WO2018123628A1 - Cell pharmaceutical composition, disease treatment kit, and cell suspension solution - Google Patents

Cell pharmaceutical composition, disease treatment kit, and cell suspension solution Download PDF

Info

Publication number
WO2018123628A1
WO2018123628A1 PCT/JP2017/044993 JP2017044993W WO2018123628A1 WO 2018123628 A1 WO2018123628 A1 WO 2018123628A1 JP 2017044993 W JP2017044993 W JP 2017044993W WO 2018123628 A1 WO2018123628 A1 WO 2018123628A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
solution
pharmaceutical composition
mesenchymal stem
Prior art date
Application number
PCT/JP2017/044993
Other languages
French (fr)
Japanese (ja)
Inventor
里枝 武田
くみ子 長岡
陽子 堀内
輝 長谷川
琴絵 玉田
Original Assignee
ロート製薬株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by ロート製薬株式会社 filed Critical ロート製薬株式会社
Priority to JP2018559040A priority Critical patent/JPWO2018123628A1/en
Publication of WO2018123628A1 publication Critical patent/WO2018123628A1/en
Priority to JP2022165115A priority patent/JP2022179749A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/35Fat tissue; Adipocytes; Stromal cells; Connective tissues
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a cell pharmaceutical composition, a disease treatment kit and a cell suspension solution.
  • stem cells such as iPS cells, hematopoietic stem cells, mesenchymal stem cells, skin cells, cardiomyocytes, etc. have moved from the basic research stage to the development stage, and are currently used in clinical settings. There are also.
  • the functions of cells themselves are used directly or indirectly for disease treatment, and the functions of cells and tissues of damaged patients are newly differentiated from stem cells and It is expected to be supplemented by organs.
  • mesenchymal stem cells are pluripotent progenitor cells isolated from bone marrow for the first time by Friedenstein (1982) (Non-patent Document 1). These mesenchymal stem cells have been revealed to exist in various tissues such as bone marrow, umbilical cord, and fat, and mesenchymal stem cell transplantation is expected as a new treatment method for various intractable diseases ( Patent Documents 1 to 4). Recently, it is known that cells having an equivalent function exist in stromal cells of fetal appendages such as adipose tissue, placenta, umbilical cord, and egg membrane. Therefore, the mesenchymal stem cell may be referred to as a stromal cell (Mesenchymal Stroma Cell).
  • the cells may be suspended in a solution for the purpose of ensuring safety and facilitating administration of the cells.
  • a solution for the purpose of ensuring safety and facilitating administration of the cells.
  • the survival rate of the cells in the suspension is gradually reduced and sufficient pharmacological action is achieved.
  • problems such as formation of emboli in the patient's pulmonary veins and the like that may not be obtained, cells may aggregate and clog in the cannula, and the like. Then, this invention makes it a subject to provide the cell pharmaceutical composition which can maintain the survival rate of a cell high over a long time.
  • the inventors of the present invention include Na + , Cl ⁇ , K + and Ca 2+ as electrolytes such as Ringer's solution, Ringer's acetate solution or Ringer's bicarbonate solution, as cells for disease treatment.
  • the present inventors have found that when a drip infusion or the like is performed after suspending in an isotonic electrolyte solution that does not contain carbohydrates, a decrease in the survival rate of cells during administration is remarkably suppressed.
  • a pharmaceutical composition containing cells can maintain a good state of cells and maintain a high survival rate over a long period of time, it has excellent therapeutic effects on various diseases. be able to. That is, the gist of the present invention is as follows.
  • the solution for cell suspension according to [1] wherein the solution for cell suspension further comprises at least one electrolyte selected from the group consisting of CH 3 COO ⁇ , HCO 3 ⁇ , citrate ions and lactate ions.
  • Cell pharmaceutical composition is an isotonic electrolytic solution containing Na + , Cl ⁇ , K + and Ca 2+ as electrolytes and not containing carbohydrates.
  • a cell suspension solution for a cell pharmaceutical composition which is an isotonic electrolyte solution containing Na + , Cl ⁇ , K + and Ca 2+ as an electrolyte and free of carbohydrates.
  • the cell pharmaceutical composition of the present invention can maintain a good cell state and maintain its survival rate in a high state for a long time, it can be expected to have an excellent therapeutic effect on various diseases.
