude is reached.
  • the clot formation time is defined as the period until the 20 mm amplitude is reached.
  • the alpha angle is defined as the angle between the centre line and a tangent to the curve through the 2 mm amplitude point, which is the end of the CT.
  • the maximum amplitude of the curve is defined as the maximum clot firmness.
  • the maximum of lysis represents the maximum fibrinolysis detected during the measurement.
  • ALDSC adult-derived human liver mesenchymal stem cells
  • TF and its natural inhibitor TFPI at the mRNA level using RT-PCR was assayed ( FIG. 8 ). Both the membrane form and the alternatively-spliced variant of TF mRNA (as-TF) were expressed in adult-derived human liver mesenchymal stem cells (ALDSC).
  • ADSC adult-derived human liver mesenchymal stem cells
  • real-time RT-PCR was used to quantify TF, as-TF, TFPI mRNA levels as shown in FIG. 9 .
  • ALDSC are suspended at a concentration of 5 ⁇ 10 6 cells/ml in a solution of Hibumin (5%), containing bicarbonate (0.84 g/l), glucose (2.5 g/l) and unfractionated heparin (10 Ul/ml).
  • the ALDSC suspension is parenterally infused in a subject.
  • the subject receives bivalirudin (1.75 mg/kg).
  • the subject receives bivalirudin (0.25 mg/kg) for 2 to 4 hours, depending on the thromboelastometry test.
  • an antithrombin activator e.g. heparin
  • a thrombin inhibitor e.g. bivalirudin
  • Concomitant administration of an antithrombin activator and a thrombin inhibitor upon cell transplantation reduces the procoagulant activity of the cells and prevents cell transplantation-associated thrombosis as well as cell transplantation-associated complications such as cell loss, cell rejection and inflammation.
  • the protocol including all experiments on human samples, and the human off label anticoagulant protocol use, and the informed consents were approved by the institution ethical review board.
  • Bone marrow samples were collected by aspiration of vertebrae or iliac crests of 3 post-mortem organ donors aged 8 to 67 years. Aspirates were collected into heparinised syringes containing 10% Hanks' balanced salt solution (Invitrogen, Merelbeke, Belgium) and were processed within 48 h according to a previously described protocol (Lysy et al., Cell Prolif. 2008; 41:36-58).
  • Human liver non-parenchymal cells were obtained after liver isolation performed in our Tissue Bank, filtration and 2 low speed centrifugations of the cell suspension from three different donors (one neonate liver and two 12-years-old donors). Next, human stellate cells were isolated by Nicodenz (Myegaard, Oslo, Norway) gradient centrifugation according to established protocols (Guimar ⁇ es et al., J. Hepatol. 2010; 52(3):389-397). Activated myofibroblasts were obtained from the isolated stellate cells.
  • the clotting time is defined as the period of time from the start of the analysis until the start of clot formation, normally until the 2 mm amplitude is reached.
  • the clot formation time is defined as the period until the 20 mm amplitude is reached.
  • the alpha angle is defined as the angle between the centre line and a tangent to the curve through the 2 mm amplitude point, which is the end of the CT.
  • the maximum amplitude of the curve is defined as the maximum clot firmness.
  • the maximum of lysis represents the maximum fibrinolysis detected during the measurement. We focused on CT.
  • cells (5 ⁇ 10exp5 cells if no specification) were incubated in 3.8 ml of citrated blood at 37° C. for 30 minutes. After incubation, whole blood was centrifuged at 4500 rpm for 10 minutes. Three hundred ⁇ l of the obtained plasma was then ready for the protocol, pipetted into the cup followed by addition or not of Innovin and CaCl2.
  • suspended cells (5 ⁇ 10exp5 cells if no specification) were added to 300 ⁇ l of plasma before addition of Innovin and CaCl2.
  • hirudin Refludan®, Cel imperative Europe Limited
  • bivalirudin Angiox®, The Medicines Company
  • Dose extrapolation was based on circulating blood volume according to weight (70 ml/kg).
  • Loops made of polyvinylchloride tubing (inner diameter 6.3 mm, length 390 mm) and treated with a Corline heparin surface were purchased from Corline (Uppsala, Sweden). Loops were supplemented with cell samples (5 ⁇ 10exp5) suspended in phosphate buffered saline before blood addition. Five mL of non-anti-coagulated blood from healthy volunteers was then added to each loop. To generate a blood flow of about 45 mL/minute, loop devices were placed on a platform rocker inside a 37° C. incubator.
