Multi-chimeric cells and transplantation therapies and treatment of immunodeficiency and genetic disorders

文档序号:1315886 发布日期:2020-07-10 浏览:17次 中文

阅读说明:本技术 多嵌合细胞以及移植疗法与免疫缺陷和遗传病症的治疗 (Multi-chimeric cells and transplantation therapies and treatment of immunodeficiency and genetic disorders ) 是由 马里亚·西米奥诺 于 2018-11-28 设计创作,主要内容包括:提供了通过来自三个或更多个不同供体的三个或更多个造血干细胞、间充质干细胞、成肌细胞、周细胞或卫星细胞或其组合的离体融合而产生的多嵌合细胞,以及这些细胞在移植疗法和治疗免疫缺陷和遗传病症中的用途。(Provided are multi-chimeric cells produced by ex vivo fusion of three or more hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes or satellite cells, or a combination thereof, from three or more different donors, and the use of these cells in transplantation therapies and in the treatment of immunodeficiency and genetic disorders.)

1. A multi-chimeric cell comprising an ex vivo fusion of three or more hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, satellite cells, or a combination thereof from three or more different donors.

2. The multi-chimeric cell of claim 1, consisting of an ex vivo fusion of a hematopoietic stem cell with two or more cells independently selected from the group consisting of a hematopoietic stem cell and a mesenchymal stem cell.

3. The multi-chimeric cell of claim 1, wherein the hematopoietic stem cell is isolated from bone marrow, cord blood, peripheral blood, or a combination thereof.

4. The multi-chimeric cell of claim 1, wherein the donor is a related donor, an unrelated donor, or a combination thereof.

5. The multi-chimeric cell of claim 1, consisting of an ex vivo fusion of a hematopoietic stem cell with two or more cells independently selected from the group consisting of a hematopoietic stem cell, a mesenchymal stem cell, and a pericyte.

6. The multi-chimeric cell of claim 5, consisting of an ex vivo fusion of hematopoietic stem cells, mesenchymal stem cells, and pericytes.

7. The multi-chimeric cell of claim 5, which consists of an ex vivo fusion of three hematopoietic stem cells.

8. The multi-chimeric cell of claim 1, consisting of an ex vivo fusion of a myoblast and two cells independently selected from a mesenchymal stem cell, a myoblast, and a satellite cell.

9. The multi-chimeric cell of claim 8, consisting of an ex vivo fusion of a myoblast, a mesenchymal stem cell, and a satellite cell.

10. The multi-chimeric cell of claim 8, which consists of an ex vivo fusion of three myoblasts.

11. A method of treating an immunodeficiency or a genetic disorder, the method comprising administering to a subject in need of treatment an effective amount of the multi-chimeric cell of claim 1, thereby treating the immunodeficiency or genetic disorder in the subject.

12. The method of claim 11, wherein the multi-chimeric cells are administered by intraosseous injection, intravenous injection, or intramuscular injection.

13. The method of claim 11, wherein the immunodeficiency or genetic disorder is selected from the group consisting of bone marrow failure, Adenosine Deaminase (ADA) deficiency, Severe Combined Immunodeficiency (SCID), digo syndrome, ataxia telangiectasia, muscular dystrophy, type I diabetes, gaucher's disease, leukemia, aplastic anemia, sickle cell anemia, lymphoma, and multiple myeloma.

14. The method of claim 11, wherein the genetic disorder is muscular dystrophy.

15. The method of claim 14, wherein the multi-chimeric cell comprises an ex vivo fusion of a myoblast and two or more cells independently selected from a mesenchymal stem cell, a myoblast, and a satellite cell.

16. The method of claim 11, wherein the immunodeficiency or genetic disorder is sickle cell anemia.

17. The method of claim 16, wherein the multi-chimeric cells comprise ex vivo fusions of hematopoietic stem cells with two or more cells independently selected from hematopoietic stem cells and mesenchymal stem cells.

18. A method of preventing the development of, or reducing the severity of, GvHD in a subject receiving an allogeneic hematopoietic stem cell transplant or organ transplant, the method comprising administering to the subject an effective amount of the multi-chimeric cell of claim 1, thereby preventing the development of, or reducing the severity of, GvHD in the subject.