  • the cell pharmaceutical composition, disease treatment kit, and cell suspension solution for injection of the present invention will be described in detail.
  • the cell pharmaceutical composition of the present invention contains (A) cells and (B) a cell suspension solution, and (B) the cell suspension solution contains Na + , Cl ⁇ , K + and Ca 2+ as electrolytes. And an isotonic electrolyte solution that does not contain carbohydrates.
  • the “cell pharmaceutical composition” refers to a pharmaceutical composition containing cells, which exhibits a therapeutic effect on diseases due to the functions of the cells.
  • the cell pharmaceutical composition of the present invention is excellent for various diseases because the cell viability can be maintained in a high state for a long time by suspending the cells in the specific solution. A therapeutic effect can be achieved.
  • the cell pharmaceutical composition of the present invention may contain other drugs having a therapeutic effect on diseases. Furthermore, as long as the effects of the present invention are not impaired, other components may be contained.
  • (A) cells, (B) cell suspension solutions, other drugs, and other components contained in the cell pharmaceutical composition of the present invention will be described in detail.
  • the (A) cell is not particularly limited as long as it has an effect on the treatment of diseases.
  • mesenchymal stem cells peripheral blood mononuclear cells (neutrophils, eosinophils, basophils, Lymphocytes, monocytes, etc.), red blood cells, T cells, NK cells, NKT cells, NKM cells, LAK cells, dendritic cells, fibroblasts, hematopoietic stem cells, iPS cells, ES cells, bone marrow cells, cardiomyocytes
  • mesenchymal stem cells, peripheral blood mononuclear cells, and bone marrow cells are preferable from the viewpoint that (B) the cell suspension solution described later is excellent in viability maintenance effect.
  • the mesenchymal stem cell refers to one or more cells belonging to the mesenchymal system, preferably two or more cells, more preferably three or more cells (bone cells, cardiomyocytes, chondrocytes, tendon cells, fat cells, etc.). It means a cell that has differentiation ability and can proliferate while maintaining the ability.
  • the term mesenchymal stem cell used in the present invention means the same cell as the stromal cell, and does not particularly distinguish them. Moreover, it may be simply described as a mesenchymal cell.
  • tissues containing mesenchymal stem cells include adipose tissue, umbilical cord, bone marrow, umbilical cord blood, endometrium, placenta, amniotic membrane, chorion, decidua, dermis, skeletal muscle, periosteum, dental follicle, periodontal ligament, Examples include dental pulp and tooth germ.
  • an adipose tissue-derived mesenchymal stem cell means a mesenchymal stem cell contained in an adipose tissue, and may be referred to as an adipose-derived mesenchymal stem cell.
  • adipose-derived mesenchymal stem cells from the viewpoint of effectiveness for treatment of various diseases, from the viewpoint of availability, adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, placenta-derived mesenchymal stem cells, Dental pulp-derived mesenchymal stem cells are preferred, and adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and bone marrow-derived mesenchymal stem cells are more preferred.
  • the mesenchymal stem cells in the present invention may be derived from the same species as the subject to be treated (subject) or from a different species.
  • examples of the mesenchymal stem cell species in the present invention include humans, horses, cows, sheep, pigs, dogs, cats, rabbits, mice, and rats, preferably cells derived from the same species as the subject to be treated (subject).
  • the mesenchymal stem cells in the present invention may be derived from the subject (subject) to be treated, that is, autologous cells (allogeneic), or derived from another subject of the same species, ie, allogeneic cells (allogeneic). ). Preferred are allogeneic cells (allogeneic).
  • mesenchymal stem cells are unlikely to cause rejection even for allogeneic subjects, cells prepared by pre-expanding donor cells that have been expanded and cryopreserved are used in the cell pharmaceutical composition of the present invention (A ) It can be used as a mesenchymal stem cell as a cell. Therefore, compared to the case of preparing and using autologous mesenchymal stem cells, commercialization is easy, and from the viewpoint that a certain effect can be easily obtained stably, the mesenchymal stem cells in the present invention, More preferably, it is allogeneic.
  • the mesenchymal stem cell means an arbitrary cell population including the mesenchymal stem cell.
  • the cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98 % Or 99% are mesenchymal stem cells.
  • an adipose tissue means a tissue containing stromal cells including adipocytes and microvascular cells, and is, for example, a tissue obtained by surgical excision or aspiration of mammalian subcutaneous fat.
