Cryopreservation of stem cells

文档序号:23270 发布日期:2021-09-21 浏览:26次 中文

阅读说明:本技术 干细胞的低温保存 (Cryopreservation of stem cells ) 是由 埃莱乌特里奥·伦巴第德拉卡马拉 梅塔恩·奥尔蒂斯维伦布拉斯 于 2020-02-11 设计创作,主要内容包括:本发明涉及用于干细胞群,包括间充质干细胞(MSCs)如脂肪来源的基质干细胞(ASCs)的低温保存的方法。更具体地,本发明涉及N-乙酰半胱氨酸(NAC)在低温保存方法中的应用、从所述方法获得的细胞群、包含所述细胞的组合物,及其应用。(The present invention relates to methods for cryopreservation of stem cell populations, including Mesenchymal Stem Cells (MSCs), such as adipose-derived stromal stem cells (ASCs). More specifically, the present invention relates to the use of N-acetylcysteine (NAC) in cryopreservation methods, cell populations obtained from said methods, compositions comprising said cells, and uses thereof.)

1. A method for the cryopreservation of stem cells, the method comprising the steps of:

a. treating a stem cell population with N-acetylcysteine (NAC) to obtain a treated stem cell population; and

b. freezing the treated stem cell population to obtain a frozen stem cell population.

2. The method of claim 1, wherein the method comprises the steps of:

a. treating the stem cell population with NAC to obtain a treated stem cell population;

b. freezing the treated stem cell population to obtain a frozen stem cell population; and

c. thawing the frozen stem cell population to obtain a thawed stem cell population.

3. The method of claim 1 or claim 2, wherein the method comprises the steps of:

a. treating the stem cell population with NAC to obtain a treated stem cell population;

b. washing the treated stem cell population to remove the NAC and obtain a washed stem cell population, and freezing the washed stem cell population to obtain a frozen stem cell population; and

c. thawing the frozen stem cell population to obtain a thawed stem cell population.

4. The method of any of the preceding claims, wherein the processing step comprises:

incubating the stem cell population with NAC for at least about 1, 2, 4, 6, 8, 10, 12, 16, 24, or 48 hours prior to freezing the stem cell population; and/or

Adding NAC to the stem cell population to an initial concentration of approximately 0.5-10mM, optionally wherein the treating step comprises one or more additional additions of NAC to maintain a predetermined level of NAC concentration.

5. The method according to any one of claims 2-4, wherein the method further comprises the steps of:

d. culturing the thawed stem cell population to obtain an expanded stem cell population.

6. The method according to any one of claims 2-4, wherein the method further comprises the steps of:

d. culturing said thawed stem cell population in the presence of NAC to obtain an expanded stem cell population, optionally wherein:

the culturing step comprises adding NAC to an initial concentration of about 0.5-5mM, further optionally wherein the culturing step comprises one or more additional additions of NAC to maintain a predetermined level of NAC concentration; and/or

The method further comprises the step of washing the expanded stem cell population to remove the NAC and obtain a washed and expanded stem cell population.

7. The method of any one of claims 2-6, wherein the method further comprises the steps of washing the thawed stem cell population or the expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier.

8. The method according to any one of claims 6-7, wherein the method further comprises the steps of:

e. freezing the expanded stem cell population or the washed and expanded stem cell population to obtain a frozen expanded stem cell population or a frozen washed and expanded stem cell population; and optionally

f. Thawing the frozen expanded stem cell population or the frozen washed and expanded stem cell population to obtain a thawed expanded stem cell population; and optionally

g. Washing the thawed expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier.

9. A method for the cryopreservation of stem cells, the method comprising the steps of:

a. freezing a stem cell population to obtain a frozen stem cell population;

b. thawing the frozen stem cell population to obtain a thawed stem cell population; and

c. culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population, optionally wherein the culturing step comprises adding NAC to an initial concentration of about 0.5-5mM, further optionally wherein the culturing step comprises one or more additional additions of NAC to maintain a predetermined level of NAC concentration.

10. The method of any one of the preceding claims, wherein the stem cells are Mesenchymal Stem Cells (MSCs), and/or wherein the stem cells are adipose-derived stromal stem cells (ASCs).

11. A population of stem cells obtained by the method of any one of claims 1-10.

12. The stem cell population of claim 11 wherein:

an increase in the number of viable cells after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population;

(ii) at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold increase in the number of viable cells after thawing, as compared to a control stem cell population;

(ii) an increase in growth rate in the stem cell population after thawing of at least about 1.03-fold, 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.6-fold, or at least about 2-fold, as compared to a control stem cell population;

at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% increase in mitochondrial activity after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population;

a reduction in the time taken for recovery of the ASCs after thawing compared to a control stem cell population; and/or

(ii) at least about 1.1-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.6-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, or at least about 5-fold decrease in the number of hours it takes for the cell to recover after thawing relative to a control stem cell population,

wherein the control stem cell population is derived from the same stem cell population as the stem cell population treated with NAC and is not treated with NAC but otherwise subjected to the same conditions.

13. The method of any one of claims 1-10, wherein:

an increase in the number of viable cells after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population;

(ii) at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold increase in the number of viable cells after thawing, as compared to a control stem cell population;

(ii) an increase in growth rate in the stem cell population after thawing of at least about 1.03-fold, 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.6-fold, or at least about 2-fold, as compared to a control stem cell population;

at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% increase in mitochondrial activity after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population;

a reduction in the time taken for recovery of the ASCs after thawing compared to a control stem cell population; and/or

The reduction in the number of hours it takes for the cell to recover after thawing is at least about 1.1-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.6-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, or at least about 5-fold relative to a control stem cell population.

14. A cryopreservation composition comprising a population of stem cells according to claim 11 or claim 12 and a cryopreservation medium, optionally wherein the composition is frozen and/or optionally wherein the composition contains NAC.

Use of NAC for cryopreservation of stem cells.

16. A cryopreservation kit comprising: a frozen vial, a NAC-containing container, and a container comprising a population of stem cells.

Technical Field

The present invention relates to methods for cryopreserving stem cell populations, including Mesenchymal Stem Cells (MSCs), such as adipose-derived stromal stem cells (ASCs). More particularly, the present invention relates to the use of N-acetylcysteine (NAC) in cryopreservation methods.

Background

The global repair and regenerative medicine market requires that the viability and function of therapeutic cells be maintained, that cells be transported from the manufacturing site to the patient, that safety and quality control tests be completed, and that cell banks be formed. Prior to or during use, the cells are cryopreserved or cryogenically maintained before returning to a normothermic temperature. The success of these therapies depends at least in part on the ability to retain not only cellular structure but also cellular function.

Regardless of type, the goal of cell preservation is to stop the biological time for a given period of time and then restore cell viability, structure and function as needed. Ideally, the cryopreserved cells/tissues should have the same characteristics after thawing. In many cases, this goal has not been achieved. Preservation results are generally characterized by a retention of high cell viability, measured immediately after storage, followed by a decline within 24-48 hours, with a concomitant decline in cell reactivity, function and reproductive capacity. For cryopreservation, the storage interval for most cell systems is typically limited to 1-3 days.

Many studies have observed that cellular properties (e.g., cell activity, survival rate, proliferation potential) are affected by the freezing and thawing process. The preservation process exerts a lot of stress on the cells due to temperature dependent uncoupling of metabolic and biochemical processes. These include, inter alia, the generation of free radicals by destruction of oxidative respiration, which are harmful to the cell due to downstream effects of lipid peroxidation, DNA and RNA damage, alterations in cytoskeletal structural components. Alterations in cell membrane structure, fluidity, and organization can also activate membrane receptors, triggering a range of intracellular events, including stimulation of stress response pathways and apoptosis. Na binding by closing the membrane+/K+Pump and Ca2+Ion channels, dysregulation of cellular ionic balance, activate stress response mechanisms including calcium release from intracellular stores, osmotic influx, and cellular swelling. Many additional stress response mechanisms can also be activated by cryopreservation, damaging the cells.

Cryoprotectants such as dimethyl sulfoxide (DMSO), glycerol, or animal-derived serum are typically added to the cryopreservation media to minimize these negative effects. However, there remains a need for improved methods for cryopreservation of stem cells.

Summary of The Invention

The present invention is summarized as providing methods and compositions relating to the cryopreservation of stem cells, including Mesenchymal Stem Cells (MSCs) such as adipose-derived stromal stem cells (ASCs), and uses of such compositions. In particular, to facilitate research and clinical applications of stem cells, the inventors have developed a new cryopreservation approach that involves treating cells with N-acetylcysteine (NAC), which results in an increase in the number of viable cells after thawing, an increase in growth rate, an increase in mitochondrial activity, and/or an improvement in recovery, while maintaining the structural and/or functional properties of the cells, such as those required for their therapeutic applications.

The present invention provides a method for cryopreservation of stem cells, said method comprising the steps of: (a) treating a stem cell population with N-acetylcysteine (NAC) to obtain a treated stem cell population; and (b) freezing the treated stem cell population to obtain a frozen stem cell population. In some embodiments, the method comprises the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; and (c) thawing the frozen stem cell population to obtain a thawed stem cell population. In some embodiments, the method comprises the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) washing the treated stem cell population to remove the NAC and obtain a washed stem cell population, and freezing the washed stem cell population to obtain a frozen stem cell population; and (c) thawing the frozen stem cell population to obtain a thawed stem cell population. In any method, the treating step can include incubating the stem cell population with the NAC for at least about 1, 2, 4, 6, 8, 10, 12, 16, 24, or 48 hours, and then freezing the stem cell population. The treating step may include adding NAC to the stem cell population to an initial concentration range of about 0.5-10 mM. The treating step may include one or more additional additions of NAC to maintain a preselected level of NAC concentration. In some embodiments, the method further comprises the steps of: (d) culturing the thawed stem cell population to obtain an expanded stem cell population. In some embodiments, the method further comprises the steps of: (d) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population. The culturing step may include the addition of NAC to an initial concentration range of about 0.5-5 mM. The culturing step may include one or more additional additions of NAC to maintain a preselected level of NAC concentration. In some embodiments, the method further comprises the step of washing the expanded stem cell population to remove the NAC and obtain a washed and expanded stem cell population. In some embodiments, the method further comprises the steps of washing the thawed or expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier. In some embodiments, the method further comprises the steps of: (e) freezing the expanded or washed and expanded stem cell population to obtain a frozen expanded stem cell population or a frozen washed and expanded stem cell population. In some embodiments, the method further comprises the steps of: (e) freezing the expanded or washed and expanded stem cell population to obtain a frozen expanded stem cell population or a frozen washed and expanded stem cell population; and (f) thawing the frozen expanded or frozen washed and expanded stem cell population to obtain a thawed expanded stem cell population. In some embodiments, the method further comprises the steps of: (g) washing the thawed expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier.

The present invention also provides a method for cryopreservation of stem cells, the method comprising the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; and (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population. The culturing step may include the addition of NAC to an initial concentration of about 0.5-5 mM. In some embodiments, the culturing step comprises one or more additional additions of NAC to maintain a predetermined level of NAC concentration.

In any of the methods of the present invention, the freezing step can comprise reducing the temperature to-70 ℃ to-130 ℃ at a rate of about-0.5 to about-10 ℃/minute. In some embodiments, the freezing step comprises reducing the temperature from +4 ℃ to-100 to-180 ℃ within 10-60 min.

In any of the methods of the invention, the population of stem cells may be thawed at 37 ℃. The cell density of the frozen stem cell population may range from about 1 million to about 5 million cells/mL, preferably about 2500 million cells/mL.

In some embodiments, the population of stem cells is substantially pure. In some embodiments, the stem cells are Mesenchymal Stem Cells (MSCs). In some embodiments, the stem cells are adipose-derived stromal stem cells (ASCs). In some embodiments, the stem cell is a human cell. In a preferred embodiment, the stem cells are human ASCs.

In any of the methods of the invention, the method may further comprise the step of resuspending the cells in a pharmaceutically acceptable carrier. The method may comprise freezing the population of stem cells in a plurality of frozen vials.

In some embodiments, the method comprises the step of replicating any one of the methods of the invention on a plurality of stem cell populations. The method may comprise freezing a plurality of stem cell populations in a plurality of frozen vials. The method may further comprise storing the plurality of cryo-preserved vials in a liquid nitrogen storage container for at least 1 month, at least 2 months, at least 3 months, at least 6 months, or at least 1 year.

The invention also provides a liquid nitrogen storage container comprising a plurality of cryo-preservation vials obtained according to the method of the invention.

The present invention provides a population of stem cells obtained by the methods of the invention.

In any of the methods of the invention or stem cell populations of the invention, the number of viable cells can be increased after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population. In any of the methods of the invention or stem cell populations of the invention, the number of viable cells after thawing can be increased at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold, compared to a control stem cell population. In any of the methods of the invention or stem cell populations of the invention, the growth rate after thawing can be increased at least about 1.03-fold, 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.6-fold, or at least about 2-fold compared to a control stem cell population. In any of the methods of the invention or stem cell populations of the invention, mitochondrial activity may be increased by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% after thawing and optionally culturing for about 1 day and/or about 4 days as compared to a control stem cell population. In any of the methods of the invention or stem cell populations of the invention, the time taken for the ASCs to recover after thawing may be reduced compared to a control stem cell population. In any of the methods of the invention or stem cell populations of the invention, the number of hours it takes for a cell to recover after thawing can be reduced by at least about 1.1 fold, at least about 1.2 fold, at least about 1.4 fold, at least about 1.6 fold, at least about 2 fold, at least about 3 fold, at least about 4 fold, or at least about 5 fold relative to a control stem cell population.

The present invention provides cryopreservation compositions comprising a population of stem cells of the invention and a cryopreservation medium. The composition may be frozen. In some embodiments, the composition contains NAC.

The invention also provides a pharmaceutical composition comprising a population of stem cells of the invention and a pharmaceutically acceptable carrier. The composition may comprise from about 1 million cells to about 1.5 million cells, preferably about 3000 million cells or about 1.2 million cells. In some embodiments, the cell density is about 100-.

The present invention provides for the use of NAC for cryopreservation of stem cells, e.g. in the methods of the invention.

The invention also provides a population of stem cells of the invention, a pharmaceutical composition of the invention or a cryopreserved composition of the invention for use in therapy.

The invention also provides a population of stem cells of the invention, a pharmaceutical composition of the invention, or a cryopreservation composition of the invention for use in a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection, in a patient in need thereof.

The invention also provides a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection, the method comprising administering to a subject in need thereof a stem cell population of the invention, a pharmaceutical composition of the invention, or a cryopreserved composition of the invention.

The present invention also provides a population of stem cells for use in a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; (c) thawing the frozen stem cell population to obtain a thawed stem cell population; (d) optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and (e) administering the population of stem cells to the patient.

The present invention also provides a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; (c) thawing the frozen stem cell population to obtain a thawed stem cell population; (d) optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and (e) administering the population of stem cells to the patient.

The present invention provides a population of stem cells for use in a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and (d) administering the population of stem cells to the patient.

The present invention also provides a method of treating fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and (d) administering the population of stem cells to the patient.

In some embodiments, the stem cell population for use according to the invention or the method of treatment according to the invention further comprises any step of the method of stem cell cryopreservation described herein prior to administration of the stem cell population to a patient.

In some embodiments of the stem cell population, the pharmaceutical composition or the cryopreservation composition for use according to the invention or the method of treatment of the invention, the method comprises administering about 100 to 1.5 million cells, preferably about 3000 million stem cells or about 1.2 million stem cells. The method may comprise administering about 100 to about 1000 ten thousand cells/kg. The methods may comprise injecting a population of stem cells, a pharmaceutical composition, or a cryopreservation composition of the invention. The stem cell may be as defined herein. In some embodiments, the stem cells are allogeneic or autologous. In a preferred embodiment, the stem cells are human allogeneic ASCs.

The present invention provides a cryopreservation kit comprising: frozen vials, NAC-containing containers, and containers containing a population of stem cells.

Brief description of the drawings

FIG. 1 shows a flow chart of an exemplary assay.

FIG. 2 MTS assay of thawed and seeded ASCs treated with various compounds (NAC; LY294,002; sc-79 or Exendin-4) prior to freezing, compared to untreated (NT) cells, 24 hours. Data representing a single experiment in six technical replicates of MTS.

FIG. 3 cell number of thawed, seeded ASCs treated with 6mM NAC (NAC) prior to freezing, compared to untreated (NT) cells 24 hours later. Data representing a single experiment in three technical replicates.

FIG. 4. cell densities (A) at 1, 4 and 7 days, and MTS measurements at 24 hours (B) and 96 hours (C) after thawing after seeding ASCs treated with 6mM NAC (NAC) prior to freezing, compared to untreated (NT) cells. MTS results are expressed as a percentage of absorbance at 490nm relative to untreated cells. Data representing a single experiment of three replicates are shown for cell count, and data representing a single experiment of 6 technical replicates are shown for MTS. The 0 day time point in fig. 4A shows the cell seeding density, rather than the number of viable adherent cells as shown by the other time points.

FIG. 5 is a graph showing the cell density of ASCs from two different donors (donor A (DON A) and donor B (DON B)) 1, 4, and 7 days after seeding after thawing. ASCs were pretreated with 6mM NAC and compared to untreated cells. Data representing one experiment out of three technical replicates.

FIG. 6 is a graph showing cell densities 7, 11 and 14 days after seeding with thawed ASC treated post-thaw with 2, 6 or 12mM NAC added to the seeding medium. Data representing two experiments in three technical replicates.

FIG. 7 ASC characterization by flow cytometry. ASCs (from donor a and treated with 6mM NAC before freezing) were analyzed two weeks after thawing and compared to untreated cells for CD29, CD73, CD90, and CD 105. The percentage of positive cells is shown in the figure. The experiment was performed in triplicate.

FIG. 8 lymphocyte proliferation assay using thawed ASCs from donor A pretreated with 6mM NAC, and compared to untreated cells. The assay was performed at 96 hours using an ASC: PBMC ratio of 1: 75. (A) Overlap between the maximal proliferation of activated PBMCs and PBMCs in the presence of ASC. (B) Comparison between lymphocyte proliferation of NAC-treated and untreated ASCs after thawing. The results are quantified in the lower right panel.