19. The method of claim 18, wherein the multi-chimeric cells comprise ex vivo fusions of hematopoietic stem cells with two or more cells independently selected from hematopoietic stem cells and mesenchymal stem cells.

20. A kit, comprising:

(a) fluxing agent, and

(b) three or more donor cells selected from the group consisting of hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, satellite cells from three or more different donors.

Background

Stem cell transplantation is a promising new strategy for the treatment of many human disorders. Hematopoietic Stem Cell (HSC) transplants are the most instructive example based on the therapeutic potential of stem cell therapies. Although autologous HSCs may not be the treatment of choice for many hematologic diseases, the availability of allogeneic stem cell transplants is limited due to the lack of matching relevant donors. Furthermore, matched unrelated HSC transplants are often hindered by excessive mortality and morbidity due to the conditioning regimen and toxic effects of graft versus host disease (GvHD). Without the risk of GvHD, improvements in the engraftment rate or the efficacy and ability to perform transplants across histocompatibility disorders would greatly increase the use of HSC transplants as a new treatment option for many diseases that are often incurable.

Chimeric cells have been developed by a variety of ex vivo fusion methods (Siemiow, et al (2012) Ann. plant. Surg.69(5): 575-9; Cwykiel & Siemiow (2015) Plastic and ReconstructivetSurgery: Experimental Models and Research Design, Siemiow (eds.), Schringge Press London Limited (Springer-Verlag L on, L td.), chapter 71-72. chimeric cell therapies applied in different Experimental Models have been described, including Vascularized Composite Allografts (VCA), radiation therapy and muscular dystrophy animal Models (Arslan, et al (2007) Microury 27: 190-9; Kulahci, et al (Transplantation 8490 (8): 843-52; Hivelin, et al (2016) Microury 27: 190-9; Microlavi, Immunoz.2016; Waltz @ 310; Waltz) Warnez et al (2016. 2016; 310) 75): 310).

Disclosure of Invention

The present invention is a multi-chimeric cell consisting of a fusion of three or more hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, satellite cells, or a combination thereof from three or more different donors. In certain embodiments, the hematopoietic stem cells are isolated from bone marrow, cord blood, peripheral blood, or a combination thereof. In other embodiments, the donor is a related donor, an unrelated donor, or a combination thereof. In some embodiments, the fusion is a combination of a hematopoietic stem cell and two or more cells independently selected from the group consisting of a hematopoietic stem cell and a mesenchymal stem cell. In other embodiments, the fusion is a combination of hematopoietic stem cells and two or more cells independently selected from hematopoietic stem cells, mesenchymal stem cells, and pericytes. In other embodiments, the fusion is a combination of a myoblast and two cells independently selected from the group consisting of a mesenchymal stem cell, a myoblast, and a satellite cell. In a particular embodiment, the fusion is a combination of: hematopoietic stem cells, mesenchymal stem cells and pericytes; three hematopoietic stem cells; myoblasts, mesenchymal stem and satellite cells; or three myoblasts. Also provided are kits comprising a fusogenic agent, and three or more donor cells selected from hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, satellite cells from three or more different donors.

The invention also provides a method of treating an immunodeficiency or a genetic disorder by administering to a subject in need of treatment an effective amount of a multi-chimeric cell of the invention. In some embodiments, the chimeric cells are administered by intraosseous injection, intravenous injection, or intramuscular injection. In other embodiments, the immunodeficiency or genetic disorder is selected from the group consisting of bone marrow failure, Adenosine Deaminase (ADA) deficiency, Severe Combined Immunodeficiency (SCID), DiGeorge Syndrome (DiGeorge Syndrome), ataxia telangiectasia, muscular dystrophy, type I diabetes, gaucher's disease, leukemia, aplastic anemia, sickle cell anemia, lymphoma, and multiple myeloma. In embodiments involving treatment of muscular dystrophy, preferably the multi-chimeric cells consist of a fusion of a myoblast and two or more cells independently selected from mesenchymal stem cells, myoblasts and satellite cells. In embodiments involving treatment of sickle cell anemia, preferably, the multi-chimeric cells are fusions of hematopoietic stem cells with two or more cells independently selected from hematopoietic stem cells and mesenchymal stem cells.