  • Adipose tissue can be obtained from subcutaneous fat. It is preferably obtained from the same animal as the subject of administration of the adipose-derived mesenchymal stem cells described later, and more preferably human subcutaneous fat in consideration of administration to humans.
  • An individual supplying subcutaneous fat may be alive or dead, but the adipose tissue used in the present invention is preferably a tissue collected from a living individual.
  • liposuction is exemplified by PAL (power assist) liposuction, Erconia laser liposuction, or body jet liposuction. From the viewpoint of maintaining the state of cells, ultrasound is used. It is preferable not to use.
  • the umbilical cord is a white tubular tissue that connects the fetus and the placenta, and is composed of umbilical vein, umbilical artery, collagenous tissue (Wharton's Jelly), umbilical matrix itself, etc., and mesenchymal stem cells Including many.
  • the umbilical cord is preferably obtained from the same animal as the subject (administration subject) using the cell pharmaceutical composition of the present invention, and more preferably in consideration of administering the cell pharmaceutical composition of the present invention to a human.
  • the human umbilical cord is preferably obtained from the same animal as the subject (administration subject) using the cell pharmaceutical composition of the present invention, and more preferably in consideration of administering the cell pharmaceutical composition of the present invention to a human.
  • the bone marrow refers to a soft tissue filling the bone lumen, and is a hematopoietic organ.
  • Bone marrow fluid is present in the bone marrow, and the cells present therein are called bone marrow cells.
  • Bone marrow cells include erythrocytes, granulocytes, megakaryocytes, lymphocytes, adipocytes and the like, as well as mesenchymal stem cells, hematopoietic stem cells, vascular endothelial progenitor cells, and the like.
  • Bone marrow cells can be collected, for example, from human iliac bone, long bone, or other bone.
  • each tissue-derived mesenchymal stem cell such as adipose-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell is a fat-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, It means any cell population containing mesenchymal stem cells derived from each tissue such as bone marrow-derived mesenchymal stem cells.
  • the cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98 % Or 99% are mesenchymal stem cells derived from various tissues such as adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and bone marrow-derived mesenchymal stem cells.
  • Mesenchymal stem cells have growth characteristics (eg, population doubling ability from aging to aging, doubling time), karyotype analysis (eg, normal karyotype, maternal line or neonatal line), flow cytometry ( For example, surface marker expression by FACS analysis, immunohistochemistry and / or immunocytochemistry (eg, epitope detection), gene expression profiling (eg, gene chip array; polymerase chain such as reverse transcription PCR, real-time PCR, conventional PCR, etc. Reaction), miRNA expression profiling, protein array, protein secretion such as cytokines (eg, plasma coagulation analysis, ELISA, cytokine array), metabolites (metabolome analysis), other methods known in the art, etc. May be.
  • growth characteristics eg, population doubling ability from aging to aging, doubling time
  • karyotype analysis eg, normal karyotype, maternal line or neonatal line
  • flow cytometry For example, surface marker expression by FACS analysis, immunohisto
  • Mesenchymal stem cells can be prepared by methods well known to those skilled in the art. Below, the preparation method of a fat-derived mesenchymal stem cell is demonstrated as an example. Adipose-derived mesenchymal stem cells may be obtained by the production method described in, for example, US Pat. No. 6,777,231, and can be produced, for example, by a method including the following steps (i) to (iii).
  • washing may be performed by sedimentation with vigorous stirring using a physiologically compatible saline solution (eg, phosphate buffered saline (PBS)).
  • PBS physiologically compatible saline
  • impurities also called debris, such as damaged tissue, blood, and red blood cells
  • washing and sedimentation are generally repeated until the debris is totally removed from the supernatant. Since the remaining cells exist as lumps of various sizes, in order to dissociate them while minimizing damage to the cells themselves, the washed cell lumps are made to have an enzyme (eg, an enzyme that weakens or breaks cell-cell junctions).
  • collagenase dispase or trypsin.
  • the amount of such enzyme and the duration of treatment vary depending on the conditions used, but are known in the art.
  • the cell mass can be broken down by other treatment methods such as mechanical agitation, ultrasonic energy, thermal energy, etc., but with minimal cell damage
  • an enzyme in order to minimize harmful effects on cells, it is desirable to deactivate the enzyme using a medium or the like after a suitable period of time.