  • Anti-Xa activity measurement was performed using the Biophen Heparin (LRT) kit adapted on a CA7000 (Siemens, Marburg, Germany). Briefly, the assay is a chromogenic kinetics method based on the inhibition of a constant amount of factor Xa, by the tested heparin (or other anti-Xa substance) in presence of endogenous antithrombin, and hydrolysis of a factor Xa specific chromogenic substrate by the factor Xa in excess. After 30 min incubation of cells suspended in albumin supplemented or not with heparin (10 Ul/ml, 50 Ul/ml, and 100 Ul/ml) in blood, anti-Xa activity was measured in plasma obtained after blood centrifugation.
  • LRT Biophen Heparin
  • TF cells were washed in phosphate-buffered saline supplemented with 0.5% bovine serum albumin (FACS buffer) and incubated for 20 minutes at 4° C. with the fluorescein isothiocyanate (FITC)-conjugated IgG1 mAb against TF no.4508CJ (American Diagnostica) or the corresponding isotype-matched control mAb (BD Biosciences, Erembogedem, Belgium) diluted in FACS buffer containing 10% decomplemented pooled human serum.
  • FACS buffer phosphate-buffered saline supplemented with 0.5% bovine serum albumin
  • cytosolic form of TF cells were incubated with Cytofix/Cytoperm (BD Biosciences) for 20 nm at room temperature and washed with Permwash (BD Biosciences). The samples were then incubated for 20 minutes at room temperature with FITC-conjugated anti TF mAb or the corresponding isotype-matched control mAb (BD Biosciences) diluted in permwash. Cell fluorescence was measured using a BD FACS CANTO II flow cytometer and analysed using the BD FACS Diva software.
  • TFPI tissue factor pathway inhibitor
  • RT-PCR reverse-transcription polymerase chain reaction
  • mRNA messenger ribonucleic acid
  • Tripure isolation reagent kit (Roche Applied Science, Brussels, Belgium) following the manufacturer's instructions.
  • One-step RT-PCR was performed on a Thermocycler instrument (Applied Biosystems, Lennik, Belgium) with primers synthesized at Invitrogen.
  • RT-PCR for TF or glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was realized with the primers detailed in Table 2.
  • Products were separated by electrophoresis on 1% agarose gel and visualized with ethidium bromide under ultraviolet lamp.
  • a real-time RT-PCR for TF, as-TF, TFPI, and cyclophilin A was also realized on a StepOnePlus real-time PCR system (Applied Biosystems, California, USA) using TaqMan® Gene Expression Assays, listed in Table B.
  • TF expression two assays were used, one (TF common) amplifying a region present in both membrane and soluble (alternative splicing, as-TF) form, and the other (TF membrane) amplifying a region present only in the membrane (classical) form.
  • the parameter Ct was derived for each cDNA sample and primer pair and the Cyclophilin A Ct was subtracted to obtain the ⁇ Ct.
  • CAPAN cell line was used as TF positive control while HUVEC cell line as TFPI positive control.
  • the first hALPCs infusion was performed under general anaesthesia without any pre-medication.
  • a transcutaneous catheter was placed in the main portal vein branch under fluoroscopy and ultrasound guidance after the injection of one dose of Cefazolin (40 mg/kg).
  • Cells infusion was performed using a syringe of 50 ml at a flow rate of 100 cc/h.
  • Immune suppression was administrated to the patient using tacrolimus (Prograft®, Astellas Pharma) monotherapy (0.1 mg/kg) corresponding to 2 mg per day in 2 divided doses to reach trough levels of 6-7 ng/ml.
  • Prophylactic antibiotherapy with Cefazolin (40 mg/kg) 2 times post infusion with 8 hours interval between the 2 doses was administrated.
  • the infusion and post infusion course were unremarkable and the child was discharged from the hospital on day 3 post-infusion.
  • the second hALPCs infusion was performed two weeks later. At that time, a partial thrombosis of an intra-hepatic portal vein branch occurred and led to arrest of infusion. This adverse event was treated by heparin and coumarinic anticoagulant (5 mg/day).
  • D-Dimer level was markedly elevated after both cell infusion courses. This adverse event was without consequence for the patient but justified to investigate further the procoagulant effect of the progenitor cells.
  • the second patient was a 24 years old man, with an intermediate type I/II Crigler-Najjar syndrome not responding to phenobarbital.