The invention also provides methods of preventing the development of GvHD or reducing the severity of GvHD in a subject by administering to a subject receiving an allogeneic hematopoietic stem cell transplant or organ transplant an effective amount of a multi-chimeric cell of the invention. According to this aspect of the invention, preferably the multi-chimeric cells are fusions of hematopoietic stem cells with two or more cells independently selected from hematopoietic stem cells and mesenchymal stem cells.

Drawings

Figure 1 shows an ex vivo fusion process for generating human multi-chimeric cells (MCCs). Human Hematopoietic Stem Cells (HSCs) are obtained from at least three or more related and/or unrelated donors. Prior to fusion, cells were fluorescently labeled with PKH26 (red) or PKH67 (green) or eFluor670 (magenta) dyes, respectively. Cell fusion of fluorescently labeled cells was performed using polyethylene glycol (PEG). Fused triple (PKH26, PKH67, and eFluor670) stained cells were selected by Fluorescence Activated Cell Sorting (FACS). Multiple chimeric (triple) cells are delivered to a graft or immunodeficient patient by systemic intraosseous injection.

Detailed Description

The present invention relates to a method for treating a patient suffering from an immune deficiency or a genetic disorder, comprising the step of contacting the patient with a chimeric cell of the invention, wherein the chimeric cell is derived from human hematopoietic stem cells (autologous and/or allogeneic) from at least three (triple chimeras) or more (quadruple chimeras, multiple generation chimeras, etc.) related and/or unrelated donors (fig. 1).

Accordingly, the present invention is a multi-chimeric cell that is the product of an ex vivo fusion of three or more hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes or satellite cells, or a combination thereof, from three or more different donors, wherein the fusion is performed in the presence of an exogenous fusogenic agent. For the purposes of the present invention, a "multiple chimeric cell" is a cell constructed from a cell fusion or cross (whole cell cross) of three or more biological cells (parental cells). Although the multiple chimeric cells of the invention are referred to as "multiple chimeric cells," the chimeric cells are intended to mean a single cell or a population of cells.

"hematopoietic stem cells" or "HSCs" refer to clonogenic, self-renewing, pluripotent cells capable of eventual differentiation into all cell types of the hematopoietic system, includingB cells, T cells, NK cells, lymphoid dendritic cells, myeloid dendritic cells, granulocytes, macrophages, megakaryocytes, and erythroid cells. Marker phenotypes for identifying HSCs are well known in the art. For human HSCs, the cell marker phenotype preferably includes CD34+CD38-CD90(Thy1)+Lin-. For mouse HSCs, an exemplary cellular marker phenotype is Sca-1+CD90+(see, e.g., Spangrude et al (1988) Science 1:661-673) or c-kit+ThyloLin-Sca-1+(see Uchida, et al (1998) J. Clin. invest.101(5): 961) -966.) alternative HSC markers such as aldehyde dehydrogenase (see Storms, et al (1999) Proc. nat' l Acad. Sci.96:9118-23), AC133 (see Yin, et al (1997) Blood 90:5002-12) and CD150(S L AM) (see Kiel (2005) Cell121(7):1109-21) may also find advantageous use.

"mesenchymal stem cells" or "MSCs" are cells that can give rise to connective tissue, bone, cartilage, and cells in the circulatory and lymphatic systems. Mesenchymal stem cells are present in the mesenchyme, which is the part of the embryonic mesoderm consisting of loosely packed fusiform or stellate non-specific cells. Mesenchymal stem cells may be obtained by conventional methods and may be identified by one or more of the following markers: CD29, CD31、CD34、CD44、CD45CD51, CD73, CD90/Thy-1, CD105, CD166, integrin α 1, PDGF R α, nestin, Sca-1+SCFR/c-kit, STRO-1, and VCAM-1. In some embodiments, the mesenchymal stem cells are derived or obtained from Bone Marrow (BM) or adipose tissue (ASC). In particular embodiments, the mesenchymal stem cells are derived or obtained from human bone marrow.

As is conventional in the art, "myoblasts" refer to primitive myocytes that have the potential to develop into myofibers. Myoblasts are characterized by the expression of the myocilin and CD56 and can be obtained from fetal or adult tissue using methods known in the art. See, for example, WO 93/03768, which discloses the isolation of myoblasts from a crude cell population by flow cytometry (e.g., FAC). Alternatively, the myoblasts may be obtained by growing and propagating myoblasts derived from muscle biopsies in culture. See, e.g., Springer, et al (1997) In: Current Human genetics. Unit 13.4, Boyle, eds.