FIG. 9 is a graph showing the schedule and time of co-culture of ASCs and monocytes, and the analysis performed to assess the effect of ASCs on macrophage and mDC differentiation and function.

FIG. 10. 2X micrograph images of mature DC cultures alone or in the presence of thawed ASCs from two different donors (donor A (DON A) and donor B (DON B)) pretreated or untreated with NAC.

FIG. 11. 20 Xmicrograph images of mature DC cultures alone or in the presence of thawed ASCs from two different donors (donor A (DON A) and donor B (DON B)) pretreated or untreated with NAC.

FIG. 12 is a histogram showing phagocytosis of Staphylococcus aureus (Staphylococcus aureus) particles by mDCs in the absence or presence of ASCs from two different donors (donor A (DON A) and donor B (DON B)) pretreated with or without NAC as measured by flow cytometry.

Figure 13 surface expression of phagocytic acceptor CD206 (mannose acceptor) of mdcs in the absence or presence of ASCs from two different donors (donor a (don a) and donor b (don b)) pretreated with or without NAC, as measured by flow cytometry. ASC induces expression of CD14, CD206, and CD163 in mdcs. ASC NAC pretreatment did not alter these effects.

Figure 14 surface expression of phagocytic acceptor CD163 (scavenger acceptor) of mdcs in the absence or presence of ASCs from two different donors (donor a (don a) and donor b (don b)) pretreated with or without NAC, as measured by flow cytometry. ASC induces expression of CD14, CD206, and CD163 in mdcs. ASC NAC pretreatment did not alter these effects.

FIG. 15 dot plots representing surface expression of CD14 and CD1a (antigen presenting molecules) of mDCs in the absence or presence of ASCs from two different donors (donor A (DON A) and donor B (DON B)) pretreated with or without NAC, as measured by flow cytometry. mdcs are CD14-CD1a +, but the presence of ASCs results in a new regulatory CD14+ CD1a-DC population. ASC NAC pretreatment did not alter this effect.

Detailed Description

The present invention relates to methods and compositions for the cryopreservation of stem cells, wherein a population of stem cells is treated with N-acetylcysteine (NAC) prior to freezing ("NAC pretreatment") and/or after thawing of the stem cells ("post-thaw treatment").

The present inventors tested a number of compounds known to modulate apoptotic damage (such as hypoxia, serum deprivation, oxidative stress (e.g., caused by hydrogen peroxide treatment), Fas ligand-induced death, etc.) with the aim of increasing the resistance of cells to the freeze-thaw process. NAC was found to confer advantages on thawed stem cells in terms of increased viable cell number, increased growth rate, increased mitochondrial activity and/or improved recovery compared to untreated control cells. It is useful to increase the number of viable cells available immediately after thawing, for example for acute treatment. These advantages will help facilitate the storage, transport, and handling of stem cell stocks and cell lines, as well as the preparation and transport of cell-based therapies, for example, by reducing the time required to recover and/or expand cryopreserved cells in culture after thawing.

N-acetylcysteine

N-acetylcysteine (NAC), also known as N-acetyl-L-cysteine, is a non-proprietary name for the N-acetyl derivative of the naturally occurring amino acid L-cysteine. It is an antioxidant with a molecular weight of 163.2gmol-1And the chemical structure is as follows:

NAC toAnd sold under trade names. It has been approved for several indications, including the treatment of paracetamol (acetaminophen) overdose (as an injection and oral agent), and as a mucolytic agent to loosen viscous mucus (ingested intravenously, orally or inhaled as a mist) in individuals with cystic fibrosis or chronic obstructive pulmonary disease. NAC has also been used or studied for the treatment of other indications, including liver failure, various cancers, methacrylonitrile poisoning, reduction of radiocontrast-induced nephropathy, and reduction of reperfusion injury during heart bypass surgery.

Pretreatment with NAC

Disclosed herein are methods for cryopreservation of stem cells, the methods comprising treating a stem cell population with NAC, i.e., a "pre-treatment" of the stem cell population, prior to freezing. Thus, "NAC-pretreated cells" refers to cells that have been treated with NAC and then frozen.

Methods of cryopreservation with stem cells may include the steps of: (a) treating a population of stem cells (e.g., ASCs) with N-acetylcysteine to obtain a treated population of stem cells; and (b) freezing the treated stem cell population to obtain a frozen stem cell population.

Treatment of a stem cell population with NAC ("treatment" or "treatment step") is typically performed by adding NAC to the appropriate cell culture medium for the stem cell population. Stock solutions of NAC can be prepared, for example in water, and the NAC can then be diluted to the desired concentration in the culture medium.

The skilled artisan will know the appropriate cell culture media to support the growth of a particular cell type. The cell culture medium may be in liquid or solid form, including gelatinous media such as agar, agarose, gelatin, and collagen matrices. The culture medium may be a "defined medium" which is made of chemically defined (usually pure) components and which does not contain poorly characterized biological extracts, such as yeast extract and beef broth. The culture medium may be a "basal medium" that promotes the growth of many types of microorganisms that do not require any special nutritional supplements. Most basal media typically contain four basic chemical groups: amino acids, sugars, inorganic salts and vitamins. Basal media are often used as the basis for more complex media to which supplements such as serum, buffers, growth factors, lipids, etc. are added. Examples of basal media include, but are not limited to: eagle basal medium, minimal essential medium, Du's Modified Eagle Medium (DMEM), Medium 199, nutrient mixture Ham's F-10 and Ham's F-12, McCoy's 5A, Du's MEM/F-12, alpha modified minimal essential medium (alpha MEM), Rosevir Pack Community Medium (Roswell Park mental Institute Media)1640(RPMI 1640), and Iscove Modified Du's Medium (IMDM). Typically, 0-20% Fetal Bovine Serum (FBS) or 1-20% horse serum will be added to the above medium to support the growth of MSCs. However, if growth factors, cytokines and hormones required for MSCs in FBS are identified and provided in the growth medium at appropriate concentrations, a defined medium may be used. Antibiotics that may be included in the culture medium include, but are not limited to, penicillin and streptomycin. The concentration of penicillin in the chemically defined medium is from about 10 to about 200 units/ml. The concentration of streptomycin in the chemically defined medium is about 10 to about 200. mu.g/ml. For example, a suitable cell culture medium for ASCs is complete DMEM (DMEM/F-12 medium-GlutaMAX)TM-I, Gibco, supplemented with 100. mu.g/mL penicillin/streptomycin and 10% FBS).

The treating step may include adding NAC to the stem cell population to an initial concentration range of about 0.5-10mM NAC, e.g., about 2-8mM or about 4-6 mM. Initial concentrations of 0.5-20mM NAC may also be used, e.g., about 3-15mM NAC, 0.5-12mM, or 4-12mM NAC. In a particularly preferred embodiment, the initial concentration of NAC is about 6 mM. By "initial concentration" is meant the concentration of NAC when added to a population of stem cells. However, it is understood that the initial concentration of NAC may be reduced upon addition to the cell, for example, by degradation or metabolism of the NAC. However, the treating step may include one or more additional additions of NAC, for example to maintain the NAC concentration to the concentration to which the stem cell population is exposed. Thus, a "treatment step" may include treating a stem cell population with an initial concentration of NAC, optionally monitoring NAC levels during the treatment step, and adding one or more additional additions of NAC to maintain the NAC concentration at the initial concentration or at a predetermined level (e.g., the NAC concentration described above).

The treating step can include incubating the stem cell population with NAC for at least about 1, 2, 4, 6, 8, 10, 12, 16, 24, or 48 hours, and then freezing the stem cell population. For example, incubating the stem cell population with NAC can be performed for about 1 to about 48 hours, about 2-24 hours, or about 6-24 hours, and then freezing the stem cell population. The incubation can be performed under any suitable conditions (e.g., where the stem cell population is stable). In a preferred embodiment, the incubation is performed under culture conditions for a particular cell type. For example, the ASCs may be in complete DMEM (DMEM/F-12 medium-GlutaMAX)TM-I, Gibco, supplemented with 100. mu.g/mL penicillin/streptomycin and 10% FBS) with NAC and 5% CO at 37 ℃2And (4) incubating. In one embodiment, the stem cell population is not incubated with NAC throughout the culture period. The culture period is the period between seeding the stem cell population and freezing the stem cell population in the cell culture vessel. In one embodiment, the population of stem cells is incubated in media without NAC addition for a first period of time, followed by a second period of incubation in media with NAC addition.

A population of stem cells that have been subjected to a NAC "treatment step" as disclosed herein is referred to as a "treated stem cell population".

After the treating step, the treated stem cell population is frozen. A population of stem cells that has been frozen as disclosed herein ("freezing step") is referred to as a "frozen stem cell population". A population of stem cells that has been thawed ("thawing step") as disclosed herein is referred to as a "thawed stem cell population". Thus, the method may comprise the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; and (c) thawing the frozen stem cell population to obtain a thawed stem cell population.

Prior to freezing the treated stem cell population, NAC can be removed (i.e., cells are thus no longer exposed to extracellular NAC). Typically, this can be accomplished, for example, by using (1) cell culture media that does not contain NAC (e.g., as used in the treatment step); (2) phosphate Buffered Saline (PBS); and/or (3) freezing the culture medium and washing the stem cell population. A population of stem cells that has been washed ("washing step") as disclosed herein is referred to as a "washed population of stem cells". Washing can also be used as a medium exchange step, so that the cells can be frozen in a different medium, such as a freezing medium. Thus, the method may comprise the steps of: (a) treating a population of stem cells with N-acetylcysteine to obtain a treated population of stem cells; (b) washing the treated stem cell population to remove N-acetylcysteine and obtain a washed stem cell population, and freezing the washed stem cell population to obtain a frozen stem cell population; and (c) thawing the frozen stem cell population to obtain a thawed stem cell population.

Washing the treated stem cell population may be performed by any suitable method. For adherent cells, a solution containing NAC (e.g., culture medium) can be changed to a different solution (e.g., a solution without NAC and/or with frozen medium) by simple pipetting. For suspension cells (including trypsinized adherent cells), the cells can be pelleted, e.g., using a centrifuge, the supernatant removed, optionally washed (e.g., with media or PBS), and then resuspended in a desired media (e.g., media or freezing media). Filtration, ultrafiltration or dialysis may also be used to wash the cells. Methods for trypsinizing adherent cells are known in the art, and suitable methods are exemplified in the examples.

After freeze thawing, the cells may be cultured ("culture" or "culturing step"), for example, to allow for cell recovery and/or to increase cell numbers. The resulting cells are referred to as "expanded stem cell populations". As used herein, the term "expanded" when referring to cells shall be taken to have its ordinary meaning in the art, i.e. cells that have been propagated in vitro. "proliferation" refers to an increase in the number of cells. "proliferation" and "proliferation" refer to the cells undergoing mitosis. Thus, the method may further comprise the steps of: (d) culturing the thawed stem cell population to obtain an expanded stem cell population.

"culturing" as used herein refers to any art-recognized term, i.e., any method of effecting cell growth in a suitable medium. The cells may be cultured by any technique known in the art for culturing stem cells. The culturing step may be on a small scale, a medium scale, or a large scale. A small scale culture can be considered if the total culture volume is less than about 100 mL. A total culture volume of between about 100mL to about 5L can be considered a medium-scale culture. Large scale can be considered if the total culture volume (e.g., in a bioreactor) is greater than about 5L, and possibly greater than 10L, 100L, 500L, or 1000L.

"cell culture" refers to the growth of cells in vitro. In such cultures, cells proliferate, but they do not organize themselves into tissues. "tissue culture" refers to the maintenance or growth of a tissue (e.g., an explant in vitro of an original or adult organ) to maintain its structure and function. "monolayer culture" refers to a culture in which cells are propagated in a suitable medium while primarily adhering to each other and to the substrate. Further, "suspension culture" refers to a culture in which cells are propagated while being suspended in a suitable medium. Likewise, "continuous flow culture" refers to culturing cells or explants in a continuous flow of fresh medium to maintain cell growth, e.g., viability. "confluent cultures" are cell cultures in which all cells are in contact and thus the entire surface of the culture vessel is covered and means that the cells have also reached their maximum density, but confluency does not necessarily mean that division will cease or that population size will not increase.

A discussion of various Culture techniques and their scale-up can be found in Freshney, RI, Culture of Animal Cells A Manual of Basic techniques and Specialized Applications, 7 th edition, Wiley-Blackwell 2016.1. The culturing step can be performed in any type of container (for a review of the manufacture of MSCs, including discussion of different types of containers, see Mizukami et al, "Mesenchyl chemical Cells: From Discovery to Manufacturing and communication" Stem Cells International (2018) article Nos. 4083921,1-13https:// doi. org/10.1155/2018/4083921). Examples of vessels that can be used in the methods disclosed herein include monolayer culture or flat two-dimensional flasks, consisting of single-compartment or multi-layered vessel Cell Factories such as Nunc Cell Factories (Nunc Cell Factories) and Corning Cell Stacks. As an alternative to a flask, a roller bottle may be used, i.e. a cylindrical bottle is placed in a rotating device in which the cells may form a monolayer on about the inner surface of the bottle. Bioreactors suitable for large scale expansion of cells include MSCs (e.g., ASCs), which are commercially available, and may include 2D (i.e., substantially planar) and 3D amplification bioreactors. Examples of such bioreactors that may be used in the methods disclosed herein include, but are not limited to, plug flow bioreactors, perfusion bioreactors, continuous stirred tank bioreactors, or fixed bed bioreactors. The bioreactor may be operated in batch, fed-batch or perfusion mode. Due to the anchorage dependence of MSCs, culture in bioreactors requires the use of microcarriers, usually small beads (100-. Examples of microcarriers include Cytodex-3 microcarriers (GE Healthcare). Cells are typically grown in a humid environment at temperatures between 31 ℃ and 37 ℃. Thus, in some embodiments, the culture of the thawed stem cell population (e.g., MSCs, such as ASCs) is performed in a large scale bioreactor using microcarriers to obtain an expanded stem cell population.

The culture of the thawed stem cell population may be performed in the presence of NAC, e.g., to improve recovery and/or increase cell number. In other words, in addition to pre-treatment with NAC, post-thaw NAC treatment may also be used. Thus, the method may further comprise the steps of: (d) culturing the thawed stem cell population in the presence of N-acetylcysteine to obtain an expanded stem cell population. Culturing the thawed stem cell population may comprise adding NAC to an initial concentration range of about 0.5-5mM NAC, such as about 0.5-4mM or about 1-2mM, preferably about 2mM, under cell culture conditions suitable for the cell type. Further NAC addition may be required to maintain the concentration of NAC in the cell culture medium (e.g., due to degradation or metabolism of NAC). Thus, the culturing step can include adding NAC to the culture medium at an initial concentration, and then further adding NAC to maintain the initial concentration of NAC or to maintain a predetermined level of NAC concentration (e.g., NAC concentration as described above). Further additives may be added as single doses of NAC alone or in combination with other nutrients (e.g. in fed-batch culture). The "culturing step" may also include monitoring the level of NAC and adding one or more further additions of NAC to maintain the initial concentration or predetermined level. Alternatively, NAC can be continuously supplemented, for example in fresh medium during perfusion culture.

NAC may be removed as desired prior to any downstream use of the stem cell population. Thus, the method may further comprise the step of washing the expanded stem cell population to remove the NAC and obtain a washed and expanded stem cell population. The washing step may allow for the medium to be replaced, for example, with a pharmaceutically acceptable carrier, a solution/medium that does not contain NAC, or a freezing medium. Washing may be performed by any suitable method, including centrifugation, filtration, ultrafiltration or dialysis. For adherent cells, a NAC-containing solution (e.g., culture medium) can be changed to a different solution by simple pipetting. For suspended cells (including trypsinized adherent cells), the cells can be pelleted (e.g., using a centrifuge), the supernatant removed, optionally washed (e.g., with culture medium or PBS), and then resuspended in a desired solution (e.g., culture medium, freezing medium, or a pharmaceutically acceptable carrier). Thus, the method may further comprise the steps of: washing the thawed or expanded stem cell population (e.g., of step (c) or (d)) and resuspending the cells (e.g., suspended cells or trypsinized adherent cells) in a pharmaceutically acceptable carrier.

The expanded stem cell population can be frozen, for example, for storage as a cell stock and/or for transport. The method may further comprise the steps of: (e) freezing the expanded stem cell population (e.g., from step (d)) to obtain a frozen expanded stem cell population. The method may further comprise the steps of: (e) freezing the expanded stem cell population to obtain a frozen expanded stem cell population; and (f) thawing the frozen expanded stem cell population to obtain a thawed expanded stem cell population. The method may comprise the steps of: (e) freezing the washed and expanded stem cell population to obtain a frozen, washed and expanded stem cell population. The method may further comprise the steps of: (e) freezing the washed and expanded stem cell population to obtain a frozen, washed and expanded stem cell population; and (f) thawing the frozen, washed and expanded stem cell population to obtain a thawed expanded stem cell population. As discussed above, since the "culturing step" of step (d) may be performed in the presence of NAC, in these cases the expanded stem cell population may be considered to be "pre-treated" with NAC prior to freezing. NAC can be removed by washing as needed, and then freezing and/or washing can be used to change the culture medium, for example, to frozen medium. Optionally, the method may further comprise the steps of: (g) the thawed expanded stem cell population is washed and the cells (e.g., suspended or trypsinized adherent cells) are resuspended in a pharmaceutically acceptable carrier.

The frozen stem cell populations (e.g., ASCs) obtained from the methods discussed above form a stock of seed cells. For example, a population of stem cells can be aliquoted into a plurality of cryovials, e.g., at least about 10, at least about 20, at least about 50, about 100, about 1000, about 2000, about 5000, or more cryovials, and cryopreserved (e.g., in liquid nitrogen storage containers). The individual frozen vials can then be individually thawed for downstream use. The thawed or expanded stem cell population (e.g., ASCs) obtained from the methods discussed above can be a therapeutic stem cell population. For example, a thawed or expanded stem cell population (e.g., ASCs) can be in a suitable formulation (e.g., a pharmaceutical composition comprising a pharmaceutically acceptable carrier) for administration to a patient in need thereof.

The method may further comprise the step of resuspending the cells in a pharmaceutically acceptable carrier.