  • the diagnosis was established at one month after birth and confirmed by DNA analysis indicating a mutation on the UDP-Glucuronosyltransferase 1A1 gene with presence of homozygous state of L443P mutation.
  • the patient received a total of 2.2 billion hALPCs administered in 7 infusions over 2 days. Prior to the portal catheter placement, the patient received pre-medication including Cefazolin (1 gr).
  • the catheter was under ultra-sound control placed in the portal system. Solumedrol (80 mg) was injected before infusion.
  • the immune suppressive treatment consisted in tacrolimus (Prograft®, Astellas Pharma), targeting blood levels of 6-8 ng/ml.
  • a specific coagulation prophylaxis was prescribed; cells were suspended in albumin 5% and heparin at a concentration of 10 Ul/ml.
  • the subject received bivalirudin (1.75 mg/kg).
  • the subject received bivalirudin (0.25 mg/kg) for 2 to 4 hours, depending on the thromboelastometry test.
  • Coagulation tests including thromboelastometry (CT), platelets count (normal values: 150-350 10exp3/ ⁇ l) and D-Dimers levels (normal values: ⁇ 500 ng/ml), Thrombin Time (TT, normal values: 15-24 sec), Prothrombin Time (PT, normal values: 9-14 sec) and Partial Thromboplastin time (PTT, normal values: 20-33 sec) were repetitively performed before, 20 min after beginning and at the end of each infusion. Liver Doppler ultra-sound was performed after each infusion to access portal flow.
  • CT thromboelastometry
  • platelets count normal values: 150-350 10exp3/ ⁇ l
  • D-Dimers levels normal values: ⁇ 500 ng/ml
  • TT Thrombin Time
  • PT Prothrombin Time
  • PTT Partial Thromboplastin time
  • the third patient was a 17 years old teenager suffering from Glycogenosis type 1a, documented by genetic analysis (G188R mutation and 380insC insertion) and absence of glucose-6 phosphatase activity on liver biopsy. He also received antibiotic prophylaxis prior to portal catheter placement and steroids before infusion. The same immune suppressive regime was applied. The patient received a total of 3 billion progenitor cells administered in 7 infusions over 3 days aiming to control recurrent hypoglycemia. The same anticoagulation protocol and coagulation including liver Doppler ultra-sound follow-up was applied.
  • PCA procoagulant activity
  • hALPCs human adult liver progenitor cells
  • a comparable PCA of hALPCs was observed when no extrinsic TF was added ( FIG. 11 ).
  • No PCA was obtained when the hALPCs culture medium, absence of cells, was placed in the thromboelastogram instead of cells ( FIG. 12 ).
  • Hepatocytes PCA was controlled by thrombin inhibitor drugs, hirudin and bivalirudin ( FIG. 15D ).
  • Control blood in absence of cells
  • Anti-vitamin K drugs (blood obtained from treated patients with INR 2 to 3) had no influence on thromboelastometry even for control (absence of cells) (data not shown).
  • non fractionated heparin can control PCA of bone marrow mesenchymal cells, skin fibroblasts but was inactive on liver myofibroblasts PCA ( FIG. 27 ).
  • the concomitant use of non fractionated heparin and bivalirudin was also shown to modulate the PCA of liver myofibroblasts, in contrast with bivalirudin alone ( FIG. 28 ).
  • the anticoagulation protocol was successfully applied as no thrombotic or hemorrhagic event occurred in both patients.
  • bivalirudin use we observed a substantial increase in TT and limited in PT. Mild D-Dimers increase was noted in both patients, reaching 1480 ng/ml for the first patient and 1840 ng/ml for the second patient. Increase in PTT was observed in both patients, correlated with detectable anti-Xa activity ( FIGS. 22A and B). No modification of portal flow by liver Doppler ultrasound was found during infusions in both patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
US13/981,720 2011-01-25 2012-01-25 Compositions and methods for cell transplantation Abandoned US20130302291A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP11152119 2011-01-25
EP11152119.1 2011-01-25
PCT/EP2012/051157 WO2012101181A1 (en) 2011-01-25 2012-01-25 Compositions and methods for cell transplantation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/051157 A-371-Of-International WO2012101181A1 (en) 2011-01-25 2012-01-25 Compositions and methods for cell transplantation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/287,061 Division US10039789B2 (en) 2011-01-25 2016-10-06 Compositions and methods for cell transplantation