As used herein, "pericytes" are pluripotent cells associated with the walls of small blood vessels previous studies have shown that pericytes can differentiate into a variety of cell types, such as adipocytes, chondrocytes, fibroblasts, and macrophages the pericytes are characterized by the expression of one of the following markers vimentin, glial 2(NG2), platelet-derived growth factor receptor β (PDGFR- β), and α smooth muscle actin (α -SMA).

"satellite cells" are mononuclear myoblasts of mature muscle fibers responsible for the growth and in vivo regeneration of the muscles after birth. Markers for the characterization of satellite cells include, but are not limited to, M-cadherin, CD34 and c-met. Satellite cells can be isolated from muscle tissue by known methods (see, e.g., US 2007/0224168).

As used herein, a donor is a subject that provides cells for use in the preparation of a multi-chimeric cell of the invention. The donor is preferably a healthy donor, i.e. an individual not suffering from a genetic disorder or disease. Further, the donor may be any mammal, including humans, mice, rats, dogs, cats, horses, etc. In a particular embodiment, the donor is a human.

The donor may be a genetic relative (e.g., parent or sibling) of the subject or cell bank donor. Thus, in some embodiments, one or more cells used to generate the multi-chimeric cells are autologous. In other embodiments, all of the cells used to generate the multi-chimeric cells are autologous. In additional embodiments, one or more cells used to generate the multi-chimeric cells are allogeneic. In still other embodiments, all of the cells used to generate the multi-chimeric cells are allogeneic.

As used herein, "autologous," when it relates to the isolation and transplantation of cells, refers to cells in which the donor and recipient are the same individual. Thus, autologous cells are harvested from a subject and then returned to the same subject. In contrast, an "allogeneic" cell is one in which the donor and recipient are genetically distinct individuals from the same species. In contrast, "xenogeneic" cells are cells in which the donor and recipient belong to different species.

As noted, the multi-chimeric stem cells of the invention are fusions of three or more hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, or satellite cells, or combinations thereof. In some embodiments, the multi-chimeric stem cell is a fusion comprising or consisting of three cells selected from the group consisting of hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes and satellite cells. In other embodiments, the multi-chimeric stem cell is a fusion comprising or consisting of four, five, six, seven, eight, nine, or ten cells selected from the group consisting of hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, and satellite cells. In embodiments involving fusion of three cells, specific combinations of cells are provided in table 1.

TABLE 1

Cell1 Cell 2 Cell 3
HSC HSC HSC
HSC HSC MSC
HSC MSC MSC
HSC HSC Pericyte
HSC MSC Pericyte
HSC Pericyte Pericyte
Myoblast cell Myoblast cell Myoblast cell
Myoblast cell Myoblast cell MSC
Myoblast cell MSC MSC
Myoblast cell MSC Satellite cell
Myoblast cell Myoblast cell Satellite cell
Myoblast cell Satellite cell Satellite cell

In embodiments involving four cell fusions, combinations of four cells are provided in table 2.

TABLE 2

Cell1 Cell 2 Cell 3 Cell 4
HSC HSC HSC HSC
HSC HSC MSC MSC
HSC MSC MSC Pericyte
HSC HSC Pericyte Pericyte
HSC MSC Pericyte Pericyte
HSC Pericyte Pericyte Pericyte
Myoblast cell Myoblast cell Myoblast cell Myoblast cell
Myoblast cell Myoblast cell MSC MSC
Myoblast cell MSC MSC MSC
Myoblast cell MSC Satellite cell Satellite cell
Myoblast cell Myoblast cell Satellite cell Satellite cell
Myoblast cell Satellite cell Satellite cell Satellite cell

In some embodiments, the multi-chimeric stem cell is an ex vivo fusion of a hematopoietic stem cell and two cells independently selected from the group consisting of a hematopoietic stem cell, a mesenchymal stem cell, and a pericyte. In a particular embodiment, the multi-chimeric stem cell is an ex vivo fusion of a hematopoietic stem cell and two cells independently selected from the group consisting of a hematopoietic stem cell and a mesenchymal stem cell. In another embodiment, the multi-chimeric stem cell is an ex vivo fusion of a hematopoietic stem cell, a mesenchymal stem cell, and a pericyte. In another specific embodiment, the multi-chimeric stem cell is an ex vivo fusion of three hematopoietic stem cells.