Post-thaw NAC treatment

Disclosed herein are methods for cryopreservation of stem cells, the methods comprising the steps of: (a) freezing a population of stem cells (e.g., ASCs) to obtain a frozen population of stem cells; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; and (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population. Culturing a thawed stem cell population in the presence of NAC (i.e., post-thaw NAC treatment) can improve recovery and/or increase viable cell numbers.

Culturing the thawed stem cell population may comprise adding NAC to an initial concentration range of about 0.5-5mM NAC, such as about 0.5-4mM or about 1-2mM, preferably about 2mM, under cell culture conditions appropriate for the cell type. Further NAC addition may be required to maintain the concentration of NAC in the cell culture medium (e.g., due to degradation or metabolism of NAC). Thus, the culturing step may include adding NAC to the culture medium at an initial concentration, and then further adding NAC to maintain the initial concentration of NAC or to maintain a predetermined level of NAC concentration (e.g., NAC concentration for post-thaw processing as described above). Further additions may be added as a single dose, optionally in combination with other nutrients (e.g. in fed-batch cultures). The "culturing step" may also include monitoring the level of NAC and adding one or more further additions of NAC to maintain the initial concentration or predetermined level. Alternatively, NAC can be continuously supplemented, for example, in fresh medium provided during perfusion culture.

The method may further comprise the step of resuspending the cells in a pharmaceutically acceptable carrier.

Low temperature preservation

Herein, the term "cryopreservation" is used to describe the storage of cells in a low temperature environment, i.e. in the range of-70 ℃ to-196 ℃. These temperatures are suitable for long-term storage (months to years). As discussed herein, the use of the terms "freezing" to "freeze" and "freeze" in the context of stem cells refers to the act of exposing the cells to such low temperatures, and the cells have been subjected to such low temperatures.

Typically, after cooling, as the external medium freezes, the cells reach equilibrium by losing water, thereby increasing the intracellular solute concentration. Intracellular freezing may occur below about-10 to-15 ℃. Both intracellular freezing and solution effects are responsible for cell damage. Physical damage caused by extracellular ice is primarily the result of plasma membrane damage caused by osmotic dehydration of cells.

Once the system is frozen, not all biological processes stop. During freezing, the cells are maintained in a biochemically active, unfrozen state while encapsulated in a frozen ice matrix. The cells do not enter the glassy state until the temperature drops below the glass transition point (Tg) of the cryoprotectant/cell solution mixture (typically below-100 ℃), where biochemical and biomolecular activities cease.

During freezing and subsequent thawing, when the temperature is above Tg, a series of important molecular and biochemical events occur within each cell, which greatly affect its post-thaw viability and function. In this temperature range (from about +15 ℃ to-99.9 ℃), many similarities can be seen in the cellular reaction mechanism between cryopreservation and cryopreservation. Such events include the formation of free radicals, uncoupling of biochemical pathways, accumulation of intracellular waste, disruption of ionic gradients, protein denaturation and degradation, and enzymatic cleavage and activation. These and other events may activate apoptotic and/or necrotic cell death pathways, which may lead to delayed onset cell death phenomena. This can be observed as a disjunction between survival measured immediately after storage and true survival after 24-48 hours.

Low-temperature preservation culture medium

The stem cell populations (e.g., ASCs) may be frozen in cryopreservation media ("freezing media"). The culture medium may retain (to some extent) one or more characteristics (e.g., viability) of the cells after freeze thawing and/or may aid in recovery. The cryopreservation media can contain NAC at a concentration of, for example, about 0.5-10 mM. In one embodiment, the cryopreservation media does not contain NAC. The cryopreservation media typically contains one or more cryopreservatives such as DMSO, PVP, sericin or methylcellulose, and/or may contain commercially available cryopreservation solutions. One or more cryopreservatives or cryopreservation solutions can be added to the stem cell culture medium, such as DMEM, to produce a cryopreservation medium. In one embodiment, the cryopreservation media does not contain any added growth factors. In one embodiment, the cryopreservation media does not contain any added EGF and bFGF. In one embodiment, the cryopreservation media is free of added sodium selenite. In one embodiment, the cryopreservation is NAC-free and does not contain any added growth factors. In one embodiment, the cryopreservation medium is NAC-free and free of any added EGF and bFGF. In one embodiment, the cryopreservation media is NAC-free and does not contain any added sodium selenite. In one embodiment, the cryopreservation media is NAC-free and does not contain any added growth factors and does not contain any added sodium selenite. In one embodiment, the cryopreservation medium is NAC-free and EGF and bFGF-free and free of any added sodium selenite.

Cryopreservatives (or cryoprotectants) are ideally non-toxic, can protect cells during freezing, replace water, and/or have a high glass transition temperature. Without wishing to be bound by theory, it is hypothesized that cryoprotectants protect cells from freezing, inter alia, by the following mechanisms: equilibrating external osmotic pressure, stabilizing biomolecules via preferential rejection, forming a protective glass around biomolecules, and preventing destructive phase transitions in lipid membranes, among other things.

In the past, DMSO, glycerol, and animal serum have been used as cryoprotectants.

DMSO is typically added to the cryopreservation media in the range of 1% -20% (v/v), such as 5% -15%, i.e. about 1%, 2%, 5%, 10% or 20%. A final concentration of about 10% is particularly preferred.

DMSO may be used in combination with serum, i.e., fetal calf/bovine serum (FCS/FBS) or human serum. For example, cryopreservation media may contain 20% -95% serum (human or FCS) and 5% -15% DMSO. Particularly preferred cryopreservation media (e.g., for MSCs, such as ASCs) for use in any of the methods described herein contain about 10% DMSO and about 90% FCS (or FBS). For example, a cryopreservation media for a population of MSCs such as human ASCs can contain 5% -15% DMSO in FBS. The freezing medium for the human embryonic stem cell population can contain 10% DMSO, 30% FBS, and 60% conditioned HES medium.

DMSO may be used in combination with human serum albumin. For example, the cryopreservation media can contain about 2% -10% human serum albumin and about 5% -15% DMSO. A particularly preferred cryopreservation medium contains about 10% DMSO and about 5% human serum albumin.

Other molecules, such as glycerol, ethylene glycol, hydroxycellulose or the disaccharide sucrose, maltose and trehalose, have been shown to enhance cell viability when combined with DMSO in freezing medium.

Trehalose is a disaccharide found in high concentrations in a variety of organisms that can survive almost complete dehydration, and has been shown to stabilize certain cells during freezing. Trehalose is believed to maintain the thermodynamic stability of the membrane by preserving phospholipid head-group spacing and inhibiting lipid phase transition and separation during freezing. Trehalose is characterized by its low tendency to penetrate lipid bilayers and must be loaded into cells by endocytosis or other methods of temporary disruption of the cell membrane. For example, cryopreservation media for ASCs may comprise trehalose at a concentration of about 50-200mM, such as about 100 mM. Trehalose can be used to reduce potential toxicity associated with other cryoprotectants, for example when used in combination with DMSO at the concentrations discussed above (see, e.g., Buchanan et al cell Preservation Technology (2005)3(4): 212-.

Polyvinylpyrrolidone (PVP), sericin and maltose, and Methylcellulose (MC) are optional cryopreservatives. These compounds have been tested as cryopreservation solutions for, for example, ASCs, as a replacement for DMSO or animal-derived serum (Miyagi Shiohira et al cell Medicine (2015)8: 3-7).

PVP is a high molecular polymer that lowers the freezing point and inhibits the increase in extracellular salt concentration, thereby stabilizing the cell membrane during freezing and thawing. PVP can be added to the cryopreservation media at a level of about 1% to 40%, such as about 8% to 25%, for example about 1%, 5%, 10%, 20%, or 40%. In addition to PVP, the cryopreservation media can also contain human serum, optionally at about 5% -20% (e.g., 10% human serum). For example, a cryopreservation media for ASCs may contain 10% PVP and 10% human serum.

MC is a high molecular polymer that can replace serum of animal origin in cryopreservation solutions, although the presence of DMSO (or another cryopreservative) is necessary to maintain cell viability after the freeze-thaw process. The cryopreservation media can contain about 0.5% to 2% w/v MC, e.g., about 1% w/v MC, in combination with a suitable concentration of DMSO as discussed above. For example, the cryopreservation media can contain about 1% MC and about 10% DMSO.

Sericin is a cocoon-derived protein that can also replace animal-derived serum in cryopreservation solutions. The cryopreservation media can contain about 0.5% to 2% w/v sericin, for example about 1% w/v sericin. Sericin can be used in combination with maltose (e.g., 50-200mM maltose) and/or DMSO at suitable concentrations as discussed above. For example, a cryopreservation medium can contain about 1% sericin, 100mM maltose, and 10% DMSO.

There are various commercially available cryopreservation solutions, for example: FM-1(Kyokuto Pharmaceutical Industrial Co., Ltd, Tokyo, Japan), cell banker cryoprotectant series (Nippon Zenyaku Kogyo Co., Ltd., Fukushima, Japan); cryostor (stem Cell technologies); Synth-a-Freeze cryopreservation Medium (Thermo Fisher Scientific) and MesenCultTMACF cryoculture medium (Stem Cell Technologies).

The Cell banker cryoprotectant series allows rapid cryopreservation of cells at-80 ℃, and its use correlates with improved survival after freezing and thawing. Cell bankers 1 and 1+ containing serum can be used for cryopreservation of almost all mammalian cells. In addition, non-serotype cell banker2 allowed cryopreservation of cells under serum-free culture conditions. On the other hand, stem cellbank marker 3 is a chemically defined cryopreservation solution for cells, free of xenobiotics (i.e., free of non-human animal products), and optimized for stem cell preservation performance, such as somatic cells and induced pluripotent stem cells.

Series (BioLife Solutions, Inc.) is a serum-free, animal component-free and defined cryopreservation medium containing various concentrations of DMSO (CS 1010% DMSO; CS 55% DMSO; CS 22% DMSO).CS10 has been used for cryopreservation of MSCs (including ASCs), embryonic stem cells (ES) and induced pluripotent stem cells (iPS). Synth-a-Freeze cryopreservation medium (Thermo Fisher Scientific) has been used to cryopreserve induced pluripotent stem cells (iPS).

Cell-specific cryopreservation media are also available, such as mFresR of ES and iPS cellsTMAnd FresrTM-S cryopreservation Medium, MesenCult of MSCsTMACF cryoculture medium and STEMdiff of neural progenitor cells derived from ES/iPS cellsTMFreezing culture medium of neural progenitor cells. For example, MSCs can be cryopreserved in mesncultTMACF cryoculture medium (Stem Cell Technologies), which may be found in MesenCultTM-ACF Plus or MesenCultTMMSCs were cryopreserved after culturing MSCs in culture medium (Stem Cell Technologies)

Exemplary cryopreservation media and cryoprotectants for various stem cell types are shown in the following table:

additional details regarding cryopreservation of MSCs are provided, for example, in Marquez-Curtis et al (Cryobiology (2015)71(2): 181-.

Freezing protocol and storage conditions

The freezing rate must be fast enough to avoid solute and electrolyte imbalances that lead to cell dehydration and damage, and slow enough to prevent the formation of extracellular and intracellular ice crystals. Cryoprotectants lower the freezing point of the medium, and therefore the mixture of cells and cryopreservation medium containing cryoprotectants is a eutectic system, since the combined freezing point is lower than the individual components. During freezing, fluid moves from the lower solute concentration in unfrozen cells into the partially frozen medium, while the plasma membrane prevents the entry of extracellular ice crystals. Slow freezing allows the liquid to move out of the cells at a rate that results in an osmotic equilibrium between the cells and the culture medium until the culture medium is frozen. If the rate is too slow, the cells may be severely dehydrated or their plasma membranes may be irreversibly damaged. If the rate is too high, fluid migration is insufficient and the cells retain high levels of freezable water during cryopreservation, which results in fatal intracellular ice damage.

In the methods described herein, the population of stem cells can be frozen using a mechanical or controlled rate freezer. The controlled rate freezer can be programmed to cool the cells to about-80 ℃ at a specific rate. Typical freezing rates for cryopreservation of most cells (including MSCs) to-80 ℃ are-1 ℃/min. For example, the freezing rate may be achieved by isolating the stem cell population prior to placing it in a mechanical-80 ℃ freezer, for example using closed cell polyethylene foam containers (e.g., such asBioCision), a styrofoam container, or an isopropyl alcohol (IPA) filled container (e.g., mrTM(Thermo Scientific))。Frosty and mrTMA freezing rate of-1 deg.c/min is specified. However, freezing protocols may require optimization for a given cell type or cell line to achieve maximum viability and maintenance of function after thawing. In the methods described herein, the freezing step can be performed at a rate of about-0.5 to about-10 deg.C/minute, preferably about-3 to about-5 deg.C/minute, such as about-1, -2, -3, -4, -5, or-10 deg.C/minute. The final freezing temperature can beBetween about-70 ℃ and about-130 ℃, therefore, in the disclosed method, the freezing step can comprise reducing the temperature to between-70 ℃ and-130 ℃ at a rate of about-0.5 to about-10 ℃/minute. The temperature can be reduced from +4 ℃ to-100 ℃ and 180 ℃ within 10-60 minutes.

The stem cell population can be frozen at any cell density. The preferred cell density of the frozen stem cell population ranges from about 100 to about 5000 ten thousand cells/mL, preferably about 2500 ten thousand cells/mL.

After freezing, the frozen cell population can be stored in liquid nitrogen at-196 ℃ until needed. The heat-dependent metabolic processes do not normally occur below-100 ℃, and stem cells are therefore in a state of metabolic arrest in liquid nitrogen. For temperatures above-100 ℃ where the low temperature mechanical stress is not too severe, a variety of vessels can be used. However, when storing substances at liquid nitrogen temperatures, containers specially designed to withstand cryogenic temperatures (i.e., "cryovials") must be used. There are a variety of containers on the market that are specifically designed for cryogenic applications, including plastic frozen vials (e.g., with screw top caps) or glass ampoules (which may be flame sealed). Common sizes are 1.2, 2.0, 4,5, 10 and 15mL frozen vials (see, e.g., forAnda vial). Typically, 0.5-1.0mL of cell suspension is placed into a 1.2 or 2.0mL vial. Liquid nitrogen storage containers of various sizes and types are commercially available (see, e.g., Thermo Scientific)TM LocatorTMPlus System and CryoExtraTMHigh efficiency cryogenic storage systems).

In a preferred embodiment, the cell population (e.g., ASCs) is frozen in cryopreservation media (e.g., 10% DMSO in FBS) at-80 ℃ in one or more cryovials, and then transferred to a liquid nitrogen storage vessel.

The methods of stem cell cryopreservation described herein may comprise freezing a population of stem cells, such as ASCs, in a plurality of frozen vials. The stem cell population in each of the plurality of frozen vials may be the same, i.e., an aliquot of a single stem cell population obtained from any of the methods disclosed herein. In some cases, the method may further comprise the step of repeating any of the stem cell cryopreservation methods described herein on a plurality of stem cell populations. The repeated steps can be carried out in sequence, i.e. following the preceding method steps. Alternatively, the repeated steps may be performed in parallel, i.e. the method steps are performed simultaneously on a plurality of stem cell populations. Each repetition may include the same method steps, or may include different method steps, as described herein. The plurality of stem cell populations may comprise stem cell populations (e.g., ASCs) obtained from the same donor (e.g., where different populations are obtained by using the same method steps described herein in separate procedures, or by using different methods as described herein). The plurality of stem cell populations may be stem cell populations obtained from different donors (e.g., ASCs). Alternatively, the plurality of stem cell populations may comprise different types of MSCs. For example, the plurality of stem cell populations may comprise one or more, two or more, three or more of the following MSCs: MSCs derived from bone marrow, umbilical cord, dental pulp, blood (e.g., peripheral, umbilical cord, or menstrual), placenta, and fat. The methods may further comprise freezing the plurality of stem cell populations in a plurality of cryovials. The method may further comprise storing the plurality of frozen vials in a liquid nitrogen storage container for at least 1 month, at least 2 months, at least 3 months, at least 6 months, or at least 1 year. The frozen vials can be frozen at-80 ℃ and then transferred to liquid nitrogen storage containers. The plurality of cryovials is more than 1 cryovial, e.g., at least about 10, at least about 20, at least about 50, about 100, about 1000, about 2000, or about 5000 or more cryovials.

Also provided herein are liquid nitrogen storage containers containing a plurality of cryo-preservation vials obtained according to the methods described herein.

Vitrification is another form of cooling that involves very rapid (> -1000 ℃/sec) cooling of cells immersed in cryopreservation media in an open storage vessel. Rapid freezing can be achieved by plunging the sample in the freezing vial into liquid nitrogen. This process inhibits ice formation, although it requires cryoprotectants at potentially cytotoxic concentrations and the use of open containers risks contamination. Vitrification human embryonic stem cells (hESCs) have been successfully cryopreserved. Capillary vitrification of human embryonic stem cells in cryopreservation media containing DMSO and ethylene glycol has been shown to increase the survival of cryopreserved cells by more than an order of magnitude compared to slow freezing and fast thawing methods. Briefly, hEScs colonies (100-400 cells) were placed in cryopreservation media containing 20% DMSO, 20% ethylene glycol, and 0.5M sucrose after equilibration in lower DMSO and EG solutions. The colonies were loaded into a pipette and dropped into liquid nitrogen.

Thawing protocol

Typically, cells are thawed at or near their growth temperature, e.g., -37 ℃. Thus, in the methods disclosed herein, the stem cell population can be thawed at 37 ℃.

During freezing and thawing, cells pass through an ice crystal formation temperature of-15 ℃ to-60 ℃. Rapid thawing by immersion in a 37 c water bath at a rate of about 90-100 c/min is typically employed to prevent ice crystal formation. However, thawing at lower temperatures or slower rates can reduce certain types of damage, such as oxidative stress detected by adhesion-mediated signaling, while allowing the membrane to seal any pores formed by ice crystallization. In the methods described herein, the stem cell population is typically thawed at 37 ℃. This rapid thawing step can be achieved by placing the cells in the frozen vials in a water bath at 37 ℃. However, the thawing protocol may need to be optimized for a given cell type or cell line to achieve maximal viability and/or maintenance of cell function.

Thawed cells can be washed to remove cryopreservation media and then cultured. Examples of washing methods discussed above (e.g., with respect to removal of NAC and/or media replacement) are also suitable for this purpose.