Publications (1)

Publication Number Publication Date
US20130302291A1 true US20130302291A1 (en) 2013-11-14

Family

ID=45607724

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/981,720 Abandoned US20130302291A1 (en) 2011-01-25 2012-01-25 Compositions and methods for cell transplantation
US15/287,061 Expired - Fee Related US10039789B2 (en) 2011-01-25 2016-10-06 Compositions and methods for cell transplantation
US15/687,340 Expired - Fee Related US10478459B2 (en) 2011-01-25 2017-08-25 Compositions and methods for cell transplantation

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/287,061 Expired - Fee Related US10039789B2 (en) 2011-01-25 2016-10-06 Compositions and methods for cell transplantation
US15/687,340 Expired - Fee Related US10478459B2 (en) 2011-01-25 2017-08-25 Compositions and methods for cell transplantation

Country Status (12)

Country Link
US (3) US20130302291A1 (de)
EP (1) EP2667878B1 (de)
JP (1) JP6012629B2 (de)
CN (1) CN103402527B (de)
CA (1) CA2825252C (de)
DK (1) DK2667878T3 (de)
ES (1) ES2573522T3 (de)
IL (1) IL227529A0 (de)
PL (1) PL2667878T3 (de)
PT (1) PT2667878E (de)
SG (1) SG192124A1 (de)
WO (1) WO2012101181A1 (de)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9775890B2 (en) 2012-01-25 2017-10-03 Université Catholique de Louvain Factor Xa inhibitor used with liver-derived progenitor cells
US10039789B2 (en) 2011-01-25 2018-08-07 Université Catholique de Louvain Compositions and methods for cell transplantation
US10874699B2 (en) 2013-07-05 2020-12-29 Université Catholique de Louvain Conditioned medium from human adult liver stem cells and its use in the treatment of liver disorders
US11020436B2 (en) * 2013-12-27 2021-06-01 Regenera—Medicina Veterinária Avançada Ltda. Multipotent and immunocompatible stem cell concentrate
US11253550B2 (en) 2017-03-03 2022-02-22 Rohto Pharmaceutical Co., Ltd. Method for treating fibrotic liver disease

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6525889B2 (ja) * 2013-01-12 2019-06-05 セスカ セラピューティクス, インコーポレイテッド 自己骨髄由来の幹細胞の迅速な注入
SG10201906304TA (en) 2013-03-15 2019-08-27 Miromatrix Medical Inc Use of perfusion decellularized liver for islet cell recellularization
PL3307279T3 (pl) * 2015-06-11 2020-07-13 Universiteit Maastricht Sposób zapobiegania odrzucenia przeszczepu przez biorcę
CN106995796A (zh) * 2016-01-26 2017-08-01 李建业 基于细胞因子的细胞处理方法、试剂盒及冻干粉
KR20200028391A (ko) * 2017-06-12 2020-03-16 더 유니버시티 오브 노쓰 캐롤라이나 엣 채플 힐 세포 생착용 패치 이식편 조성물
JP7264589B2 (ja) 2017-12-25 2023-04-25 株式会社 資生堂 トロンビンの抑制作用を指標とした皮膚状態改善剤のスクリーニング方法、及びトロンビン作用阻害剤を含む皮膚状態改善剤
CN112608890A (zh) * 2020-12-17 2021-04-06 陕西佰傲干细胞再生医学有限公司 一种防止间充质干细胞粘连的培养方法