In other embodiments, the multi-chimeric stem cell is an ex vivo fusion of a myoblast and two cells independently selected from the group consisting of a mesenchymal stem cell, a myoblast, and a satellite cell, or a combination thereof. In particular embodiments, the multi-chimeric stem cell is an ex vivo fusion of a myoblast, a mesenchymal stem cell, and a satellite cell. In another specific embodiment, the multi-chimeric stem cell is an ex vivo fusion of three myoblasts.

Cells useful for making the multi-chimeric stem cells of the invention can be isolated and optionally purified. As used herein, the term "isolated" is intended to describe a cell of interest that is in an environment that is different from the environment in which the element naturally occurs. As used herein, "purified" refers to cells that are removed from the environment in which the cells are produced and are at least 60% free, preferably 75% free, and most preferably 90% free from other components that are naturally associated with the cells or with the cells during production.

Purification and/or identification of the cells of interest can be accomplished by any means known in the art, such as immunologically, histochemical staining, flow cytometry, Fluorescence Activated Cell Sorting (FACS), western blot analysis, enzyme-linked immunosorbent assay (E L ISA), density gradient separation (e.g.,ficoll 400), immunomagnetic bead separation, and combinations thereof. Flow immunocytochemistry may be used to detect cell surface markers, and immunohistochemistry (e.g., fixed cell immunohistochemistry) may be used to detect intracellular or cell surface markers. Western blot analysis can be performed on cell extracts. Enzyme-linked immunosorbent assays can be used for cell extracts or products secreted into the culture medium. Antibodies for identifying stem cell markers can be obtained from commercial sources, such as from Chemicon International (temkura, canada).

The multi-chimeric stem cells of the present invention are prepared by: contacting three or more cells from three or more different donors ex vivo with an exogenous fusion agent, thereby promoting ex vivo fusion of the three or more different donor cells. By "ex vivo" is meant that the cells are manipulated in vitro. Cell fusion is a process in which three or more cells are fused together by fusing their plasma membranes. The multi-chimeric stem cells can be prepared using a fusogenic agent and methods known in the art, including but not limited to exposing the cells to a chemical that promotes fusion, such as polyethylene glycol (PEG); use of inactivated viruses, such as Sendai virus; and using electrical stimulation. See, for example, Kennett (1979) Methods enzymol.58: 345-359 reviewed the commonly used methods based on Sendai virus-induced cell fusion or polyethylene glycol (PEG) -induced cell fusion. Briefly, the cells to be fused are incubated with a fusing agent such as Sendai virus or PEG. Centrifugation or agitation can be used to promote agglutination and close apposition of cell membranes. Variables such as time, temperature, cell concentration, and fusogen concentration can be optimized for each cell combination. In contrast to electrofusion, a short electrical pulse is passed through the cell mixture to stimulate fusion. See, for example, Neil & Zimmermann (1993) Methods Enzymol.220: 174. sup. 196. In certain embodiments, the multi-chimeric stem cells are prepared by polyethylene glycol cell fusion.

Prior to fusion, the donor cells may or may not be cultured to increase their numbers. In addition, the donor cells may or may not be labeled (e.g., with a membrane dye) to monitor fusion of the donor cells. Illustratively, HSCs from the first donor were labeled with PKH26 (emission at 567 nm), MSCs from the second donor were labeled with eFluor670 (emission at 670 nm), and pericytes from the third donor were labeled with PKH67 (emission at 502 nm), such that after fusion, the fused cells fluoresce at 502nm, 567nm, and 670nm (fig. 1). To identify the multiple chimeric cells of the invention, the fused cells are selected by cell sorting, such as Fluorescence Activated Cell Sorting (FACS). Once identified, the multiple chimeric cells can be cryopreserved, stored, and stockpiled as cell lines. Alternatively, successful fusion of three or more cells can be assessed by morphological, phenotypic, or genotypic characterization.