Post-thaw evaluation

Post-thaw evaluation of stem cell populations (e.g., to examine the effects of NAC pretreatment or post-thaw treatment) may include one or more (or all) of the following tests: cell viability, morphology, cell surface marker assessment, differentiation assays, and analysis of other functional properties. Exemplary evaluations are provided in the examples.

Vitality of the body

As used herein, the term "viable" or "viable" refers to cells that are capable of normal growth and development after cryopreservation and thawing. Thus, assessing the viability of a stem cell population relative to a similar stem cell population that has not been treated with NAC pretreatment, post-thaw NAC treatment, or both, can be used to confirm that the cells have not been negatively affected (i.e., have reduced viability) as a result of NAC pretreatment and/or post-thaw treatment (however, NAC pretreatment and/or post-thaw treatment may have a positive impact on viable cell number, growth rate, recovery rate, and the like, as discussed further below).

Examples of experiments that can be used in the disclosed methods to determine the level of cell viability include trypan blue staining and MTS assays, as discussed in the examples. The MTS assay is a measure of functional viability (i.e. metabolism), whereas the trypan blue assay measures structural viability (i.e. membrane integrity). Other methods known to those skilled in the art, such as the alamar blue assay, may also be used for the measurement of cell viability.

The MTS assay is a colorimetric method for determining the number of viable cells in a proliferation or cytotoxicity assay. For example, CellTiterThe AQueous One Solution reagent contains a novel tetrazolium compound [3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfophenyl) -2H-tetrazolium, inner salt; MTS (a)]And an electron coupling reagent (phenazine ethosulfate; PES). PES has enhanced chemical stability, enabling it to be combined with MTS to form stable solutions. This convenient "One Solution" format is for the first version of CellTiterAn improvement in the AQueous assay wherein Phenazine Methosulfate (PMS) is used as the electron coupling reagent and the PMS solution and MTS solution are provided separately. MT (multiple terminal)The tetrazolium compound (Owen reagent) is bioreduced by metabolically active cells into colored formazan products, which are soluble in tissue culture medium. Can be prepared by directly mixing a small amount of CellTiterThe measurement was performed by adding the AQueous One Solution reagent to the culture well, incubating for 1-4 hours, and then recording the absorbance at 490nm with a 96-well plate reader.

Ability to differentiate

After cryopreservation, in order for stem cells to be suitable for various therapeutic applications, the cells must remain viable, remain in an undifferentiated state and retain their differentiation capacity. Any differentiation will limit their use in downstream applications. Thus, assessing the differentiation ability of a stem cell population relative to a similar stem cell population without NAC pretreatment, NAC treatment after thawing, or both, can be used to confirm that the characteristics of the cells are not affected by NAC pretreatment and/or NAC treatment after thawing.

As used herein, the term "differentiation" or "differentiation" refers to the process by which pluripotent or multipotent (unspecified) stem cells are converted into more specialized cell types.

One method of determining the differentiation potential or pluripotency of an embryo or induced pluripotent stem cell is to measure the levels of surface markers such as OCT4 and SSEA-4, for example by immunofluorescence microscopy (Xu, c., et al, (2001) Nat biotechnol.19: 971-. OCT4 and SSEA-4 are markers of undifferentiated stem cells (i.e., have the potential to differentiate into other lineages). OCT4 is an embryonic gene transcription factor that plays a role in controlling developmental pluripotency, and thus differentiation occurs when OCT4 gene activity is inhibited in pluripotent stem cell differentiation. SSEA4 expression can also be determined by flow cytometry.

MSCs have the ability to differentiate into different tissues such as bone, cartilage, tendon and adipose tissue. They are considered multipotent adult progenitor cells because their differentiation potential is more limited than that of pluripotent/totipotent stem cells, such as embryonic or induced pluripotent stem cells, which have the potential to differentiate into all adult tissues (Jiang et al, (2002) Nature 418(6893): 41-49). Methods for testing the differentiation potential of MSCs in different tissues are known in the art (e.g., Guilak et al, J Cell Physiol. (2006)206(1): 229-an 237; Zuk et al, Mol Biol Cell. (2002)13(12): 42794295).

Cell morphology and/or size

The phenotype of the stem cell population may be assessed by morphology and/or size. The term "phenotype" refers to an observable characteristic of a cell, such as size, morphology, protein expression, including cell surface markers, and the like. Thus, assessing the cell morphology and/or size of a stem cell population relative to a similar stem cell population that has not been treated with NAC pretreatment, post-NAC thawing treatment, or both, can be used to confirm that the characteristics of the cells are not affected by NAC pretreatment and/or post-thawing treatment.

Cell morphology and/or size can be viewed and imaged using an inverted culture microscope.

Human iPSCs and ESCs have similar characteristics including morphology, proliferation, surface markers, gene expression, in vitro differentiation capacity and teratoma formation (see, e.g., Thomson et al science (1998)282(5391): 1145-.

Depending on the tissue of origin, MSCs are morphologically and immunophenotypically similar but not identical (Colter et al, Proc. Natl Acad. Sci. USA (2000)97(7): 3213-.

Characterization of cell surface markers

Phenotypic characterization of the stem cell population may be performed by analysis of one or more cell surface markers. Thus, assessment of the expression of one or more cell surface markers on a population of stem cells relative to a similar population of stem cells that have not been subjected to NAC pretreatment, NAC post-thaw treatment, or both can be used to confirm that the characteristics of the cells are not affected by NAC pretreatment and/or post-thaw treatment

The presence or absence of antibodies that bind to a cell surface marker of interest can be determined by different methods, including but not limited to immunofluorescence microscopy, radiography, and flow cytometry. The determination of the antibody surface marker expression profile may be direct, using a labeled antibody, or it may be indirect, using a second labeled antibody directed against the first specific antibody of the target cell marker, thereby achieving signal amplification. In flow cytometry, the level of a fluorescent dye can be correlated with the number of cell surface markers that specifically bind to the antibody by using a labeled antibody. Differential expression of one or more cell surface markers in a stem cell population allows for identification and/or isolation of the population, for example using FACS (fluorescence activated cell sorting).

For example, according to the International Society for Cellular Therapy, the minimum criteria defining MSCs may be expression of CD105, CD73, CD44 and CD90, and lack of expression of CD45, CD14 or CD11b, CD79 α or CD19 and HLA class II (Dominici et al, Cytotherapy. (2006)8(4): 315-7). Examples of antibodies that can be used to assess CD73, CD90, and CD105 markers are provided in example 5. Antibodies useful for assessing other markers are commercially available, for example from Beckton Dickinson, examples of which are listed below.

Marker substance Fluorescent dyes Sources of antibodies
CD45 FITC Mouse IgG1k
CD34 APC Mouse IgG1
CD14 APC Mouse IgG2ak
CD11b PE Mouse IgG1k
CD79α PE Mouse IgG1k
CD19 APC Mouse IgG1
HLA class II APC Mouse IgG1

For example, post-thaw assessment of a population of ASCs can be performed by examining expression of CD29, CD73, CD90, and CD105 (e.g., as in example 5). This assay can be used to confirm that the properties of the cells are not affected by NAC pretreatment or post-thaw treatment.

Cell surface markers associated with a particular stem cell type are known and exemplified below.

Other functional characteristics

Assessing other functional characteristics of the stem cell population (relative to a similar stem cell population without NAC pretreatment, NAC post-thaw treatment, or both) can be used to confirm that the characteristics of the cells are not affected by the pretreatment and/or NAC post-thaw treatment. For example, for ASCs, other functional characteristics that may be evaluated include: the ability of ASCs to inhibit the proliferation of stimulated lymphocytes (e.g., as in example 6); immunomodulatory capacity of ASCs, e.g., to differentiate monocytes (e.g., as in example 7); the ability of ASCs to regulate phagocytosis, such as staphylococcus aureus granules, by mature dendritic cells (mDCs); ASC-mediated upregulation of one or both of CD206 and CD163 on the cell surface of mDCs (e.g., as in example 9); and/or ASC mediated modulation of CD14-CD1a + mDCs to CD14+ CD1a-mDCs (e.g., as in example 9).

Thus, in any of the methods disclosed herein, the population of thawed ASCs can be assessed for one or more, two or more, three or more, four or more, five or more, six or more, or all seven of the following characteristics: (1) cell viability; (2) expression of cell surface markers CD29, CD73, CD90 and CD 105; (3) the ability to inhibit proliferation of stimulated lymphocytes; (4) immunoregulatory effects on monocyte differentiation; (5) the ability to modulate phagocytosis of mature dendritic cells, such as staphylococcus aureus particles; (6) the ability to upregulate one or both of CD206 and CD163 on the cell surface of mDCs; and (7) modulating CD14-CD1a + mDCs through CD14+ CD1a-mDCs, for each property, can be evaluated relative to a population of similar ASCs without NAC pretreatment, after NAC thawing treatment, or both, to allow confirmation that the property of the cells is not affected and/or that cell viability is not negatively affected (i.e., decreased cell viability) by NAC pretreatment and/or after NAC thawing treatment. Similarly, populations of ASCs obtained by any of the methods described herein are also disclosed that have one or more, two or more, three or more, four or more, five or more, six or more, or all seven of these properties (e.g., as evaluated relative to a similar population of ASCs without NAC pretreatment, NAC post-thaw treatment, or both, as discussed above).

Type of Stem cell

The Stem Cell population may be a population of pluripotent Stem cells or Mesenchymal Stem Cells (MSCs), such as bone marrow-derived, umbilical cord tissue-derived, blood-derived (including umbilical cord blood-derived), menstrual, dental pulp-derived, placenta-derived, or adipose-derived MSCs (Huang et al, J.Dent.Res. (2009)88(9) 792-806; Carvalho et al, curr.Stem Cell Ther. (2011)6(3) 221-8; Harris et al, Curr Stem Cell Res The (2013)8(5) 394-9; Li al, Ann.N.Y. Acad.Sci. (2016)1370(1) 109-18). In a preferred embodiment, the stem cells are human cells (e.g., human ASCs). In a preferred embodiment of the invention, the population of stem cells are adipose-derived stromal stem cells (ASCs). The ASCs used in the cryopreservation methods described herein can be an expanded population of ASCs.

Methods for producing and culturing stem cell populations according to the invention are well known.

The population of stem cells can be substantially pure. The term "substantially pure" in reference to a population of stem cells (e.g., a population of MSCs such as a population of ASCs) refers to a population of stem cells that is at least about 75%, typically at least about 85%, more typically at least about 90%, and most typically at least about 95% homogeneous. Homogeneity can be assessed by morphology and/or by cell surface marker characteristics. Techniques for assessing morphological and cell surface marker characteristics are disclosed herein.

Pluripotent stem cells

Pluripotent stem cells come from two sources. First, Embryonic Stem Cells (ESCs) are derived from the internal cell mass of the embryo pre-implantation blastocyst and pluripotency is controlled by the intrinsic regulatory network of the core transcription factor, the octamer-binding transcription factor 4(OCT4), the sex-determining region Y-box 2(SOX2) and the Nanog homeobox (Nanog). In one embodiment, embryonic stem cell lines are used. Embryonic stem cell lines include constantly dividing cells produced from a set of parental cells harvested from a single embryo. The embryonic stem cell lines used in the present invention are not obtained by disrupting human embryos. Embryonic stem cell lines are commercially available, for example, from ATCC. Embryonic stem cells of the embryonic stem cell line do not lose their pluripotency during the culturing process. In particular, embryonic stem cells of the embryonic stem cell line do not differentiate during the culture process. Second, Induced Pluripotent Stem Cells (iPSCs) derive from ectopic or elevated expression of the four transcription factors OCT4, SOX2, Kruppel-like factor 4(KLF4), and the MYC proto-oncogene (C-MYC) that is essential for inducing pluripotency in somatic cells.

Techniques for isolating stable (undifferentiated) Embryonic Stem cells, such as Human Embryonic Stem Cell cultures, are well established (e.g., U.S. Pat. No. 5,843,780; Thomson et al, Science (1998)282: 1145-. In one embodiment, the method of obtaining embryonic stem cells does not comprise disrupting one or more human embryos.

Since their discovery by Yamanaka in 2007, techniques for generating iPSCs have been well established (e.g., Takahashi et al, Cell (2007)131(5): 861-72). Since then, new and improved methods for iPSC generation have been developed, including non-integrated and feeder-free methods and automated high-throughput derivatization (Paull et al, Nature Mehtods (2015)12(9): 885-.

ipscs are characterized by expression of a series of pluripotency markers: NANOG, SOX2, SSEA4, TRA1-81, TRA1-60, and lack lineage specific markers. Pluripotency of ipscs is demonstrated by their ability to differentiate into three germ layers in an embryoid body assay, and posterior analysis of differentiation markers Tuj1 (ectodermal marker), SMA (mesodermal marker) and SOX17 (endodermal marker) from the three germ layers was performed by immunohistochemistry (Paull et al, Nature Mehtods (2015)12(9): 885-.

MSCs

"mesenchymal stem cells" (also referred to herein as "MSCs") are multipotent stromal cells. They are generally derived from connective tissue and are non-hematopoietic cells. The population of MSCs (according to Dominici et al 2006(Cytotherapy 8(4):315-317) can (1) adhere to plastic under standard culture conditions (e.g., minimal essential medium plus 20% fetal bovine serum), (2) express (i.e., greater than or equal to 80% of the population of MSCs) CD105, CD90, CD73 and CD44, (3) lack of expression of CD45, CD14 or CD11b, CD79 alpha or CD19 and HLA DR (HLA class II) (e.g., less than or equal to 5% of the population of MSCs), and (4) be capable of differentiating into osteoblasts, adipocytes and chondroblasts.

MSCs can be obtained using standard methods from, for example, bone marrow, umbilical cord tissue and blood, menses, dental pulp, umbilical cord blood, placenta, and adipose tissue.

Although MSCs obtained from different tissues are similar, there are some differences in phenotypic and functional characteristics. For example, the expression levels of the cell surface markers CD54 and CD106 may differ depending on the source/origin of the MSCs. These can be measured by flow cytometry. The mRNA levels of some genes such as SOX2, IL1 α, IL1 β, IL6, and IL8 may be differentially expressed by MSCs from different tissues and may be measured by conventional methods. IL6 and PGE2 secretion may also differ between MSCs from different sources, and thus cells may have different regulatory capacities (see, e.g., Yang et al plos ONE (2013)8(3) e 59354).

Bone marrow derived MSCs (BMSCs)

Bone marrow mesenchymal stem cells (BM-MSCs) are similar to MSCs from other tissue sources. However, there were some differences in phenotypic and functional characteristics of MSCs from other tissue sources, such as umbilical cord MSCs, placental MSCs, endodontic MSCs, and menstrual MSCs. Although their lowest characterization criteria are the same, including their ability to adhere to plastics, lowest surface feature markers, and the ability to differentiate into bone, cartilage, tendon, and adipose tissue, they all have some subtle differences. These properties include different expression levels of surface markers such as CD105, different levels of secreted soluble factors associated with their immunomodulatory and regenerative potential, and in general, slightly different functional properties that may make each source or origin more suitable for a particular therapeutic indication (Miura et al, Int J Hematology (2016)103(2): 122-.

Umbilical cord-derived and endodontic-derived MSCs

Huang et al (J.Dent.Res. (2009)88(9):792-806) discusses MSCs from dental pulp and compares their characteristics to MSCs from other sources. Carvalho et al (Curr Stem Cell Res Ther. (2011)6(3): 221-.

ASCs

Adipose-derived mscs (ascs) are usually isolated from subcutaneous adipose tissue, which makes them available in large quantities. ASCs proliferate rapidly with high cellular activity, making them ideal sources for obtaining MSCs.

By "adipose tissue" is meant any adipose tissue. The adipose tissue may be brown or white adipose tissue, derived from subcutaneous, omentum/visceral, mammary, gonadal or other adipose tissue sites. Typically, the adipose tissue is subcutaneous white adipose tissue. Such cells may comprise a primary cell culture or an immortalized cell line. The adipose tissue may be from any organism having adipose tissue. Typically, the adipose tissue is mammalian, most typically, the adipose tissue is human. A convenient source of adipose tissue is from liposuction surgery, however, the source of adipose tissue or the method of isolation of adipose tissue is not critical to the present invention.

The population of stem cells may be a population of ASCs generated using any of the methods described in example 1 or described herein.

Preferred ASCs are the product "Darvadstrocel" (trade name ") Human allogenic adipose-derived stem cells (human eASCs). These expanded ASCs express the cell surface markers CD29, CD73, CD90, and CD 105. These cells are capable of expressing a variety of factors, such as Vascular Endothelial Growth Factor (VEGF), transforming growth factor-beta 1 (TGF-beta 1), interleukin 6(IL-6), matrix metalloproteinase inhibitor-1 (TIMP-1), and interferon-gamma(IFN-. gamma.) and inducible indoleamine 2, 3-dioxygenase (IDO). Thus, the population of ASCs may be characterized by at least about 50%, at least about 60%; at least about 70%; at least about 80%; at least about 85%; at least about 90% or at least about 95% or more express one or more of CD29, CD73, CD90, and/or CD 105. The population of ASCs may be characterized by at least about 50%, at least about 60%; at least about 70%; at least about 80%; at least about 85%; at least about 90% or at least about 95% of the cell population expresses all of CD29, CD73, CD90, and CD 105. In general, a population of ASCs may be characterized by at least about 80% of the cell population expressing all of CD29, CD73, CD90, and CD 105.

According to Bourin et al (Cytotherapy (2013)15(6):641-648), the population of ASCs can be defined as positive for CD13, CD29, CD44, CD73, CD90 and CD105 expression and negative for CD31 and CD45 expression. In the population of ASCs, at least about 50%, at least about 60%; at least about 70%; at least about 80%; at least about 85%; at least about 90% or at least about 95% of the cell population may express CD13, CD29, CD44, CD73, CD90, and CD105, and less than about 5%, about 4%, about 3%, or about 2% of the population of ASCs may express CD31 and CD 45. Typically, of the population of ASCs, at least about 80% of the population of cells may express CD13, CD29, CD44, CD73, CD90, and CD105, and less than about 5% of the population of ASCs may express CD31 and CD 45.

ASCs may adhere to plastic under standard culture conditions.