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9010552D0 (en) 1990-05-10 1990-07-04 Erba Carlo Spa Method for the recombinant production of hirudins and novel hirudins
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5486359A (en) 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5837539A (en) 1990-11-16 1998-11-17 Osiris Therapeutics, Inc. Monoclonal antibodies for human mesenchymal stem cells
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5827740A (en) 1996-07-30 1998-10-27 Osiris Therapeutics, Inc. Adipogenic differentiation of human mesenchymal stem cells
GB0014136D0 (en) 2000-06-10 2000-08-02 Astrazeneca Ab Combination therapy
DE10202982A1 (de) 2002-01-26 2003-07-31 Augustinus Bader Verfahren und Vorrichtung zum Einbringen von lebenden Zellen in eine biologische Körperflüssigkeit
US20060222640A1 (en) * 2005-03-29 2006-10-05 Boehringer Ingelheim International Gmbh New pharmaceutical compositions for treatment of thrombosis
WO2006126219A1 (en) 2005-05-26 2006-11-30 Fresenius Medical Care Deutschland G.M.B.H. Liver progenitor cells
AU2006292203A1 (en) 2005-09-21 2007-03-29 Dask Technologies, Llc Methods and compositions for organ and tissue functionality
AU2006328988B2 (en) 2005-12-21 2012-08-02 Universite Catholique De Louvain Isolated liver stem cells
CA2825252C (en) 2011-01-25 2018-10-16 Universite Catholique De Louvain Compositions of an antithrombin activator and a thrombin inhibitor for use in cell transplantation
JP6034406B2 (ja) 2012-01-25 2016-11-30 ユニヴェルシテ カソリック ド ルーヴァンUniversite Catholique De Louvain 細胞移植のための組成物および方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Lisman et al Rebalanced hemostasis in patients with liver disease: evidence and clinical consequences. Blood 12 AUGUST 2010. Vol 116(6). pp 878-885 *
Lisman et al. J Hepatology 2010 vol 52/ pp355-361. *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10039789B2 (en) 2011-01-25 2018-08-07 Université Catholique de Louvain Compositions and methods for cell transplantation
US10478459B2 (en) 2011-01-25 2019-11-19 Universite Catholique De Louvain Compositions and methods for cell transplantation
US9775890B2 (en) 2012-01-25 2017-10-03 Université Catholique de Louvain Factor Xa inhibitor used with liver-derived progenitor cells
US10874699B2 (en) 2013-07-05 2020-12-29 Université Catholique de Louvain Conditioned medium from human adult liver stem cells and its use in the treatment of liver disorders
US11793837B2 (en) 2013-07-05 2023-10-24 Université Catholique de Louvain Conditioned medium from human adult liver stem cells and its use in the treatment of liver disorders
US11020436B2 (en) * 2013-12-27 2021-06-01 Regenera—Medicina Veterinária Avançada Ltda. Multipotent and immunocompatible stem cell concentrate
US11253550B2 (en) 2017-03-03 2022-02-22 Rohto Pharmaceutical Co., Ltd. Method for treating fibrotic liver disease

Also Published As

Publication number Publication date
CA2825252C (en) 2018-10-16
SG192124A1 (en) 2013-08-30
CN103402527B (zh) 2016-04-20
IL227529A0 (en) 2013-09-30
CA2825252A1 (en) 2012-08-02
EP2667878A1 (de) 2013-12-04
JP2014508138A (ja) 2014-04-03
PT2667878E (pt) 2016-06-03
ES2573522T3 (es) 2016-06-08
US10039789B2 (en) 2018-08-07
US20170354723A1 (en) 2017-12-14
EP2667878B1 (de) 2016-03-30
WO2012101181A1 (en) 2012-08-02
US10478459B2 (en) 2019-11-19
US20170042940A1 (en) 2017-02-16
PL2667878T3 (pl) 2016-10-31
CN103402527A (zh) 2013-11-20
DK2667878T3 (en) 2016-06-27
JP6012629B2 (ja) 2016-10-25

Similar Documents

Publication Publication Date Title
US10039789B2 (en) Compositions and methods for cell transplantation
US9775890B2 (en) Factor Xa inhibitor used with liver-derived progenitor cells
EP3091991B1 (de) Zusammensetzungen mit immunomodulatorischer wirkung
AU2017328913B2 (en) Macrophage-based therapy for use in the treatment of liver injury
JP2020050659A (ja) 溶剤/界面活性剤処理された血漿(s/d血漿)を含む製剤及びその使用
Liuwantara et al. Characterizing the mechanistic pathways of the instant blood-mediated inflammatory reaction in xenogeneic neonatal islet cell transplantation
US20220233592A1 (en) Treatment of kidney failure using ex vivo reprogrammed immune cells
CN113677790B (zh) 用于治疗慢加急性肝衰竭的成体肝祖细胞
US20220202874A1 (en) Adult Liver Progenitor Cells for Treating Non-Alcoholic Fatty Liver Disease
US20220049225A1 (en) Cell composition comprising liver progenitor cells expressing hla-e
den Tierkliniken Comparative Characterization of Human and Equine Mesenchymal Stromal Cells: A Basis for Translational Studies in the Equine Model

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITE CATHOLIQUE DE LOUVAIN, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STEPHENNE, XAVIER;SOKAL, ETIENNE;NAJIMI, MUSTAPHA;AND OTHERS;SIGNING DATES FROM 20130812 TO 20130904;REEL/FRAME:031185/0172

AS Assignment

Owner name: PROMETHERA BIOSCIENCES S.A., BELGIUM

Free format text: LICENSE;ASSIGNORS:UNIVERSITE CATHOLIQUE DE LOUVAIN;SOPARTEC SA;REEL/FRAME:038683/0239

Effective date: 20160315

STCB Information on status: application discontinuation

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