  • the cell suspension obtained by the step (i) includes a slurry or suspension of aggregated cells, and various kinds of contaminated cells such as erythrocytes, smooth muscle cells, endothelial cells, and fibroblasts. Therefore, the cells in the aggregated state and these contaminated cells may be separated and removed, but they can be removed by adhesion and washing in step (iii) to be described later. May be. When contaminating cells are separated and removed, this can be achieved by centrifugation that forcibly separates the cells into a supernatant and a precipitate. The resulting precipitate containing contaminating cells is suspended in a physiologically compatible solvent.
  • Suspended cells may contain erythrocytes, but erythrocytes are excluded by selection by adhesion to the individual surface described later, and thus a lysis step is not always necessary.
  • a method for selectively lysing erythrocytes for example, a method well known in the art such as incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride can be used. After lysis, the lysate may be separated from the desired cells, for example, by filtration, centrifugation, or density fractionation.
  • the cells in step (ii), in order to increase the purity of the mesenchymal stem cells in the suspended cells, the cells may be washed once or continuously several times, centrifuged, and resuspended in the medium. Alternatively, cells may be separated based on cell surface marker profile or based on cell size and granularity.
  • the medium used in the resuspension is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes basal medium added with serum and / or albumin, transferrin, fatty acid, One or more serum substitutes such as insulin, sodium selenite, cholesterol, collagen precursor, trace elements, 2-mercaptoethanol, 3′-thiolglycerol, and the like may be added.
  • substances such as lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular compounds, antibiotics, antioxidants, pyruvate, buffers, inorganic salts, etc. are added as necessary. May be.
  • basal medium examples include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, Ham's F16 medium.
  • EMEM Eagle's Minimum Essential Medium
  • DMEM Dulbecco's modified Eagle's Medium
  • Ham's F12 medium Ham's F16 medium.
  • Examples include a medium, Fischer's medium, MCDB201 medium, and a mixed medium thereof.
  • the serum examples include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like.
  • FBS fetal bovine serum
  • bovine serum bovine serum
  • calf serum goat serum
  • horse serum horse serum
  • pig serum sheep serum
  • rabbit serum rat serum
  • 5 v / v% to 15 v / v% preferably 10 v / v% may be added to the basal medium.
  • fatty acid examples include, but are not limited to, linoleic acid, oleic acid, linolenic acid, arachidonic acid, myristic acid, palmitoyl acid, palmitic acid, and stearic acid.
  • lipid examples include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, phosphatidylcholine, and the like.
  • Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like.
  • proteins include, but are not limited to, ecotin, reduced glutathione, fibronectin and ⁇ 2-microglobulin.
  • polysaccharide examples include glycosaminoglycans, and among the glycosaminoglycans, hyaluronic acid, heparan sulfate and the like are particularly exemplified, but the polysaccharide is not limited thereto.
  • Growth factors include, for example, platelet derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF- ⁇ ), hepatocyte growth factor (HGF), epidermal growth factor (EGF) Examples include, but are not limited to, connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF), and the like. From the viewpoint of using the adipose-derived mesenchymal stem cells obtained in the present invention for cell transplantation, it is preferable to use a (xenofree) medium that does not contain components derived from different species such as serum.
  • a (xenofree) medium that does not contain components derived from different species such as serum.
  • Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from, for example, PromoCell, Lonza, Biological Industries, Veritas, R & D Systems, Corning, and Roto. Has been.
  • the appropriate cell density and the appropriate cell medium are used on the solid surface without differentiating the cells in the cell suspension obtained in step (ii).
  • Culture under culture conditions the “solid surface” means any material capable of binding / adhering the adipose-derived mesenchymal stem cells in the present invention.
  • such a material is a plastic material that has been treated to promote the attachment and adhesion of mammalian cells to its surface.
  • the shape of the culture vessel having a solid surface is not particularly limited, but a petri dish or a flask is preferably used. The cells are washed after incubation to remove unbound cells and cell debris.
  • cells that finally remain attached and adhered to the solid surface can be selected as a cell population of adipose-derived mesenchymal stem cells.
  • the surface antigen may be analyzed by a conventional method using flow cytometry or the like. Further, the ability to differentiate into each cell line may be examined, and such differentiation can be performed by conventional methods.