The multi-chimeric stem cells of the invention are particularly useful for treating immunodeficiency and genetic disorders. Accordingly, the present invention also provides methods for treating an immunodeficiency or a genetic disorder in a subject by administering to a subject in need of treatment an amount of a multi-chimeric stem cell of the invention or a composition containing the same effective for treating the immunodeficiency or the genetic disorder. "treating" a subject having a disease or disorder refers to achieving one or more of the following: (a) lessening the severity of the disease; (b) arresting the development of the disease or disorder; (e) inhibiting the exacerbation of a disease or disorder; (d) limiting or preventing relapse of the disease or disorder in a patient previously suffering from the disease or disorder; (e) causing regression of the disease or condition; (f) ameliorating or eliminating symptoms of the disease or disorder; and (g) increasing survival rate. In some embodiments, the multi-chimeric stem cells used in the methods of the invention are fusions of three or more hematopoietic stem cells, mesenchymal stem cells, or myoblasts from three or more different donors.

Immunodeficiency and genetic disorders that may be treated according to the invention include bone marrow failure, Adenosine Deaminase (ADA) deficiency, Severe Combined Immunodeficiency (SCID), dychis syndrome, ataxia telangiectasia, muscular dystrophy, type I diabetes, gaucher's disease, leukemia, aplastic anemia, lymphoma and multiple myeloma. In certain embodiments, the immunodeficiency and genetic disease is selected from the group consisting of bone marrow failure, muscular dystrophy, sickle cell anemia, leukemia, and type I diabetes. In other embodiments, the genetic disease is muscular dystrophy.

Muscular dystrophy is a group of genetic diseases characterized by progressive weakness and degeneration of skeletal muscles that control movement. Examples of muscular dystrophy include duchenne muscular dystrophy, becker muscular dystrophy, limb girdle muscular dystrophy, myotonic muscular dystrophy, facioscapulohumeral muscular dystrophy, oculopharyngeal muscular dystrophy, Emery-Dreifuss muscular dystrophy, fushan congenital muscular dystrophy, sanhao distal myopathy, Ullrich congenital muscular dystrophy, Steinert muscular dystrophy. In certain embodiments, the muscular dystrophy is Duchenne Muscular Dystrophy (DMD). In a particular embodiment, the muscular dystrophy is treated with a multi-chimeric cell that is a fusion of a myoblast and two or more cells independently selected from the group consisting of a mesenchymal stem cell, a myoblast, and a satellite cell.

In another embodiment, the immunodeficiency or genetic disorder is sickle cell anemia. According to this embodiment, the sickle cell anemia is treated with a multi-chimeric cell that is a fusion of a hematopoietic stem cell and two or more cells independently selected from the group consisting of a hematopoietic stem cell and a mesenchymal stem cell.

To the extent that the multiple chimeric cells of the invention provide tolerogenic and immunomodulatory effects in the context of organ and HSC transplantation, the invention also includes methods of preventing the development of, or reducing the severity of, GvHD in a subject receiving an allogeneic hematopoietic stem cell transplant or organ transplant. As known in the art, a "graft versus host response" or "GVH" or "GVHD" refers to a cellular response that occurs when lymphocytes of different MHC classes are introduced into a host, resulting in a reaction of the lymphocytes against the host. According to the method, an effective amount of a multi-chimeric cell is administered to a subject receiving an allogeneic hematopoietic stem cell transplant or organ transplant to prevent the development of, or reduce the severity of, GvHD.

The multi-chimeric cells can be administered to a subject alone and/or included as part of a hematopoietic stem cell graft or organ graft. Treatment with the multiple chimeric cells of the invention can significantly reduce the incidence and severity of GvHD and prevent allograft rejection, lessen the immune response and/or reduce/eliminate the need for life-long immunosuppression in transplanted patients. In this regard, the multi-chimeric cells of the invention are useful for preventing the development of GvHD related symptoms and signs in various organs and systems, including skin, nails, mouth, eye, female genitalia, gastrointestinal tract, liver, lung, muscle, fascia, joints, etc. In a particular embodiment, GvHD is treated with a multi-chimeric cell that is a fusion of a hematopoietic stem cell and two or more cells independently selected from the group consisting of a hematopoietic stem cell and a mesenchymal stem cell.