The expanded asc (eas) exhibits a fibroblast-like morphology in culture. In particular, these cells are large and morphologically characterized by shallow cell bodies with few long and thin cell processes. The nucleus is large and round, and the nucleolus is prominent, so that the appearance of the nucleus is clear. Most of the eASCS show this spindle-shaped morphology, but usually some cells acquire polygonal morphology (Zuk et al tissue Eng (2001)7(2): 211-228).

ASCs may be positive for surface markers HLA I, CD29, CD44, CD59, CD73, CD90 and CD 105. In some embodiments, the population of ASCs may be characterized by a population of ASCs that is at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%; at least about 90% or at least about 95% of the ASCs population express the surface markers HLA I, CD29, CD44, CD59, CD73, CD90, and CD 105. Typically, at least about 80% of the etascs express surface markers HLA I, CD29, CD44, CD59, CD73, CD90, and CD 105.

ASCs may be negative for the surface markers HLAII, CD11b, CD11c, CD14, CD45, CD31, CD80, and CD 86. In some embodiments, the population of ASCs may be characterized by less than about 5% of the population of ASCs expressing the surface markers HLAII, CD11b, CD11c, CD14, CD45, CD31, CD80, and CD 86. More typically, less than about 4%, 3%, or 2% of the population of ASCs express the surface markers HLAII, CD11b, CD11c, CD14, CD45, CD31, CD80, and CD 86. In one embodiment, less than about 1% of the population of ASCs express the surface markers HLAII, CD11b, CD11c, CD14, CD45, CD31, CD80, and CD 86.

In some cases, in a population of ASCs, at least about 80% of the cell population expresses all of CD29, CD73, CD90, and CD105, and less than about 5% of the population of ASCs expresses surface markers HLAII, CD11b, CD11c, CD14, CD45, CD31, CD80, and CD 86.

In some embodiments, the population of ASCs may express one or more (e.g., two or more, three or more, four or more, five or more, six or seven) of HLA I, CD29, CD44, CD59, CD73, CD90 and CD 105. In some embodiments, the esascs do not express one or more (e.g., two or more, three or more, four or more, five or more, six or more, seven or eight) of HLAII, CD11b, CD11c, CD14, CD45, CD31, CD 80. In some embodiments, the esascs express four or more of HLA I, CD29, CD44, CD59, CD73, CD90, and CD105, and do not express four or more of HLAII, CD11b, CD11c, CD14, CD45, CD31, CD 80.

Expression of CD34 may be negative or lower, e.g., expressed by 0 to about 30% of the ASCs population. Thus, in some cases, ASCs as described above may express CD34 at low levels, e.g., in about 5% to about 30% of the population. Alternatively, in other cases, the described ASCs do not express CD34, e.g., less than about 5% of the population of ASCs express CD 34.

In some embodiments, a population of ASCs (e.g., at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 85%; at least about 90%, or at least about 95% of a population of cells) can express one or more (e.g., two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more (e.g., up to 13)) of the markers CD9, CD10, CD13, CD29, CD44, CD49A, CD51, CD54, CD55, CD58, CD59, CD90, and CD 105. For example, ASCs may express one or more (e.g., two, three, or all) of the markers CD29, CD59, CD90, and CD105, such as CD59 and/or CD 90.

In some embodiments, the population of ASCs may not express one or more (e.g., two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, or ten or more (e.g., up to 15)) of the markers factor VIII, alpha-actin, desmin, S-100, keratin, CD11b, CD11c, CD14, CD45, HLAII, CD31, CD45, STRO-1, and CD133, e.g., the ASCs do not express one or more (e.g., two, three, or all) of the markers CD45, CD31, and CD14, e.g., CD31 and/or CD 45.

In certain embodiments, ascs as described above (i) do not express markers specific for Antigen Presenting Cells (APCs); (ii) does not constitutively express IDO; and/or (iii) does not significantly constitutively express MHC II. Expression of IDO or MHC II can be induced by stimulation with IFN- γ in general.

In certain embodiments, ASCs as described above do not express Oct 4.

Method for preparing ASCs population

Methods for isolating and culturing ASCs to provide the esascs and stem cell populations of the invention, as well as compositions comprising populations of the stem cell populations of the invention, are known in the art. ASCs are typically prepared from stromal parts of adipose tissue and are selected by adhesion to a suitable surface, such as plastic. Thus, the methods of stem cell cryopreservation disclosed herein may include the following initial steps (prior to step (a) of either method): (i) isolating a population of ASCs from a stromal fraction of adipose tissue obtained from the patient, and (ii) culturing the population of ASCs. The ASCs may optionally be selected in step (i) to adhere to a suitable surface, for example a plastic. Optionally, the phenotype of the ASCs may be assessed during and/or after the culturing step (ii).

ASCs may be obtained by any means standard in the art. The cells are typically obtained by separating the cells from the source tissue (e.g., lipoaspirate or adipose tissue), typically by treating the tissue with a digestive enzyme such as collagenase. The digested tissue material is then typically filtered through a filter of about 20 microns to 1 mm. The cells are then isolated (typically by centrifugation) and cultured on an adherent surface (typically a tissue culture plate or flask). Such methods are known in the art and are disclosed, for example, in U.S. patent No. 6777231. According to this methodology, lipoaspirate is obtained from adipose tissue and cells derived therefrom. During this methodology, the cells may be washed to remove contaminating debris and red blood cells, preferably with PBS. Cells were digested with collagenase in PBS (e.g., 0.075% collagenase for 30 min at 37 ℃; type I, Invitrogen, Carlsbad, Calif.). To eliminate the remaining red blood cells, the digested sample can be washed (e.g., with 10% fetal calf serum), washed with 160mmol/L NH4Cl and finally suspended in DMEM complete medium (DMEM with 10% FBS, 2mmol/L glutamine and 1% penicillin/streptomycin). Cells can be filtered through a 40 μm nylon mesh.

Cultured human ASCs according to certain embodiments of the invention are described in Delarosa et al (Tissue Eng Part A (2009)15(10): 2795-. In one embodiment (as described in Lopez-Santalla et al 2015), human adipose tissue aspirates from healthy donors were washed twice with phosphate buffered saline and digested with 0.075% collagenase (type I; Invitrogen). The digested sample was used as a 10% embryoBovine Serum (FBS) wash with 160mM NH4The remaining erythrocytes were eliminated by Cl treatment and suspended in a medium (Du's modified Eagle Medium (DMEM) containing 10% FBS). Subjecting cells (2-3. multidot.10)4Individual cell/cm2) Inoculated in a tissue culture flask and cultured (37 ℃, 5% CO)2) The medium was changed every 3-4 days. When the cells reached 90% confluence, the cells were transferred to a new flask (10)3Individual cell/cm2). Cells were expanded to up to 12-14 replications and frozen. Experiments were performed with cells from two male and two female adult donors at 12-14 population doublings. ASCs from the same freezer were thawed and inoculated prior to each experiment. ASCs are defined according to the international society for cell therapy standards: positive for HLA-I, CD73, CD90 and CD105, and negative for CD11b, CD14, CD31, CD34 and CD 45.

In another embodiment (as described by Delarosa et al 2009), lipoaspirate obtained from human adipose tissue from healthy adult donors was washed twice with PBS and digested for 30 minutes at 37 ℃ for 18U/mL type I collagenase in PBS. One unit of collagenase releases 1mM L-leucine equivalent from collagen within 5 hours at 37 deg.C, pH 7.5(Invitrogen, Carlsbad, Calif.). Digested samples were washed with 10% Fetal Bovine Serum (FBS) and 160mM NH4Treated with Cl, suspended in culture medium (DMEM containing 10% FBS) and filtered through a 40-mm nylon mesh. Cells (2-3X 10)4Individual cell/cm2) Inoculating onto tissue culture flasks and culturing at 37 deg.C and 5% CO2Amplification was performed under conditions in which the medium was changed every 7 days. When the culture reached 90% confluence, the cells were transferred to a new flask. The phenotype of cells is characterized by their ability to differentiate into cartilage, bone and fat genetic lineages. In addition, hASCs were verified by staining with specific surface markers. hASCs are positive for HLA-I, CD90 and CD105, and negative for HLA-II, CD40, CD80, CD86, and CD 34. The study used a collection of six healthy donors (three men and three women, between the ages of 35-47). Cells from passage 4-6 were used.

Culturing the ASCs in a suitable tissue culture vessel, including suitable ASCss a surface to which it adheres, for example plastic. Non-adherent cells are removed, e.g., by washing in a suitable buffer, to provide an isolated adherent stromal cell population (e.g., ASC). Cells isolated in this manner can be seeded (preferably 2-3X 10)4Individual cell/cm2) Onto tissue culture flasks and incubated at 37 ℃ and 5% CO2And (5) carrying out amplification, and replacing the culture medium every 3-4 days. When the culture reaches about 90% confluence, the cells are preferably detached from the adherent surface (e.g., by means of trypsin) and transferred ("passaged") to a new culture flask (1,000 cells/cm)2)。

ASCs may be cultured for at least about 15 days, at least about 20 days, at least about 25 days, or at least about 30 days. Typically, expansion of the cells in culture increases the homogeneity of the cell phenotype in the population, thereby obtaining a substantially pure population.

In some embodiments, the ASCs are expanded in culture for at least three culture generations or "passaged at least 3 times". In other embodiments, the cells are passaged at least four times, at least five times, at least six times, at least seven times, at least eight times, at least nine times, or at least ten times. Preferably, the cells are passaged more than 3 times to improve homogeneity of the cell phenotype in the cell population. In fact, cells can expand indefinitely in culture as long as the homogeneity of the cell phenotype is improved and the differentiation capacity is maintained.

In some embodiments, the ASCs are propagated in culture for at least three population doublings, e.g., the cells are expanded in culture for at least four, five, six, seven, eight, nine, ten, 15, or 20 population doublings. In some embodiments, the cells are expanded in culture for less than seven, eight, nine, ten, 15, or 20 population doublings. In certain embodiments, the cells are expanded in culture for about 5-10 population doublings. In certain embodiments, the cells are expanded in culture for about 10-15 population doublings. In certain embodiments, the cells are expanded in culture for about 15-20 population doublings, e.g., about 16 population doublings.

The ASC isolation is preferably performed under sterile or GMP conditions.

The stem cell population (e.g., ASCs) may be allogeneic, i.e., not isolated from the subject to which the stem cell population is to be administered as a treatment.

Stem cell populations

Pre-treatment with NAC, post-thaw treatment with NAC, or a combination of pre-treatment with NAC and post-thaw treatment according to the methods disclosed herein can result in one or more, two or more, three or more, or all four of the following characteristics: increased viable cell number, increased growth rate, increased mitochondrial activity and increased recovery rate compared to control stem cell populations. A control stem cell population is the same stem cell population that has not been pre-treated with NAC, post-NAC thaw treatment, or both pre-treated with NAC and post-NAC thaw treatment, but that has otherwise been subjected to the same conditions. In another embodiment, the control stem cell population is derived from the same stem cell population as the stem cell population that was pre-treated with NAC, post-NAC-thaw treatment, or both pre-treated with NAC and post-NAC-thaw treatment, but the control population is not pre-treated with NAC, post-NAC-thaw treatment, or both pre-treated with NAC and post-NAC-thaw treatment, but otherwise experiences the same conditions.

Also provided are populations of stem cells (e.g., ASCs) having one or more, two or more, three or more, or all four of these properties obtained by any one of the methods described herein.

The number of viable cells can be increased for the stem cell population after thawing and optionally culturing for about 1 day, about 2 days, about 3 days, about 4 days, about 7 days, or about 10 or more days, as compared to a control cell population. For example, the number of viable cells in the stem cell population can be increased at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold or more after thawing and culturing for 1 day (and/or 4 days) compared to a control stem cell population. For example, FIG. 4A shows 1 day of culture (. about.5,000 vs. 3,000 cells/cm) versus untreated cells2) And cultured for 4 days (12,500 vs 9,000 cells/cm)2) Thereafter, the number of viable cells of ASCs pretreated with 6mM NAC increased. In another example, FIG. 6 shows that relative to untreated cells,culturing for 7 days (6,300 vs-5,600 cells/cm)2) 11 days (-18,700 vs-17,500 cells/cm)2) And 14 days (. about.18,300 vs. 15,200 cells/cm)2) Post-thaw treatment with 2mM NAC increased the number of viable cells. Suitable methods for measuring the number of viable cells are described above.

Growth rate of the stem cell population (i.e., number of viable cells per cm per day) compared to the control stem cell population2Increase in) may be increased. The growth rate of the stem cell population can be increased at least about 1.03-fold, about 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, or at least about 2-fold or more after thawing (e.g., between days 1 and 4 of culture after thawing) compared to a control stem cell population. For example, fig. 4A shows the increase in growth rate from day 1 to day 4 in ASCs cultured with 6mM NAC pre-treated relative to untreated cells. Specifically, cells pretreated with NAC had a growth of about 2500 cells/cm from day 1 to day 42Day, compared to about 2000 cells/cm for untreated cells2A day, i.e., an approximately 1.25-fold increase. In other examples, fig. 6 shows that the growth rate of ASCs treated after thawing with 2mM NAC increases from day 7 to day 11 relative to untreated cells, i.e., about 3100 cells/cm2Day, compared to about 3000 cells/cm for untreated cells2The day is.

The mitochondrial activity (as measured, for example, by an MTS assay) of a stem cell population of cells can be increased after thawing and optionally culturing for about 1 day, about 2 days, about 3 days, about 4 days, about 7 days, or about 10 or more days, as compared to a control stem cell population. Mitochondrial activity may be increased by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% or more in a stem cell population that is thawed and cultured for 1 day (and/or 4 days) compared to a control stem cell population. For example, figure 4B shows that mitochondrial activity increased by more than 35% following pretreatment with 6mM NAC compared to untreated cells, as measured after 24 hours of culture after thawing (MTS assay reading at 490nm normalized to 100% for control). In another example, fig. 4C shows an increase in mitochondrial activity of more than 15% after pretreatment with 6mM NAC compared to untreated cells, as measured after 96 hours of culture after thawing.

For adherent cells (e.g., ASCs), post-thaw "recovery" can be defined as the point at which the number of viable cells of the adherent cells increases above the initial seeding density during culture. For cells grown in suspension, "recovery" after thawing can be defined as when the number of viable cells increases during culture beyond the initial seeding density. The recovery rate of the thawed stem cell population, i.e., the time it takes for the cells to recover after thawing, may be increased (i.e., decreased) compared to the control stem cell population. For example, the number of hours spent to recover after thawing may be reduced by at least about 1.1 fold, at least about 1.2 fold, at least about 1.4 fold, at least about 1.6 fold, at least about 2 fold, at least 3 fold, at least 4 fold, or at least 5 fold or more compared to a control stem cell population. For example, fig. 4A shows that ASCs pretreated with 6mM NAC recovered after 1 day of post-thaw culture, while untreated cells did not recover.

In a preferred method or stem cell population as disclosed herein, the stem cell population has one or more, two or more, three or more, four or more or all five of the following characteristics: (a) at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold or more increase in the number of viable cells after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population; (b) the growth rate of the thawed stem cell population (e.g., day 1 to day 4 of post-thaw culture) is increased by at least about 1.03-fold, about 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, or at least about 2-fold or more compared to the control stem cell population; (c) at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% increase in mitochondrial activity after thawing and optionally culturing for about 1 day and/or about 4 days, as compared to a control stem cell population; (d) a reduction in time taken for cell recovery after thawing compared to a control stem cell population; and/or (e) at least about 1.1-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.6-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, or at least about 5-fold decrease in the number of hours it takes for cells to recover after thawing, relative to a control stem cell population

In a preferred method or stem cell population as disclosed herein, the population of ASCs has one or more, two or more, three or more, four or more, five or more, or all six of the following characteristics: (1) at least about a 1.5-fold increase in the number of viable cells after thawing and culturing for about 1 day compared to a control stem cell population (e.g., after 24 hours of pretreatment with 6mM NAC); (2) the number of viable cells after thawing and culturing for about 4 days is at least about 1.3-fold compared to the control stem cell population (e.g., after 24 hours of pretreatment with 6mM NAC); (3) the growth rate of the post-thaw culture from day 1 to day 4 is increased by at least about 1.25-fold compared to a control stem cell population (e.g., after 24 hours of pretreatment with 6mM NAC); (4) an increase in mitochondrial activity of at least about 35% after thawing and culturing for about 1 day compared to a control stem cell population (e.g., after 24 hours of pretreatment with 6mM NAC); (5) an increase in mitochondrial activity of at least about 15% after thawing and culturing for about 4 days compared to a control stem cell population (e.g., after 24 hours of pretreatment with 6mM NAC); and/or (6) a reduction in the time taken for ASCs to recover following thawing (e.g., after 24 hours of pretreatment with 6mM NAC) compared to a control stem cell population.

In a preferred method or stem cell population as described herein, the population of ASCs has one or more, two or more, three or more, or all four of the following characteristics: (a) the number of viable ASCs increases by at least about 1.1-fold after 7 days of treatment following thawing with NAC (e.g., 2mM) compared to a control stem cell population; (b) the number of viable ASCs increases by at least about 1.05-fold after 11 days of treatment following thawing with NAC (e.g., 2mM) compared to a control stem cell population; (c) the number of viable ASCs increases by at least about 1.2-fold after 14 days of treatment following thawing with NAC (e.g., 2mM) compared to a control stem cell population; and/or (d) at least about a 1.03-fold increase in growth rate after treatment with NAC (e.g., 2mM) thawing, as measured on days 7 to 11 in culture, as compared to a control stem cell population.

Low temperature storage composition

Disclosed are cryopreservation compositions comprising stem population cells (e.g., ASCs) prepared by any of the methods disclosed herein and cryopreservation media. The cryopreservation composition can be frozen. The cryopreservation composition can contain NAC, e.g., in a concentration range of about 0.5-10mM, e.g., about 2-8mM or about 4-6 mM. In a particularly preferred embodiment, the concentration of NAC in the cryopreservation composition is about 6 mM.

In practicing the methods of the invention, it is envisioned that cryopreservation processes may have an impact on a variety of cellular processes. As discussed above, the freezing process may stop intracellular reactions, including gene transcription. These effects may also arise from or be in addition to the chemical composition of the cryopreservation medium (e.g. metabolism of the cryoprotectant, ion concentration) or pretreatment of the cells with NAC. Furthermore, in cryopreservation, freezing-induced stresses can affect cell transport processes involving heat shock or membrane destabilization of proteins.