  • Mesenchymal stem cells in the present invention can be prepared as described above, but may be defined as cells having the following characteristics; (1) It exhibits adhesiveness to plastic under the culture conditions in a standard medium. (2) Surface antigens CD44, CD73, CD90 are positive, CD31, CD45 are negative, and (3) Differentiated into bone cells, adipocytes, and chondrocytes under culture conditions.
  • peripheral blood mononuclear cells refer to fractions containing lymphocytes, neutrophils, eosinophils, basophils, and monocytes, which are obtained from human or animal peripheral blood.
  • Peripheral blood mononuclear cells can be separated from peripheral blood by density gradient centrifugation using Ficoll-hypaque (registered trademark) or the like.
  • Peripheral blood mononuclear cells as cells (A) in the present invention may be cells separated from peripheral blood, or they may be cultured with various factors, low molecular compounds, antibodies, etc. as necessary. It may be proliferated and activated.
  • the (A) cell in the present invention may be a cell that has been appropriately cryopreserved and thawed as long as it has a therapeutic effect on various diseases.
  • cryopreservation can be performed by suspending (A) cells in a cryopreservation solution well known to those skilled in the art and cooling. Suspension can be performed by detaching the cells with a release agent such as trypsin as necessary, transferring them to a cryopreservation container, treating them appropriately, and adding a cryopreservation solution.
  • the cryopreservation solution may contain DMSO (Dimethylsulfoxide) as a frost damage protective agent.
  • DMSO Dimethylsulfoxide
  • DMSO is also known to have differentiation-inducing properties for mesenchymal stem cells.
  • glycerol, propylene glycol or polysaccharides are exemplified.
  • DMSO When DMSO is used, it contains a concentration of 5% to 20%, preferably a concentration of 5% to 10%, more preferably a concentration of 10%.
  • additives described in WO2007 / 058308 may be included.
  • cryopreservation solution for example, cryopreservation provided by Bioverde, Nippon Genetics, Reprocell, Xenoac, Cosmo Bio, Kojin Bio, Thermo Fisher Scientific, etc.
  • a liquid may be used.
  • the cooling rate may be appropriately controlled using a program freezer.
  • the cooling rate may be appropriately selected depending on the components of the cryopreservation solution, and may be performed according to the manufacturer's instructions for the cryopreservation solution.
  • the upper limit of the storage period is not particularly limited as long as the cells cryopreserved under the above conditions are thawed and retain the same properties as before freezing, for example, 1 week or more, 2 weeks or more, 3 weeks or more, 4 weeks or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, 6 months or more, 1 year or more, or more. Since cell damage can be suppressed by storing at a lower temperature, it may be transferred to a gas phase on liquid nitrogen (from about ⁇ 150 ° C. to ⁇ 180 ° C.). When storing in a gas phase on liquid nitrogen, it can be performed using a storage container well known to those skilled in the art. Although not particularly limited, for example, when storing for 2 weeks or more, it is preferable to store in a gas phase on liquid nitrogen.
  • the thawed (A) cells may be appropriately cultured before the next cryopreservation.
  • the mesenchymal stem cells are cultured using the above-described medium capable of culturing mesenchymal stem cells, and are not particularly limited, but contain CO 2 at a culture temperature of about 30 to 40 ° C., preferably about 37 ° C. It may be performed in an air atmosphere.
  • the CO 2 concentration is about 2-5%, preferably about 5%.
  • the culture after reaching the appropriate confluency for the culture container (for example, cells may occupy 50% to 80% of the culture container), the cells are detached with a release agent such as trypsin.
  • the culture may be continued by seeding in a separately prepared culture vessel at an appropriate cell density.
  • typical cell densities are 100 cells / cm 2 to 100,000 cells / cm 2 , 500 cells / cm 2 to 50,000 cells / cm 2 , 1,000 to 10,000 cells. / Cm 2 , 2,000 to 10,000 cells / cm 2, etc. are exemplified. In a particular embodiment, the cell density is between 2,000 and 10,000 cells / cm 2 . It is preferable to adjust the period until reaching appropriate confluency from 3 days to 7 days. During culture, the medium may be changed as necessary.
  • Thawing of cryopreserved cells can be performed by methods well known to those skilled in the art. For example, the method performed by standing or shaking in a 37 degreeC thermostat or a hot water bath is illustrated.
  • the cell (A) contained in the cell pharmaceutical composition of the present invention may be a cell in any state, for example, a cell recovered by detaching a cell in culture, or frozen in a cryopreservation solution It may be a cell in a state of being formed.