According to the methods of treatment herein, the multi-chimeric cells or compositions comprising the same are administered to a subject in need of treatment. In some embodiments, a combination of multiple chimeric cells of the invention may be administered. The multiple chimeric cells or combination of cells can be administered by implantation, wherein the cells are injected into the subject, e.g., intravenously, intramuscularly, intra-arterially, intraosseously, etc. In particular embodiments, the multi-chimeric stem cells are administered by intraosseous injection, intravenous injection, or intramuscular injection. In certain embodiments, administration involves implantation of about 102、104、106、107、108、109、1010、1012Or more cells. The number of implanted cells can be selected based on the route of administration and/or the severity of the condition for which the implanted cells are used to treat. Advantageously, the multi-chimeric stem cells of the invention will successfully engraft and complement genetics/immunityDefective in immunogenicity and provides tolerogenicity and immunomodulation in the case of organ and HSC transplants.

Compositions comprising a multi-chimeric cell or a combination of multi-chimeric cells can be prepared by mixing the cell or combination of cells with a pharmaceutically acceptable carrier or an aqueous medium. The phrase "pharmaceutically or pharmacologically acceptable" refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or human. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Unless any conventional media or agent is incompatible with the cells of the present disclosure, its use in therapeutic compositions is contemplated. The pharmaceutical composition may be determined by one skilled in the art based on, for example, the intended route of administration, the form of delivery, and the desired dosage. See, for example, Remington, The Science and Practice of Pharmacy, version 22, 2012.

The compositions of the present invention may be incorporated into injectable formulations. The formulation may also include necessary physiologically acceptable carrier materials, excipients, lubricants, buffers, surfactants, antibacterial agents, fillers (e.g., mannitol), antioxidants (ascorbic acid or sodium bisulfite), and the like.

Suitable formulation materials may include, but are not limited to, amino acids (e.g., glycine, glutamine, asparagine, arginine, or lysine), antibacterial agents, antioxidants (e.g., ascorbic acid, sodium sulfite, or sodium bisulfite), buffers (e.g., borates, bicarbonates, tris hydrochloride, citrates, phosphates, or other organic acids), bulking agents (e.g., mannitol or glycine), chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA)), coffein (e.g., caffeine, polyvinylpyrrolidone, β -cyclodextrin, or hydroxypropyl- β -cyclodextrin), fillers, disaccharides, and other carbohydrate compounds (e.g., glucose, mannose, or dextrin), proteins (e.g., albumin, gelatin, or immunoglobulin), coloring agents, flavoring agents and diluents, emulsifying agents, hydrophilic polymers (e.g., polyvinylpyrrolidone), low molecular weight solubilizing agents such as sodium chloride, sorbitol, or sodium chloride, as a pharmaceutically active sorbitol, sodium chloride, or sodium chloride solubilizing or sodium chloride, sorbitol, or sorbitol excipient (e.g., sorbitol), as sorbitol, sodium chloride, sorbitol.

The primary vehicle or carrier in the pharmaceutical composition may be aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier may be water for injection, physiological saline solution, or artificial cerebrospinal fluid, possibly supplemented with other materials common in parenterally administered compositions. Neutral buffered saline or saline mixed with serum albumin are other exemplary vehicles. The pharmaceutical composition may comprise tris buffer at about pH 7.0-8.5, or acetate buffer at about pH 4.0-5.5, which may further comprise sorbitol or a suitable substitute thereof. The pharmaceutical compositions of the present invention may be prepared for storage by mixing the selected composition of the desired purity with optional formulation agents (Remington, the science and Practice of Pharmacy, Id.) in the form of a lyophilized cake or an aqueous solution.

The cells or composition may be provided by a slow release system, by encapsulation or by an implanted device. The composition may be administered by bolus injection or continuously by infusion or by implantation device. The composition may also be administered topically by implanting a membrane, sponge, or another suitable material that has absorbed or encapsulated cells. Where a grafting device is used, the device may be implanted into any suitable tissue or organ. Injections can be given as a one-time treatment, repeatedly (daily, weekly, monthly, yearly, etc.) to achieve the desired therapeutic effect.