Pharmaceutical composition

Disclosed are pharmaceutical compositions comprising a population of stem cells (e.g., ASCs) prepared by any one of the methods disclosed herein and a pharmaceutically acceptable carrier.

The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

Examples of pharmaceutically acceptable carriers include pharmaceutically acceptable substances, compositions or vehicles, such as liquid or solid fillers, diluents, excipients or solvent encapsulating substances, involved in carrying or transporting the subject compound from one organ, or part of the body, to another organ, or part of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.

The pharmaceutical composition may be sterile, free of unwanted viruses, bacteria and other pathogens, and free of pyrogens. That is, for human administration, the subject compositions should comply with sterility, pyrogenicity, and general safety and purity standards as required by FDA Office of Biologics (FDA Office of Biologics) standards.

The stem cell populations disclosed herein may be obtained from allogeneic sources due to the difficulty in obtaining sufficient autologous stem cells. It is known in the art that bone marrow-derived MSCs and ASCs do not elicit an allogeneic lymphocyte response in vitro, and therefore, these cells can be used in any patient regardless of MHC incompatibility. Thus, the population of stem cells (e.g., bone marrow-derived MSCs or ASCs) in the pharmaceutical composition may be allogeneic with respect to the intended transplant host.

The pharmaceutical composition may comprise a suspension of the stem cell population in various solutions or substances, for example for use as a medicament or biomaterial, as described in more detail below. The pharmaceutical composition may comprise suspended cell stem cells (e.g., allogeneic ASCs) in ringer's solution and HSA. The pharmaceutical composition may comprise a suspension of stem cells (e.g., allogeneic ASCs) in sterile buffered saline solution. The cells may be provided in a single use vial without preservatives. Cells can be administered at a dose of 1.2 million cells (e.g., at a concentration of 500 million cells/mL). Cells (e.g., ASCs) may also be administered at about 100 to 1000 ten thousand cells/kg.

In certain embodiments, the pharmaceutical composition is a suspension of stem cells (e.g., allogeneic ASCs) in a substance such as a polymer, a gel, or the like. Such suspensions may be prepared, for example, by precipitating the stem cells from the culture medium and resuspending them in the desired solution or substance. The cells may be pelleted and/or replaced from the culture medium, for example by centrifugation, filtration, ultrafiltration, and the like.

The concentration of the subject adipose tissue-derived stromal stem cells in the composition comprising the subject adipose tissue-derived stromal stem cells can be at least about 500, at least about 1000, at least about 2000, at least about 3000, or at least about 4000 million cells/mL. Typically, a concentration of about 100 to 1000 ten thousand cells/mL, for example about 500 to 1000 ten thousand cells/mL. In certain embodiments, the cell density in the pharmaceutical composition is about 500 ten thousand cells/mL.

In certain embodiments, the pharmaceutical composition comprises about 1000 to 1.5 million cells, preferably about 3000 million cells or about 1.2 million cells.

In some cases, the pharmaceutical composition may comprise NAC. In other cases, the pharmaceutical composition may not comprise NAC.

Pharmaceutically acceptable carriers and diluents include saline, buffered aqueous solutions, solvents and/or dispersion media. The use of such carriers and diluents is well known in the art. The solution is generally sterile and fluid to the extent that ease of injection occurs. Generally, the solutions are stable under the conditions of manufacture and storage and are protected from the contaminating action of microorganisms such as bacteria and fungi by the use of, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. The pharmaceutical compositions may be prepared by suspending a population of stem cells (e.g., ASCs) as described herein in a pharmaceutically acceptable carrier or diluent, and as necessary, the other ingredients listed above, followed by filter sterilization.

Some examples of materials and solutions that can be used as pharmaceutically acceptable carriers include: (1) sugars such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) tragacanth powder; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) a ringer's solution; (19) ethanol; (20) a pH buffer solution; (21) polyesters, polycarbonates and/or polyanhydrides; and (22) other non-toxic compatible materials commonly used in pharmaceutical formulations.

In certain embodiments, the pharmaceutical composition further comprises a binder. The adhesive may be a fibrin-based adhesive, such as fibrin gel or fibrin glue or fibrin-based polymers or adhesives, or other tissue adhesives or surgical glues, such as, for example, cyanoacrylate, collagen, thrombin and polyethylene glycol. Other substances that may be used include, but are not limited to, calcium alginate, agarose, type I, II, IV or other collagen isoforms, polylactic/polyglycolic acid, hyaluronic acid derivatives or other substances (Perka et al J.biomed.Mater. Res. (2000)49: 305-311; Securiest et al J.biomed.Mater. Res. (2000)49: 534-541; Chuu et al J.biomed.Mater. Res. (1995)29: 1147-1154; Hendrickson et al ortho p.Res. (1994)12: 485-497). In other embodiments, the adhesive is a liquid bandage in which a population of stem cells (e.g., ASCs) is mixed with a liquid bandage substance. A "liquid bandage" is a solution containing a compound, such as a polymeric material, which is applied to a wound with a spray or brush, and then the solvent is removed by evaporation to provide a protective film on the wound.

The pharmaceutical composition may also be used to coat a support, such as a medical device. For example, the support may be a suture or thread. The support may be coated with cells in any manner known to those skilled in the art, such as by soaking, spraying, painting, stamping, and the like. In one embodiment, the support is a suture, staple, absorbable thread, non-absorbable thread, natural thread, synthetic thread, monofilament thread, or multifilament thread (also known as a braid). A preferred method of preparing sutures and other supports for closing wounds coated with adipose-derived stromal stem cells is disclosed in U.S. patent application No. 11/056,241, "biomaterials for suturin," filed on 14/2/2005, which is incorporated herein by reference in its entirety. The pharmaceutical compositions disclosed herein represent novel compositions that can be used with the methods disclosed in U.S. patent application No. 11/056,241.

In addition, in any of the disclosed pharmaceutical compositions, at least one therapeutic agent can be incorporated into the composition (although not required, and can optionally be excluded). For example, the pharmaceutical composition may contain an analgesic (e.g., to help treat inflammation or pain), or an anti-infective agent to prevent infection at the site treated with the composition.

More specifically, non-limiting examples of useful therapeutic agents that may be included in the pharmaceutical compositions described herein include the following therapeutic classes: analgesics, such as non-steroidal anti-inflammatory drugs, opioid agonists, and salicylates; anti-infective agents such as anthelmintics, anti-anaerobics, antibiotics, aminoglycoside antibiotics, antifungal antibiotics, cephalosporin antibiotics, macrolide antibiotics, promiscuous β -lactam antibiotics, penicillin antibiotics, quinolone antibiotics, sulfonamide antibiotics, tetracycline antibiotics, antimycobacterial agents, antituberculotic agents, antiprotozoal agents, antimalarial antiprotozoal agents, antiviral agents, antiretroviral agents, antimycotic agents, antiinflammatory agents, corticosteroid antiinflammatory agents, antipruritic/local anesthetics, local anti-infective agents, antifungal local anti-infective agents, antiviral local anti-infective agents; electrolyte and renal agents such as acidifiers, alkalizers, diuretics, carbonic anhydrase inhibitor diuretics, loop diuretics, osmotic diuretics, potassium sparing diuretics, thiazide diuretics, electrolyte substitutes, and uricosuric agents; enzymes, such as pancreatin and thrombolytic enzyme; gastrointestinal agents such as antidiarrheals, antiemetics, gastrointestinal anti-inflammatory agents, salicylate gastrointestinal anti-inflammatory agents, antacid anti-ulcer agents, gastric acid pump inhibitor anti-ulcer agents, gastric mucosa anti-ulcer agents, H2 blocker anti-ulcer agents, gallstone dissolving agents, digestive agents, emetic agents, laxatives and stool softeners, and prokinetic agents; general anesthetics, such as inhalation anesthetics, halogenated inhalation anesthetics, intravenous anesthetics, barbiturate intravenous anesthetics, benzodiazepine intravenous anesthetics, and opioid agonist intravenous anesthetics; hormones and hormone modulators, such as abortifacients, adrenal agents, corticosteroid adrenal agents, androgens, antiandrogens, immunobiological agents, such as immunoglobulins, immunosuppressants, toxoids, and vaccines; local anesthetics, such as amide local anesthetics and ester local anesthetics; musculoskeletal agents, such as anti-gout anti-inflammatory agents, corticosteroid anti-inflammatory agents, gold compound anti-inflammatory agents, immunosuppressive anti-inflammatory agents, non-steroidal anti-inflammatory drugs (NSAIDs), salicylate anti-inflammatory agents, minerals; and vitamins such as vitamin a, vitamin B, vitamin C, vitamin D, vitamin E and vitamin K.

Preferred classes of useful therapeutic agents from the above classes include: (1) general analgesics such as lidocaine or a derivative thereof, and non-steroidal anti-inflammatory drugs (NSAIDs) analgesics including diclofenac, ibuprofen, ketoprofen, and naproxen; (2) opioid agonist analgesics such as codeine, fentanyl, hydromorphone, and morphine; (3) salicylate analgesics, such as aspirin (ASA) (enteric ASA); (4) h1 blocker antihistamines such as clemastine and terfenadine; (5) anti-infective agents, such as mupirocin; (6) anti-anaerobic anti-infective agents such as chloramphenicol and clindamycin; (7) antifungal antibiotic anti-infectives, such as amphotericin b, clotrimazole, fluconazole, and ketoconazole; (8) macrolide antibiotic antiinfectives, such as azithromycin and erythromycin; (9) miscellaneous beta-lactam antibiotic anti-infectives, such as aztreonam and imipenem; (10) penicillin antibiotic anti-infectives, such as nafcillin, oxacillin, penicillin G and penicillin V; (11) quinolone antibiotic anti-infectives, such as ciprofloxacin and norfloxacin; (12) tetracycline antibiotic anti-infectives, such as doxycycline, minocycline, and tetracycline; (13) antituberculous anti-mycobacterial anti-infectives such as Isoniazid (INH) and rifampicin; (14) antiprotozoal anti-infectives, such as atovaquone and dapsone; (15) anti-malarial antiprotozoal anti-infective agents such as chloroquine and pyrimethamine; (16) antiretroviral anti-infectives, such as ritonavir and zidovudine; (17) antiviral anti-infective agents such as acyclovir, ganciclovir, interferon alpha and rimantadine; (18) antifungal topical anti-infective agents, such as amphotericin B, clotrimazole, miconazole, and nystatin; (19) antiviral topical anti-infective agents, such as acyclovir; (20) electrolytes and renal agents, such as lactulose; (21) loop diuretics, such as furosemide; (22) potassium sparing diuretics, such as triamterene; (23) thiazide diuretics, such as Hydrochlorothiazide (HCTZ); (24) uricosuric agents, such as probenecid; (25) enzymes such as rnases and dnases; (26)antiemetics, such as prochlorperazine; (27) salicylate gastrointestinal anti-inflammatory agents, such as sulfasalazine; (28) gastric acid pump inhibitors antiulcer agents, such as omeprazole; (29) h2 blockers antiulcer agents such as cimetidine, famotidine, nizatidine and ranitidine; (30) digestive agents, such as pancreatic lipase; (31) prokinetic agents, such as erythromycin; (32) ester local anesthetics, such as benzocaine and procaine; (33) musculoskeletal corticosteroid anti-inflammatory agents, such as beclomethasone, betamethasone, cortisone, dexamethasone, hydrocortisone, and prednisone; (34) musculoskeletal anti-inflammatory immunosuppressants such as azathioprine, cyclophosphamide and methotrexate; (35) musculoskeletal non-steroidal anti-inflammatory drugs (NSAIDs), such as diclofenac, ibuprofen, ketoprofen, ketobutyric acid, and naproxen; (36) minerals such as iron, calcium and magnesium; (37) vitamin B compounds, e.g. cyanocobalamin (vitamin B)12) And nicotinic acid (vitamin B)3) (ii) a (38) Vitamin C compounds, such as ascorbic acid; and (39) vitamin D compounds, such as calcitriol.

In certain embodiments, the therapeutic agent may be a growth factor or other molecule that affects cell differentiation and/or proliferation. Growth factors that induce a terminally differentiated state are well known in the art and may be selected from any such factor that has been shown to induce a terminally differentiated state. In certain embodiments, the growth factor used in the methods described herein can be a functional variant or fragment of a naturally occurring growth factor. For example, variants can be generated by making conservative amino acid changes and testing the resulting variants using assays known in the art to test growth factor function.

Use and application

Use of NAC

The use of NAC for cryopreservation of stem cells is disclosed, e.g., in any of the methods disclosed herein.

Medical applications

Stem cells are being used to treat an increasing number of diseases and disorders. Thus, a population of stem cells made according to any one of the methods disclosed herein, a pharmaceutical composition disclosed herein, or a cryopreservation composition disclosed herein can be used for treatment. The term "treatment" is intended to encompass the treatment and/or prevention of a disease, disorder, or symptom in a patient. The terms "subject", "recipient" and "patient" are used interchangeably herein and refer to any human or non-human animal (e.g., a mammal) in need of treatment unless specifically stated otherwise. In a preferred embodiment, the patient is a human. When the patient is a human, the stem cell population is typically human.

Disclosed is a stem cell population, a pharmaceutical composition, or a cryopreservation composition as described herein for use in a method of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof. The cell population used in the method can be prepared by any of the methods disclosed herein for cryopreservation of stem cells.

Also disclosed is the use of a stem cell population, a pharmaceutical composition or a cryopreservation composition as described herein for the preparation of a medicament for treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g. type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof.

Also disclosed are methods of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection, the method comprising administering to a subject in need thereof a stem cell population, pharmaceutical composition, or cryopreservation composition disclosed herein.

The stem cell populations, pharmaceutical compositions, or cryopreservation compositions described herein, particularly when the stem cell populations are ASCs, may be used to treat fistulas. The term "fistula" refers to any abnormal passage or communication or connection, typically between two internal organs or from an internal organ to a body surface, such as a connection or passage between organs or blood vessels that are not normally in communication. For example, the types of fistulas, named after the body regions in which they occur, include anorectal or anal or fecal fistulas (between the rectum or other anorectal region and the skin surface), arteriovenous or AV fistulas (between the arteries and veins), biliary fistulas (between the bile duct to the skin surface, typically caused by gallbladder surgery), cervical fistulas (abnormal opening of the cervix), cranial sinus fistulas (between the intracranial space and the paranasal sinus), enterointestinal fistulas (between two parts of the intestine), enterocutaneous fistulas (between the intestine and the skin surface, i.e., from the duodenum or jejunum or ileum), enterovaginal fistulas (between the intestine and the vagina), gastric fistulas (between the stomach and the skin surface), uteroperitoneal fistulas (between the uterus and the abdominal cavity), perilymph fistulas (between the membranes between the middle and inner ear), pulmonary arteriovenous fistulas (between the pulmonary artery and veins, resulting in a shunt of blood flow), Rectovaginal fistula (between the rectum and the vagina), umbilical fistula (between the umbilicus and the intestine), tracheoesophageal fistula (between the respiratory tube and the feeding tube), and vesicovaginal fistula (between the bladder and the vagina). Causes of fistulas include trauma, complications from medical treatment and disease. Inflammatory bowel diseases, such as crohn's disease and ulcerative colitis, are the major cause of anorectal, enterointestinal and enterocutaneous fistulas. In certain embodiments, the fistula is a perianal fistula, such as a refractory complex perianal fistula in a crohn's disease patient. For intralesional injection, a population of stem cells (e.g., allogeneic ASCs) can be administered at a dose of about 1.2 million cells (e.g., about 500 ten thousand cells/mL).

Disclosed is the use of a stem cell population as disclosed herein in a method for treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; (c) thawing the frozen stem cell population to obtain a thawed stem cell population; (d) optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and (e) administering the population of stem cells to the patient.

Also disclosed is the use of a population of stem cells disclosed herein for the manufacture of a medicament for treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) administering NAC stem cell population to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; (c) thawing the frozen stem cell population to obtain a thawed stem cell population; (d) optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and (e) administering the population of stem cells to the patient.

Also disclosed are methods of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of: (a) treating a stem cell population with NAC to obtain a treated stem cell population; (b) freezing the treated stem cell population to obtain a frozen stem cell population; (c) thawing the frozen stem cell population to obtain a thawed stem cell population; (d) optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and (e) administering the population of stem cells to the patient.

In certain embodiments, the methods of treatment and/or prevention further comprise any of the steps (e.g., "pre-treatment") as defined in the methods disclosed herein prior to administering the population of stem cells to the patient.

Disclosed is the use of a population of stem cells as described herein in a method of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and (d) administering the population of stem cells to the patient.

Also disclosed is the use of a population of stem cells as described herein for the manufacture of a medicament for treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and (d) administering the population of stem cells to the patient.

Also disclosed are methods of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of: (a) freezing a stem cell population to obtain a frozen stem cell population; (b) thawing the frozen stem cell population to obtain a thawed stem cell population; (c) culturing the thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and (d) administering the population of stem cells to the patient.

In certain embodiments, the methods of treatment and/or prevention further comprise any of the steps (e.g., "post-thaw treatment") as defined in the methods disclosed herein prior to administering the population of stem cells to the patient.

The stem cell population, pharmaceutical composition, or cryopreservation composition may be administered at a dose of about 100 million to 1.5 million stem cells (e.g., allogeneic ASCs). In preferred embodiments, the stem cells (e.g., allogeneic ASCs) may be administered at a dose of about 3000 million or about 1.2 million cells.

Administration of the stem cell populations, pharmaceutical compositions, or cryopreservation compositions disclosed herein to a subject, particularly a human subject, can be performed by injecting or implanting cells into a target site in the subject. For example, a delivery device that is easily introduced into a subject by injection or implantation may be used. Such delivery devices include tubes, such as catheters, for injection into a recipient subject. In a preferred embodiment, the tube additionally has a needle, e.g., a syringe, through which the stem cell population, pharmaceutical composition or cryopreservation composition can be introduced into the subject at a desired location.

In a preferred embodiment, the population of stem cells-including those in the pharmaceutical composition and/or cryopreservation composition-are ASCs.

The stem cells may be allogeneic or autologous.

Toxicity and therapeutic efficacy of the subject compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining LD50And ED50. Compositions exhibiting a large therapeutic index are preferred. Although compounds exhibiting toxic side effects may be used, care should be taken to design a delivery system that targets the agent to the desired site to reduce side effects.