  • Use of the same lot of cells obtained by expanding and culturing in the same lot is preferable in that the same action and effect can be stably obtained and the handling property is excellent.
  • the (A) cells in the cryopreservation state may be thawed immediately before use and directly mixed with the cell suspension solution (B) described later while being suspended in the cryopreservation solution.
  • the cryopreservation solution may be removed by a method such as centrifugation and then suspended in the (B) cell suspension solution.
  • the dose (dosage) of (A) cells of the present invention may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.), dosage form, etc., but from the standpoint of sufficient therapeutic effect, the amount From the viewpoint of suppressing the occurrence of side effects, a smaller amount tends to be preferable.
  • the number of cells is 1 ⁇ 10 3 to 1 ⁇ 10 12 cells / time, preferably 1 ⁇ 10 4 to 1 ⁇ 10 11 cells / time, more preferably 1 ⁇ 10 5 to 1 ⁇ 10 10 cells / time, particularly preferably 5 ⁇ 10 5. 6 to 1 ⁇ 10 9 pieces / time.
  • this dose may be administered once as a single dose, or the dose may be divided into multiple doses.
  • the dose (dosage) of the cell (A) of the present invention may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.) and the dosage form of the composition of the present invention, but is usually administered to an adult.
  • the number of cells is 1 ⁇ 10 to 5 ⁇ 10 10 cells / kg, preferably 1 ⁇ 10 2 to 5 ⁇ 10 9 cells / kg, more preferably 1 ⁇ 10 3 to 5 ⁇ 10 8 cells / kg, particularly preferably 1 ⁇ 10 4 to 5 ⁇ 10 7 cells / kg. It is.
  • this dose may be administered once as a single dose, or the dose may be divided into multiple doses.
  • the (B) cell suspension solution of the present invention is an isotonic electrolyte solution containing Na + , Cl ⁇ , K + and Ca 2+ as electrolytes and not containing carbohydrates.
  • the cell suspension solution may further contain at least one electrolyte selected from the group consisting of CH 3 COO ⁇ , HCO 3 ⁇ , citrate ions and lactate ions.
  • the solution for cell suspension can be said to be an isotonic electrolyte infusion preparation having an extracellular fluid replacement effect in a living body, and is specifically selected from the group consisting of Ringer's solution, Ringer's acetate solution, Ringer's bicarbonate bicarbonate solution, and Ringer's lactate solution At least one of them is preferred.
  • (B) cell suspension solution having such a composition in the cell pharmaceutical composition of the present invention, (A) the state of the cells is kept good, and the survival rate is high over a long period of time. Can be maintained.
  • the cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Cl ⁇ as electrolytes, Na + is 140 mEq / L to 160 mEq / L, and K + is 1 mEq / L.
  • a Ringer solution having L to 10 mEq / L, Ca 2+ of 1 mEq / L to 10 mEq / L, and Cl ⁇ of 140 mEq / L to 170 mEq / L can be mentioned as a preferable example.
  • the above Ringer solution can be prepared with each electrolyte in the above concentration range, such as Ringer solution “Otsuka” (Nippon Ringer solution, Otsuka Pharmaceutical Factory), Ringer solution “Fuso” (Nippon Ringer solution, Fuso Pharmaceutical Co., Ltd.), etc. Commercial products can also be used.
  • the cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Cl ⁇ , CH 3 COO ⁇ as electrolytes, and Na + is 120 mEq / L to 140 mEq / L
  • the above-mentioned Ringer's solution can be prepared by adjusting the concentration of each electrolyte to the above-mentioned concentration range. Commercial products such as F infusion (Terumo Corporation) can also be used.
  • the cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Mg 2+ , Cl ⁇ , HCO 3 ⁇ , citrate ions (Citrate 3+ ) as electrolytes,
  • the electrolyte concentrations of Na + are 120 mEq / L to 145 mEq / L
  • K + is 1 mEq / L to 10 mEq / L
  • Ca 2+ is 1 mEq / L to 10 mEq / L
  • Mg 2+ is 0.1 mEq / L to 10 mEq / L.
  • the bicarbonate Ringer's solution can be prepared with each electrolyte in the above-mentioned concentration range.
  • commercially available products such as Bicanate (registered trademark) infusion (Otsuka Pharmaceutical Factory), Bicarbon (registered trademark) infusion (Ai Pharma Co., Ltd.), etc. Can also be used.