Cell encapsulation methods have previously been described which allow transplantation of encapsulated cells for use in the treatment of Parkinson's disease (Tresco, et al (1992) ASAIO J.38:17-23) or amyotrophic lateral sclerosis (Aebishcher, et al (1996) hum. Gene Ther.7: 851-860). According to this embodiment, the cells are encapsulated by a compound that forms a microporous membrane. Capsules, e.g., about 1cm in length, containing cells of interest can be prepared using hollow microporous membranes made from Polyethersulfone (PES) (Akzo Nobel Faser AG, Wupetal, Germany; D teglon, et al (1996) hum. Gene ther.7: 2135-2146).

These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Prevention of the action of microorganisms can be ensured by including various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like.

Supplementary active ingredients may also be incorporated into the composition. The active compositions of the present disclosure may comprise classical pharmaceutical preparations. Administration of these compositions according to the present disclosure will be by any of the usual routes, so long as the target tissue is available by that route.

As used herein, the term "amount effective for …", "effective amount", or "therapeutically effective amount" refers to an amount of a cell or composition of the invention sufficient to achieve the desired result. The amount of a cell or composition that constitutes an "effective amount" or "therapeutically effective amount" can depend on the severity of the disease; the condition, weight or age of the patient to be treated; the dosing frequency or route of administration will vary, but can be routinely determined by one of ordinary skill in the art. The clinician can set the dosage or route of administration to achieve the optimal therapeutic effect.

The invention also relates to kits for treating immunodeficiency or genetic disorders and/or preventing or alleviating GvHD. The kits can be used to practice the methods of the invention described herein. The kit is a component that includes at least one material or component of the composition of the invention. Thus, in some embodiments, the kit comprises a fusogenic agent for ex vivo cell fusion, and three or more donor cells selected from hematopoietic stem cells, mesenchymal stem cells, myoblasts, pericytes, satellite cells from different donors (e.g., donor cells from a cell bank), and optionally material for obtaining donor cells, as described above.

The exact nature of the components configured in the kit of the invention will depend on their intended purpose. For example, some embodiments are configured for the purpose of treating muscular dystrophy. In one embodiment, the kit is specifically configured for the purpose of treating a human subject. In another embodiment, the kit is specifically configured for the purpose of treating an adult, human subject. In another embodiment, the kit is specifically configured for the purpose of treating children. In another embodiment, the kit is specifically configured for the purpose of treating DMD and may comprise a myoblast and two or more cells independently selected from the group consisting of mesenchymal stem cells, myoblasts and satellite cells. In another embodiment, the kit is specifically configured for the purpose of treating GvHD and may comprise hematopoietic stem cells and two or more cells independently selected from the group consisting of hematopoietic stem cells and mesenchymal stem cells. In another embodiment, the kit is particularly configured for treating sickle cell anemia, and may comprise hematopoietic stem cells and two or more cells independently selected from hematopoietic stem cells and mesenchymal stem cells. In another embodiment, the kit is specifically configured for the purpose of providing a continuous daily dosage. In another embodiment, the kit is specifically configured for the purpose of providing the required dosage for use. In additional embodiments, the kit is configured for veterinary applications to treat subjects such as, but not limited to, farm animals, livestock, and laboratory animals.

The kit may contain instructions for use. "instructions for use" generally include tangible expressions which describe techniques to be used in using the components of the kit to achieve a desired result, e.g., for treating muscular dystrophy, for treating sickle cell anemia, or for treating GvHD. Optionally, the kit further comprises other useful components, such as diluents, buffers, pharmaceutically acceptable carriers, syringes, catheters, applicators, pipetting or measuring instruments, dressing materials, or other useful means, as will be readily recognized by those skilled in the art.

The materials or components assembled in the kit can be provided to the practitioner in any convenient and suitable manner that maintains their operability and usefulness. For example, the components may be in dissolved, dehydrated or lyophilized form; they may be provided at room temperature, refrigerated or frozen temperatures. The components are typically contained in a suitable packaging material. As used herein, the phrase "packaging material" refers to one or more physical structures used to contain the contents of a kit. The packaging material is constructed by well-known methods, preferably for providing a sterile, contaminant-free environment. The packaging material used in the kit is a packaging material generally used for therapy. As used herein, the term "package" refers to a suitable solid substrate or material, such as glass, plastic, paper, foil, and the like, capable of holding the various kit components. The packaging material typically has an external label that indicates the contents and/or purpose of the kit and/or its components.

The following non-limiting examples are provided to further illustrate the invention.

15页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于生物标志物分析的设备、系统和方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!