The data obtained from cell culture assays and animal studies can be used to formulate a range of doses for use in humans. The dose of any therapeutic agent, or alternatively any component thereof, is generally within the circulating concentration range, including ED50, with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For the agents of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. The dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 determined in cell culture (i.e., the concentration of the test compound that achieves half-maximal inhibition of symptoms). Such information can be used to more accurately determine useful doses in humans. Levels in plasma can be measured, for example, by high performance liquid chromatography.

Reagent kit

Disclosed is a cryopreservation kit comprising: a frozen vial, a NAC-containing container, and a container comprising a population of stem cells. The kit may comprise instructions for use. Disclosed is a cryopreservation kit comprising: a plurality of frozen vials, a container comprising NAC, and a container comprising a population of stem cells. The population of stem cells can be provided in a kit as a composition or pharmaceutical composition as disclosed herein.

General definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

The articles "a" and "an" refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, "an element" means one element or more than one element.

The terms "comprising" and "comprising" are used in an inclusive, open sense, meaning that additional elements may be included.

In general, a method that "comprises" many steps does not require that the steps be performed in a particular order. If the method includes a plurality of sequentially numbered or alphabetically arranged steps (e.g., (1), (2), (3) or (a), (b), (c), etc.), this means that the steps must be performed in the order specified, unless otherwise specified. However, this language does not exclude the possibility of performing additional steps between each specified step.

The term "including" is used herein to mean "including but not limited to". "include" and "include but are not limited to" are used interchangeably.

Examples

The present invention will now be generally described, as will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.

Example 1 ASC isolation and culture

Human samples were obtained with informed consent (reference tissue acquisition site approved by the spanish ethics committee; ciionica de la Luz hospital, spanish madrid). As previously disclosed (A), (B)-Corvo et al, Frontiers in Immunology (2017),8,462; menta et al, Frontiers in Immunology (2014),8,462) obtained ASC. Briefly, human adipose tissue aspirates from healthy donors were washed twice with Phosphate Buffered Saline (PBS) and digested with 0.075% collagenase (type I, Invitrogen, Carlsbad, CA, USA). Digested samples were washed with 10% Fetal Bovine Serum (FBS) and 160mM NH4Cl to remove remaining erythrocytes and suspended in medium (duller's modified Eagle medium (DMEM) with 10% FBS). Cells were seeded in tissue culture flasks and expanded (37 ℃, 5% CO)2) The medium was changed every 3-4 days. When it reached 90% confluence, the cells were transferred to a new flask. Cells were expanded to replicate 12-14 times and frozen in FBS with 10% DMSO (when ASCs were frozen throughout all examples described herein, FBS with 10% DMSO was used as the freezing medium). Experiments were performed with cell banks from three male and three female adult donors, with 12-14 population doublings. Amplified ACSs (eASCs) were confirmed to meet the definition according to the criteria of the International society for cell therapy (dominci et al, Cytotherapy (2006)8(4):315- & 317), to be positive for CD 56 73(AD2) and CD90(5E10) from Becton Dickinson (Franklin Lakes, NJ, USA) and CD105(43A3) from Biolegend (San Diego, CA, USA), and to be negative for CD14(RM052), CD19(4G7), HLA-DR (L243) from Immunotech (Monrovia, CA, USA), CD34(8G12) from Becton Dickinson and CD45(J33) from Beckman Coulter (Brea, CA, USA).

Example 2 evaluation of various pretreatment steps for the number of post-thaw ASC cells ASC pretreatment

By warming the vials in a 37 ℃ water bath and adding fresh complete DMEM (DMEM/F-12 medium-GlutaMAX)TM-I, Gibco, supplemented with 100 μ g/mL penicillin/streptomycin and 10% FBS) to dilute the freezing medium containing DMSO to thaw ASCs from donor a. Cells were centrifuged at 450g for 6 min at room temperature to eliminate residual DMSO and at 20.000 cells/cm in complete DMEM2Inoculated in a T-175 flask. After thawing 24 hours, the cells were treated with the appropriate concentrations of the compounds shown in the following table for 24 hours:

600mM NAC (SIGMA) stock was prepared in Milli-Q water (Millipore). This stock solution was used for pretreatment and post-treatment by adding 50. mu.L of the stock solution directly to 5mL of the medium per well to a final concentration of 6 mM. For 2mM, only 16.7. mu.L per well was added, and in the case of 12mM, 100. mu.L of stock solution was added. DMSO was used as vehicle for sc79 and LY 294.

After the pretreatment step, the medium was removed, the cells were washed with PBS and trypsinized using trypsin-EDTA 0.25% (ThermoFisher) for 8 minutes at 37 ℃. After trypsin was inactivated with complete DMEM, cells were harvested and centrifuged, then resuspended in freezing medium (FBS with 10% DMSO) and frozen into 500,000 or 100 ten thousand cells per vial and stored in liquid nitrogen for further use. Specifically, cells are incubated in CoolThe cells were frozen in the apparatus (BioCision) at-80 ℃ for 24 hours and then transferred to a liquid nitrogen storage vessel. All experiments were performed in an incubator at 37 ℃ and 5% CO2The process is carried out as follows.

Evaluation of various pretreatment steps on cell number and growth after thawing

ASCs were seeded in 96-well flat-bottom plates (1000 or 2000 ASCs per well), cultured for 24 hours, and then assayed using the MTS assay (CellTiter) according to the manufacturer's instructions(ii) an aquous One Solution cell proliferation assay; promega) to assess the number of viable cells. CellTiterThe Aqueous One Solution cell proliferation assay is a ratio used to determine the number of viable cellsAnd (4) a color method. CellTiterAqueous One Solution reagent containing tetrazolium compound [3- (4, 5-dimethylthiazol-2-yl) -5- (3-carboxymethoxyphenyl) -2- (4-sulfophenyl) -2H-tetrazole, MTS]And an electron coupling reagent (phenazine ethosulfate; PES). The MTS tetrazole compound is bioreduced by cells (possibly through NADPH or NADH produced by dehydrogenases in metabolically active cells) into a colored formazan product that is soluble in tissue culture medium.

Briefly, 40 μ L of the reagent was added to 200 μ L of complete DMEM in each well, and the absorbance was measured at 490nm using a Navision system (Microsoft) after 2-3 hours. Each condition was measured in 6 technical replicates. MTS assay results are expressed as percentage of absorbance at 490nm relative to untreated (NT) cells.

NAC pretreatment resulted in an increase in cell number 24 hours after seeding compared to untreated (NT), as assessed by MTS assay (fig. 2) and cell density (fig. 3).

In addition to NAC, exenatide (Exendin-4), IL6, sc79, and LY294 pretreatments were also evaluated. Sc79 is an activator, proliferation and survival promoting signal for the PI3K pathway (Jo et al proceedings of the National Academy of Sciences (2012),109(26): 10581-. DMSO was used as vehicle for sc79 and LY 294. Pretreatment with compounds other than NAC showed no reproducible effect on cell number.

Example 3-further evaluation of NAC pretreatment step ASC proliferation assay on post-thaw ASC cell counts

ASCs pretreated with NAC from donor a or donor B Final Drug Substance (FDS) according to the method described in example 2 were thawed by heating the vial in a 37 ℃ warm bath and rapidly diluting the freezing medium containing DMSO (FBS with 10% DMSO) with fresh complete DMEM. The cells were incubated at room temperature450g were centrifuged for 6 min to remove residual DMSO and seeded in P6 well plates (Falcon #353046) in triplicate in 5mL complete DMEM per well at 3000 cells per well. Cells were washed with 1 × PBS and trypsinized using trypsin-EDTA 0.25% (ThermoFisher) for 8 min at 37 ℃. After trypsin was inactivated using complete DMEM, cells were harvested, centrifuged and resuspended in fresh DMEM; triplicate wells were pooled into a single sample for counting purposes. At 24 hours, 96 hours and 7 days post inoculation, cells were counted in triplicate using an Invitrogen Countess automatic cell counter (Invitrogen) and adding trypan blue as a viability stain (fig. 4A). Using units per surface (cm)2) Viable ASC (trypan blue negative) were counted to perform cell density calculations.

Pretreatment with NAC increased the number of cells counted at 24 hours and 4 days (12,500 cells compared to 9,200 cells/cm at 4 days post-thaw in untreated controls2As shown in fig. 4A). These findings are supported by parallel MTS data showing 15% -20% increase in mitochondrial activity following NAC pretreatment (fig. 4B)&C) In that respect ASC growth had reached confluence before day 7, so there was no significant growth at this time point compared to day 4.

To again confirm the data discussed above, growth assays were performed with two different ASC donors (donor a (don a) and donor b (don b)) after NAC pretreatment. After thawing NAC-pretreated or non-treated cells, the number of cells per donor was analyzed on days 1, 4 and 7 after thawing NAC-pretreated or non-treated cells (fig. 5A & B). Cells from both donors showed an increase in cell number 24 hours after seeding and this increase was maintained in culture for one week (fig. 5). This data demonstrates that NAC pretreatment increases the number of cells after freeze-thaw recovery.

Example 4 Effect of post-thaw treatment with different concentrations of NAC after thawing on cell growth

The effect of post-thaw treatment with different concentrations of NAC (post-thaw NAC treatment) on cell growth was also investigated. ASCs were frozen (without NAC pretreatment), thawed, and then treated with three different NAC concentrations (2, 6, and 12mM) in complete DMEM prior to seeding as discussed above, and analyzed for cell numbers on days 4, 11, and 14. Post-thaw treatment with 2mM NAC resulted in an increase in cell number, which lasted up to 2 weeks in culture (fig. 6). One possible explanation for the lower cell densities observed after thaw treatment with 6mM and 12mM NAC is that these NAC concentrations may affect adhesion of thawed ("floating") ASCs to culture plates.

Example 5 NAC Pre-treatment did not affect the characteristics of the ASCs after thawing and culture

Expression of the four surface markers (CD29, CD73, CD90 and CD105, in compliance with the International society for cell therapy standards (domiinici et al, Cytotherapy (2006)8(4): 315-.

After two weeks in culture (after thawing), the cells were analyzed for characteristics according to standard protocols. Harvested cells were stained with appropriate concentrations of antibody (diluted according to the manufacturer's instructions) as shown in the table below and evaluated using a FACSCalibur cytometer (BD).

Data was analyzed using FCS Express software. Fig. 7 shows that cells express CD29, CD73, CD90, and CD105, and demonstrates that pretreatment of cells with NAC before freezing does not alter expression of ASC trait markers after thawing and culture.

Example 6-NAC Pre-treatment did not significantly affect the ability of thawed ASCs to inhibit stimulated lymphocyte proliferation

After showing that pretreatment of ASCs with NAC leads to in vitro growth advantages, experiments were performed to see if NAC pretreatment affected ACS functional properties. First, the ability of thawed and expanded ASCs (pretreated with NAC2 according to the method in example 2) to inhibit the proliferation of stimulated lymphocytes was measured.

Immunosuppressive assays as previously published: (-Corvo et al, Frontiers in Immunology (2017),8,462; menta et al, Frontiers in Immunology (2014),8,462), isolated Peripheral Blood Mononuclear Cells (PBMCs) from buffy coats provided by the National Transfusion center of the Committed Autonoma of Madrid, using Ficoll-Paque Plus (GE Healthcare Biosciences AB, Uppsala, Sweden) by density centrifugation gradient, and splenocytes obtained from C57/BL6 male mice. For carboxyfluorescein diacetate N-succinimidyl ester (CFSE) labeling, PBMCs or splenocytes were washed extensively to remove FBS, resuspended in 10 μ M CFSE (Sigma-Aldrich, St Louis, MO, USA) solution (107 PBMC or splenocytes per 200 μ l solution), and incubated with constant shaking at 37 ℃ for 10 min. The reaction was stopped by adding ice-cold medium (RPMI + 10% FBS) and the cells were washed 3 times with ice-cold PBS. The cells were then cultured overnight and one aliquot was used to set and control the FL-1 voltage of the CFSE. After standing overnight, CFSE-labeled PBMCs were activated with a pan T cell activation kit (microbeads coated with anti-CD 3, anti-CD 2, and anti-CD 28; Miltenyi Biotec, Auburn, Calif., USA) according to the manufacturer's instructions. CFSE-labeled splenocytes were activated with anti-CD 3(Becton Dickinson) and IL-2(Novartis, Basel, Switzerland). PBMCs or splenocytes (100 ten thousand cells/well) were cultured in 24-well plates in a total volume of 2mL RPMI + 10% FBS alone or with eASCs (4X 10)4Individual cells/well; eASC: PBMC or eASC: splenocytes at a ratio of 1: 25). A ratio of ASC to PBMC of 1:75 allows the assessment of differences between samples under suboptimal conditions. After 5 days of PBMCs and 3 days of splenocytes, cells were harvested, labeled with 7-AAD and anti-CD 3 antibodies, and cell proliferation of the CD3 +/7-AAD-population (viable CD 3T lymphocytes) was determined by flow cytometry based on the loss of CFSE signal. FCSExpress 4(De Novo Software, Glendale, CA, USA) and BD CellQuest were usedTMPro analysis (Becton Dickinson) software analyzed the data. CaliBRITE beads (BD Bioscience, Eremodegem-Aalst, Belgium) were used to calibrate the collection events in the cytometer.

ASCs pretreated with NAC prior to freezing had similar inhibitory potency to untreated cells (fig. 8) (a slight trend was also observed in one or both experiments for NAC pretreatment to increase ASC inhibitory potency).

Example 7 evaluation of NAC Pre-treatment on the Effect of ASC on macrophage and mDC differentiation and function

A second functional in vitro assay performed to assess the effect of NAC on the immunomodulatory capacity of ASCs is the modulation of monocyte differentiation. Fig. 9 shows the time and settings of the experiment.

Blood sample

Buffy coat was obtained from a blood transfusion center in madrid municipality. Approximately 50-60mL of blood was diluted with PBS at room temperature and partitioned between 50mL tubes on top of 15mL room temperature Ficoll Hypaque Plus. The tubes were then centrifuged at 2000rpm for 40 minutes at 10 ℃ without braking or acceleration. White rings of PBMCs were collected, washed in 50mL cold PBS, and centrifuged at 1800rpm for 15 minutes at 10 ℃ without brake or acceleration. After a second wash with 50mL of cold RPMI complete medium (RPMIc: RPMI with 10% FBS, 2mM L-Glu and 100. mu.g/mL penicillin/streptomycin), the tubes were centrifuged at 1500rpm for 15 minutes at 10 ℃ with brake or acceleration. The final wash was performed in 50mL of cold RPMIc and centrifuged at 1200rpm for 15 minutes at 10 ℃ with braking or acceleration. PBMCs were resuspended in RPMIc and counted. Cells were resuspended at 1 hundred million cells/mL under ice-cold and the same volume of cold RPMIc supplemented with 10% DMSO, i.e., the final concentration was 5% DMSO, was added. PBMCs were frozen in liquid nitrogen in 5000 ten thousand vials of PBMCs.

CD14+Isolation of monocytes

Frozen PBMCs vials were thawed and CD14 was counted and isolated using the Dynabeads Untouched human monocyte kit (Dynal #11350D) according to the manufacturer's instructions+CD16-A monocyte.

Culture and differentiation of human monocytes

Separating CD14+CD16-Monocytes (see above) were seeded in 5mL of RPMIc under normoxic conditions at 150 ten thousand cells per 6 wells (Falcon # 353046). The following factors were added to differentiate into non-polarized M0 macrophages or further polarized into M1,M2 macrophage and mature dendritic Cell (mDC) populations (based on a number of publications including Beyer et al, PLoS One (2012)7(9): e 45466; Erbel et al, J.Vis.exp. (2013)76: e 50332; Zhou et al, 2014; Tarique et al, American Journal of Respiratory Cell and Molecular Biology 2015; 53(5):676 688).

Immature DC (iDC) RPMIc +5ng/mL GM-SCF +10ng/mL IL-4 for 5 days

Maturation of DCs (mDC) on day 5, 40ng/mL LPS was added (i.e., 500. mu.L/well RPMIc supplemented with 400ng/mL LPS was added to the pre-existing medium).

Human recombinant GM-CSF (#100-22B) and IL-4(#200-04) were from Peprotech. LPS (# L8274) is from SIGMA. The addition of GM-CSF and IL4 mediated differentiation into Immature Dendritic Cells (iDCs); iDCs were induced to mature into mDCs 5 days after LPS addition, and the phenotype and function of these mature DCs were analyzed 2 days later in the presence or absence of ASC.

Co-culture experiments with ASCs

Freshly isolated human CD14+CD16-Monocytes were co-cultured with ASCs from donor a or donor B in polycarbonate 6 well transwells (Corning #3412 chamber and Falcon #353046 plate).

NAC pretreated ASCs (according to the method in example 2) or untreated ASCs were thawed and 150,000 ASCs were seeded on a transwell chamber 16 hours or 24 hours prior to co-culture set-up in 1mL RPMIc medium and 150 ten thousand monocytes were seeded at the bottom of the wells in 4mL RPMIc medium. Differentiation was performed using the same factors as those used for differentiating monocytes alone (see above, i.e., addition of GM-CSF and IL4 to induce differentiation into iDCs; LPS was added after 5 days to induce maturation of iDCs into mDCs, and after 2 days, the phenotype and function of these mature DCs were analyzed in the presence or absence of ASC). Throughout the duration of the differentiation process, ASCs were stored in the transwell chamber.

No activated clusters formed on the plates after cocultivation of mDCs with NAC-pretreated or untreated ASCs, indicating that ASCs modulate the activation of mDCs and that this effect is not disrupted by NAC pretreatment (see microscope images 2x magnification (fig. 10) and 20x magnification (fig. 11)).

Example 8-NAC Pre-treatment did not significantly alter the ability of ASCs to modulate phagocytosis of mDC by Staphylococcus aureus particles

The effect of NAC pretreatment (according to the method in example 2) on the ability of thawed ACS to modulate the phagocytosis of staphylococcus aureus particles of mDCs was analyzed.