  • the (B) a solution for cell suspension, free of carbohydrates, and Na +, K +, Ca 2+ as the electrolyte, Cl -, lactate (Lactate -) include respective electrolyte concentration, Na + is 120 mEq / L to 140 mEq / L, K + 1 mEq / L to 10 mEq / L, Ca 2+ 1 mEq / L to 10 mEq / L, Cl ⁇ 90 mEq / L to 130 mEq / L, Lactate ⁇ 15 mEq / L to 45 mEq / L
  • a preferred example is Lactated Ringer's solution which is L.
  • the above Lactated Ringer solution can be prepared with each electrolyte in the above-mentioned concentration range.
  • Kriticare Co., Ltd. Hartmann Infusion "NP” (Nipro Corporation), Hartmann Infusion pH 8 "NP” (Nipro Corporation), Lactec (registered trademark) Note (Otsuka Pharmaceutical Factory), Lactolin Gel Solution “Fuso” (Fuso Pharmaceutical) Commercial products such as Kogyo Co., Ltd. can also be used.
  • the cell pharmaceutical composition of the present invention may contain one or more other drugs having a therapeutic effect on a disease.
  • Other drugs that can be used as liver disease drugs, heart disease drugs, inflammatory bowel disease drugs, respiratory drugs, nervous system drugs, cardiovascular drugs, cerebral circulation improving drugs, immunosuppressive drugs Of these drugs.
  • liver diseases examples include hepatitis B therapeutic agents (lamivudine, adefovir, entecavir, tenofovir, etc.), interferon preparations (interferon ⁇ , interferon ⁇ -2b, interferon ⁇ , peginterferon ⁇ -2a, peginterferon ⁇ -2b, etc.
  • hepatitis C drugs ribavirin, terapyrevir, simeprevir, vaniprevir, daclatasvir, asunaprevir, sofosbuvir, etc.
  • corticosteroids prednisolone, methylprednisolone sodium succinate, etc.
  • anticoagulants dry concentrated human antithrombin III
  • antidote calcium edetate disodium hydrate, glutathione, dimethicaprol, sodium thiosulfate hydrate, Sugamasekuna Human serum albumin, liver extract, ursodeoxycholic acid, glycyrrhizic acid, azathioprine, bezafibrate, amino acids (glycine, L-cysteine, L-isoleucine, L-leucine, L-valine, L-threonine, L-serine,
  • therapeutic agents for heart diseases include ACE inhibitors, angiotensin II receptor antagonists, ⁇ -blockers, antiplatelet agents, warfarin, calcium antagonists, nitrates, diuretics, HMG-CoA reductase inhibitors, ancalons, etc. Is mentioned.
  • Examples of the therapeutic agent for inflammatory bowel disease include salazosulfapyridine and mesalazine.
  • Examples of the respiratory medicine include dimorpholine, doxapram hydrochloride hydrate, cyberestat sodium hydrate, pirfenidone, pulmonary surfactant, and Dornase alpha.
  • Examples of the nervous system drug include edaravone, interferon beta-1a, interferon beta-1b, fingolimod hydrochloride, riluzole, tartilelin hydrate and the like.
  • circulatory drugs examples include hepronicart, midodrine hydrochloride, amedinium methylmethyl sulfate, ethylephrine hydrochloride, phenylephrine hydrochloride, and the like.
  • cerebral circulation improving drug examples include ifenprodil tartrate, nicergoline, ipdilast, dihydroergotoxin mesylate, nizophenone fumarate, fasudil hydrochloride hydrate and the like.
  • immunosuppressant examples include cyclosporine, azathioprine, mizoribine, basiliximab, tacrolimus hydrate, gusperimus hydrochloride, mycophenolate mofetil, everolimus and the like.
  • the cell pharmaceutical composition of the present invention contains the above-mentioned other drug, at the time of storage, it may be stored in a container different from (A) the cell and (B) the cell suspension solution. It may be contained in a form blended with. Depending on the type of disease, treatment method, patient condition, etc., other drugs and (A) cells and (B) cell suspension solutions may be administered simultaneously, or at regular intervals. Also good.
  • the cell pharmaceutical composition of the present invention is within a range not impairing the effects of the present invention, in addition to the above (A) cell and (B) cell suspension solution, according to its use and form, according to a conventional method, Other components such as pharmaceutically acceptable carriers and additives may be included.