Following in vitro mono-or co-culture differentiation (differentiation conditions including cytokines used, differentiation concentration and number of differentiation provided in example 7), macrophages and mdcs were harvested with 0.05% trypsin-EDTA at 37 ℃ for 10 minutes. The phagocytic potential of polarized macrophages or mdcs was assessed using pHRodo Red-conjugated staphylococcus aureus particles (Life Technologies # a10010) according to the manufacturer's instructions. Briefly, 50,000 mdcs were transferred to 96-well U-bottom wells (Corning #3799) and the cells were allowed to stand in RPMIc for 60 minutes. Prior to use, lyophilized pHRodo-coupled particles were reconstituted in 1mL per vial of RPMIc and the particles were sonicated at 20% amplitude for 5 minutes. Then, 50. mu.L of pHRodo Zymosan was added to each well, and the cells were incubated at 37 ℃ for 60 minutes under normal oxygen. Then, phagocytosis was stopped on ice and cells were washed and stained with 5 μ L7-aminoactinomycin D (7AAD) before FACS analysis in a Fortessa cytometer (BD). Negative controls for phagocytosis were cells without pHRodo reagent. The results were analyzed in FlowJo software. The fluorescence intensity in the PE channel is proportional to the amount of bacterial particles phagocytosed by each cell.

Figure 12 shows that the presence of ASC under non-contact conditions (i.e. co-culturing mDC and ASC cells in a transwell plate) results in a new population of cells that appear more intense in the fluorescent channel, i.e. cells that have phagocytosed fluorescent particles. NAC pretreatment of ASCs with NAC did not alter their ability to increase phagocytic potential of mDCs.

Example 9-NAC pretreatment effect on ASC-mediated surface expression on mature dendritic cells.

The ability of mdcs to phagocytose bacteria is associated with the expression of phagocytic markers, such as CD209(DC-SIGN), CD206 (mannose receptor), or CD163 (scavenger receptor). These membrane receptors recognize specific patterns on fungal, bacterial and parasitic surfaces and mediate their phagocytosis by monocytes, macrophages and DCs. The CD163 receptor also intervenes in the clearance of cellular debris from apoptotic cells following tissue injury, promoting the wound healing process.

The effect of NAC pretreatment (according to example 2) on thawed ASC mediated surface expression of the mature dendritic cell phagocytic receptors CD206 (mannose receptor) and CD163 (scavenger receptor) was measured by flow cytometry.

Phenotypic characterization

Following in vitro mono-or co-culture differentiation (differentiation conditions, including cytokines used, differentiation concentration and number of differentiation times, provided in example 7), macrophages and mdcs were harvested with 0.05% trypsin-EDTA at 37 ℃ for 10 minutes after collecting the supernatant and freezing for future cytokine and/or HPLC analysis. After mDC counting, they were dispensed into 96-well V-plates for staining (Nunc # 249570). Cells were incubated on ice for 15 min in Blue MACS buffer with 1% human serum to block Fc γ receptor-mediated nonspecific antibody binding. Subsequently, the cells were stained on ice for 20 minutes with the following antibody mixture (staining in 50. mu.L of 1:10 antibody dilution; it was 1:20 dilution except CD 64):

key dyeing (all holes have 7AAD)
1 CD14-APC/HLAII-FITC 1:50/CD86-PE
2 CD14-APC/CD206-PE/CD209-FITC
3 CD14-APC/CD163-PE
4 CD14-APC/CD80-FITC/CD64-PE 1:20
5 CD14-APC/CD1a-PE

Details of the antibodies used are listed in the following table:

name (R) Fluorescent dyes Host computer Cloning Classification number Company(s)
CD1a PE Mouse HI149 555807 BD
CD14 APC Mouse M5E1 555399 BD
CD64 PE Mouse 10.1 CD6404 Miltenyi Biotech
CD68 PeCy7 Mouse 27-35 560542 BD
CD80 PE Mouse L307.4 557227 BD
CD86 PE Mouse IT2.2 555665 BD
CD206 PE Mouse 19.2 555954 BD
CD209 FITC Mouse DCN46 551264 BD
HLA-II PE Mouse WR18 MA1-80680 Ebiosciences

Cell viability was assessed by adding 5 μ L of 7AAD to each well and staining for 10min on ice, and samples were collected in a BD Fortessa cytometer. The results were analyzed in FSC Express software.

The ability of mdcs to phagocytose bacteria is associated with the expression of phagocytic markers, such as CD209(DC-SIGN), CD206 (mannose receptor), or CD163 (scavenger receptor). These membrane receptors recognize specific patterns on the surface of fungi, bacteria and parasites and mediate their phagocytosis by monocytes, macrophages and DCs. The CD163 receptor additionally intervenes in the clearance of cellular debris from apoptotic cells following tissue injury, promoting the wound healing process. The effect of NAC pretreatment (according to example 2) on thawed ASC mediated surface expression of the mature dendritic cell phagocytic receptors CD206 (mannose receptor) and CD163 (scavenger receptor) was measured by flow cytometry.

ASCs upregulated the expression of CD206 and CD163 markers on the surface of monocytes, macrophages and mdcs, and this upregulation was intact even when ASCs were pre-treated with NAC (fig. 13 and 14).

The effect of NAC pretreatment (according to example 2) on thawed ASC-mediated effects on CD14 and CD1a surface expression on mature dendritic cells was also measured by flow cytometry. mdcs are CD14-CD1a +. CD1a is an antigen presenting molecule and mediates the presentation of antigens to other cells of the immune system via mdcs to activate their responses. ASCs modulate the phenotype of these mdcs, transforming them into CD14+ CD1 a-cells. This population has been suggested to have anti-inflammatory and regulatory properties (Chang et al, Journal of Immunology,165(7), 3584-3591).

Fig. 15 shows that NAC pretreatment ASCs did not alter the ability of thawed ASCs to induce this mDC-mediated population formation.

Numbered embodiments

The invention also provides the following numbered embodiments:

1. a method for the cryopreservation of stem cells, the method comprising the steps of:

a. treating a stem cell population with N-acetylcysteine (NAC) to obtain a treated stem cell population; and

b. freezing the treated stem cell population to obtain a frozen stem cell population.

2. The method of embodiment 1, wherein the method comprises the steps of:

a. treating the stem cell population with NAC to obtain a treated stem cell population;

b. freezing the treated stem cell population to obtain a frozen stem cell population; and

c. thawing the frozen stem cell population to obtain a thawed stem cell population.

3. The method of embodiment 1 or embodiment 2, wherein the method comprises the steps of:

a. treating a stem cell population with NAC to obtain a treated stem cell population;

b. washing the treated stem cell population to remove NAC and obtain a washed stem cell population, and freezing the washed stem cell population to obtain a frozen stem cell population; and

c. thawing the frozen stem cell population to obtain a thawed stem cell population.

4. The method of any one of the preceding embodiments, wherein the treating step comprises incubating the stem cell population with NAC for at least about 1, 2, 4, 6, 8, 10, 12, 16, 24, or 48 hours prior to freezing the stem cell population.

5. The method of any one of the preceding embodiments, wherein the treating step comprises adding NAC to the stem cell population to an initial concentration of about 0.5-10 mM.

6. The method of embodiment 5, wherein the treating step comprises one or more further additions of NAC to maintain a predetermined level of NAC concentration.

7. The method of any one of embodiments 2-6, wherein the method further comprises the steps of:

d. culturing the thawed stem cell population to obtain an expanded stem cell population.

8. The method of any one of embodiments 2-6, wherein the method further comprises the steps of:

d. culturing said thawed stem cell population in the presence of NAC to obtain an expanded stem cell population.

9. The method of embodiment 8, wherein the culturing step comprises adding NAC to an initial concentration of about 0.5-5 mM.

10. The method of embodiment 9, wherein said culturing step comprises one or more further additions of NAC to maintain a predetermined level of NAC concentration.

11. The method according to any one of embodiments 8-10, wherein the method further comprises the step of washing the expanded stem cell population to remove the NAC and obtaining a washed and expanded stem cell population.

12. The method of any one of embodiments 2-11, wherein the method further comprises the step of washing the thawed stem cell population or expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier.

13. The method of any one of embodiments 7-12, wherein the method further comprises the steps of:

e. freezing the expanded stem cell population or the washed and expanded stem cell population to obtain a frozen expanded stem cell population or a frozen, washed and expanded stem cell population.

14. The method of any one of embodiments 7-13, wherein the method further comprises the steps of:

e. freezing the expanded stem cell population or the washed and expanded stem cell population to obtain a frozen expanded stem cell population or a frozen, washed and expanded stem cell population; and

f. thawing the frozen expanded stem cell population or the frozen, washed and expanded stem cell population to obtain a thawed expanded stem cell population.

15. The method of embodiment 14, wherein the method further comprises the steps of:

g. washing the thawed expanded stem cell population and resuspending the cells in a pharmaceutically acceptable carrier.

16. A method for the cryopreservation of stem cells, the method comprising the steps of:

a. freezing a stem cell population to obtain a frozen stem cell population;

b. thawing the frozen stem cell population to obtain a thawed stem cell population; and

c. culturing said thawed stem cell population in the presence of NAC to obtain an expanded stem cell population.

17. The method of embodiment 16, wherein said culturing step comprises adding NAC to an initial concentration of about 0.5-5 mM.

18. The method of embodiment 17, wherein said culturing step comprises one or more further additions of NAC to maintain a predetermined level of NAC concentration.

19. The method of any one of the preceding embodiments, wherein the freezing step comprises reducing the temperature to-70 ℃ to-130 ℃ at a rate of about-0.5 to about-10 ℃/minute.

20. The method of any one of the preceding embodiments, wherein the freezing step comprises reducing the temperature from +4 ℃ to-100 to-180 ℃ within 10-60 min.

21. The method of any one of the preceding embodiments, wherein the population of stem cells is thawed at 37 ℃.

22. The method of any one of the preceding embodiments, wherein the cell density of the frozen stem cell population ranges from about 100 to about 5000 ten thousand cells/mL, preferably about 2500 ten thousand cells/mL.

23. The method of any one of the preceding embodiments, wherein the population of stem cells is substantially pure.

24. The method of any one of the preceding embodiments, wherein the stem cells are Mesenchymal Stem Cells (MSCs).

25. The method of any one of the preceding embodiments, wherein the stem cells are adipose-derived stromal stem cells (ASCs).

26. The method of any one of the preceding embodiments, wherein the stem cell is a human cell.

27. The method of any one of the preceding embodiments, wherein the method further comprises the step of resuspending the cells in a pharmaceutically acceptable carrier.

28. The method of any one of the preceding embodiments, wherein the method comprises freezing the population of stem cells in a plurality of frozen vials.

29. The method of any one of the preceding embodiments, wherein the method comprises repeating the steps of any one of the preceding embodiments for a plurality of stem cell populations.

30. The method of embodiment 29, wherein the method comprises freezing the plurality of stem cell populations in a plurality of frozen vials.

31. The method of embodiment 28 or embodiment 30, wherein the method comprises storing the plurality of cryo-preservation vials in a liquid nitrogen storage container for at least one month, at least 2 months, at least 3 months, at least 6 months, or at least 1 year.

32. A liquid nitrogen storage container containing a plurality of cryopreserved vials obtained according to the method of embodiment 28 or embodiment 30.

33. A population of stem cells obtained by the method of any one of embodiments 1-31.

34. The method of any one of embodiments 1-31 or the stem cell population of embodiment 33, wherein the number of viable cells is increased after thawing, and optionally culturing, for about 1 day or about 4 days, as compared to a control stem cell population.

35. The method of any one of embodiments 1-31 and 34 or the stem cell population of embodiment 33 or 34, wherein the number of viable cells after thawing is increased at least about 1.05-fold, at least about 1.1-fold, at least about 1.2-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.5-fold, at least about 1.6-fold, at least about 2-fold, or at least about 5-fold compared to a control stem cell population.

36. The method of any one of embodiments 1-31, 34 or 35 or the stem cell population of embodiments 33-35, wherein the growth rate of the stem cell population after thawing is increased at least about 1.03-fold, 1.05-fold, at least about 1.1-fold, at least about 1.15-fold, at least about 1.2-fold, at least about 1.25-fold, at least about 1.3-fold, at least about 1.4-fold, at least about 1.6-fold, or at least about 2-fold compared to a control stem cell population.

37. The method of any one of embodiments 1-31, 34-36 or the stem cell population of embodiments 33-36, wherein mitochondrial activity is increased by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 30%, at least about 35%, at least about 40%, or at least about 50% after thawing and optionally culturing for about 1 or about 4 days as compared to a control stem cell population.

38. The method of any one of embodiments 1-31, 34-37 or the stem cell population of embodiments 33-37, wherein the time taken for the ASCs to recover after thawing is reduced compared to a control stem cell population.

39. The method of any one of embodiments 1-31, 34-38 or the stem cell population of embodiments 33-38, wherein the number of hours it takes for a cell to recover after thawing is reduced by at least about 1.1-fold, at least about 1.2-fold, at least about 1.4-fold, at least about 1.6-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, or at least about 5-fold relative to a control stem cell population.

40. A cryopreservation composition comprising a stem cell population of any one of embodiments 33-38 and a cryopreservation medium.

41. The cryopreservation composition of embodiment 40 wherein the composition is frozen.

42. The cryopreservation composition of embodiment 40 or embodiment 41 wherein the composition contains NAC.

43. A pharmaceutical composition comprising a population of stem cells of any one of embodiments 33-38 and a pharmaceutically acceptable carrier.

44. The pharmaceutical composition of embodiment 43, wherein the composition comprises from about 100 million cells to about 1.5 million cells, preferably about 3000 million cells or about 1.2 million cells.

45. The pharmaceutical composition of embodiment 43 or embodiment 44, wherein the cell density is about 100-2000 ten thousand cells/mL.

Use of NAC for cryopreservation of stem cells.

47. Use of a NAC according to embodiment 46 in the method of any one of embodiments 1-31 and 34-39.

48. Use of the stem cell population of any one of embodiments 33-39, the pharmaceutical composition of any one of embodiments 43-45, or the cryopreservation composition of embodiments 40-42 for treatment.

49. Use of a stem cell population of any one of embodiments 33-39, a pharmaceutical composition of any one of embodiments 43-45, or a cryopreserved composition of embodiments 40-42 for treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof.

50. A method of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease, or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g., type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection, the method comprising administering to a subject in need thereof a stem cell population according to any one of embodiments 33-39, a pharmaceutical composition according to any one of embodiments 43-45, or a cryopreservation composition according to embodiments 40-42.

51. A stem cell population for use in a method for treating fistulas and/or treating and/or preventing inflammatory disorders, autoimmune diseases or immune-mediated diseases, such as sepsis, rheumatoid arthritis, allergies (e.g. type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of:

a. treating a stem cell population with NAC to obtain a treated stem cell population;

b. freezing the treated stem cell population to obtain a frozen stem cell population;

c. thawing the frozen stem cell population to obtain a thawed stem cell population;

d. optionally culturing the thawed stem cell population to obtain an expanded stem cell population; and

e. administering the population of stem cells to the patient.

52. A method of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g. type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of:

a. treating a stem cell population with NAC to obtain a treated stem cell population;

b. freezing the treated stem cell population to obtain a frozen stem cell population;

c. thawing the frozen stem cell population to obtain a thawed stem cell population;

d. optionally culturing the thawed stem cell population to obtain an expanded stem cell population; to be provided with

e. Administering the population of stem cells to the patient.

53. The stem cell population for use according to embodiment 51 or the method of treatment of embodiment 52, wherein the method further comprises any of the steps as defined in embodiments 3-14, 18-31 or 34-39 prior to administering the stem cell population to a patient.

54. A stem cell population for use in a method for treating fistulas and/or treating and/or preventing inflammatory disorders, autoimmune diseases or immune-mediated diseases, such as sepsis, rheumatoid arthritis, allergies (e.g. type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, wherein the method comprises the steps of:

a. freezing a stem cell population to obtain a frozen stem cell population;

b. thawing the frozen stem cell population to obtain a thawed stem cell population;

c. culturing said thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and

d. administering the population of stem cells to the patient.

55. A method of treating a fistula and/or treating and/or preventing an inflammatory disorder, an autoimmune disease or an immune-mediated disease, such as sepsis, rheumatoid arthritis, allergy (e.g. type IV hypersensitivity), irritable bowel disease, crohn's disease, ulcerative colitis, or organ rejection in a patient in need thereof, the method comprising the steps of:

a. freezing a stem cell population to obtain a frozen stem cell population;

b. thawing the frozen stem cell population to obtain a thawed stem cell population;

c. culturing said thawed stem cell population in the presence of NAC to obtain an expanded stem cell population; and

d. administering the population of stem cells to the patient.

56. The stem cell population for use according to embodiment 54 or the method of treatment of embodiment 55, wherein the method further comprises any step as defined in any one of embodiments 15-31 or 34-39 prior to administering the stem cell population to a patient.

57. The stem cell population, pharmaceutical composition or cryopreservation composition for use according to any of embodiments 48, 49, 51, 53, 54 or 56, or the method of any of embodiments 50, 52, 53, 55 or 56, wherein the method comprises administering between about 100 and 1.5 million cells, preferably about 3000 million stem cells or about 1.2 million stem cells.

58. The stem cell population, pharmaceutical composition or cryopreservation composition for use according to any of embodiments 48, 49, 51, 53, 54, 56 or 57, or the method of any of embodiments 50, 52, 53, 55-57, wherein the method comprises administering from about 100 to about 1000 ten thousand cells/kg.

59. The stem cell population or the pharmaceutical composition or the cryopreservation composition for use according to any one of embodiments 48, 49, 51, 53, 54, 56-58, or the method of any one of embodiments 50, 52, 53, 55-58, wherein the method comprises injecting the stem cell population or the pharmaceutical composition of any one of embodiments 43-45 or the cryopreservation composition of any one of embodiments 40-42.

60. A stem cell population or a pharmaceutical or cryopreservation composition for use according to any of embodiments 48, 49, 51, 53, 54, 56-59, or a method of any of embodiments 50, 52, 53, 55-59, wherein the stem cells are as defined in any of embodiments 23-26.

61. The stem cell population or the pharmaceutical or cryopreservation composition for use according to any one of embodiments 48, 49, 51, 53, 54, 56-60, or the method of any one of embodiments 50, 52, 53, 55-60, wherein the stem cells are allogeneic or autologous.

62. A cryopreservation kit comprising: a frozen vial, a NAC-containing container, and a container comprising a population of stem cells.

59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:过氧化氢消毒剂组合物

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!