  • Such carriers and additives may be contained in the (B) cell suspension solution, or may be contained separately from the (B) cell suspension solution.
  • Examples of such carriers and additives include isotonic agents, thickeners, sugars, sugar alcohols, preservatives (preservatives), bactericides or antibacterial agents, pH regulators, stabilizers, chelating agents.
  • Oily base Oily base, gel base, surfactant, suspending agent, binder, excipient, lubricant, disintegrant, foaming agent, fluidizer, dispersant, emulsifier, buffer, solubilizer , Antioxidants, sweeteners, sour agents, colorants, flavoring agents, fragrances or refreshing agents, but are not limited thereto.
  • the cell pharmaceutical composition of the present invention can be used in various forms depending on the purpose, for example, injections (including infusions, implantable injections, continuous injections, injections prepared at the time of use), dialysis agents, patches. It can be used in the form of a poultice.
  • the cell pharmaceutical composition of the present invention can also be applied to the affected area by spraying, and the cell pharmaceutical composition of the present invention can also be used in the form of gelation or sheeting at the affected area after spraying.
  • the cell pharmaceutical composition of the present invention can also be applied to the affected area after the (A) cell is made into a sheet or a three-dimensional structure.
  • (A) cells, (B) cell suspension solutions, other drugs, and other components are sealed and stored in separate containers, and are used by mixing them at the time of use. Also good.
  • the above (A) cells, (B) cell suspension solution, other drugs, and other components may be stored under conditions suitable for each, for example, freezing conditions, refrigerated conditions, room temperature Any of conditions etc. may be sufficient.
  • the pH of the cell pharmaceutical composition of the present invention is not particularly limited as long as it is within a pharmaceutically, pharmacologically (pharmaceutical) or physiologically acceptable range.
  • the range is 9.0, preferably 2.5 to 8.5, and more preferably 4.0 to 8.0.
  • the osmotic pressure of the cell pharmaceutical composition of the present invention is not particularly limited as long as it is within the range acceptable to the living body.
  • An example of the osmotic pressure ratio of the cell pharmaceutical composition of the present invention is preferably in the range of 0.6 to 1.5, more preferably 0.7 to 1.2, and still more preferably 0.8 to 1.0. It is done.
  • the osmotic pressure can be adjusted by a method known in the technical field using the above-described electrolyte or the like.
  • the osmotic pressure ratio is the ratio of the osmotic pressure of the sample to the osmotic pressure of 286 mOsm (0.9 w / v% sodium chloride aqueous solution) based on the 15th revised Japanese Pharmacopoeia. Measure by referring to the freezing point method.
  • the standard solution for osmotic pressure ratio measurement (0.9 w / v% sodium chloride aqueous solution) was prepared by drying sodium chloride (Japanese Pharmacopoeia standard reagent) at 500 to 650 ° C. for 40 to 50 minutes, and then in a desiccator (silica gel).
  • the mixture is allowed to cool and 0.900 g is accurately weighed and dissolved in purified water to make exactly 100 mL, or a commercially available standard solution for osmotic pressure ratio measurement (0.9 w / v% sodium chloride aqueous solution) is used.
  • the concentration of (A) cells in the cell pharmaceutical composition of the present invention is the cell type, cell suspension although it may vary depending on the solution used, it is usually 1 ⁇ 10 2 to 2.5 ⁇ 10 8 cells / mL, preferably 1 ⁇ 10 3 to 2.5 ⁇ 10 7 cells / mL, more preferably 1 ⁇ 10 4 to 2.5 ⁇ 10 6 cells / mL.
  • human mesenchymal stem cells are contained in a cell suspension solution at a density of 1 ⁇ 10 4 to 2.5 ⁇ 10 6 cells / mL,
  • the turbid solution contains 125 to 150 mEq / L sodium ion, 100 to 160 mEq / L chloride ion, 3 to 5 mEq / L potassium ion and 2 to 6 mEq / L calcium ion, and has an osmotic pressure ratio of 0 to physiological saline.
  • a pH of 4.0 to 8.0 is a cell suspension solution at a density of 1 ⁇ 10 4 to 2.5 ⁇ 10 6 cells / mL.
  • the administration route to the subject of the cell pharmaceutical composition of the present invention includes subcutaneous administration, intramuscular administration, intravenous administration, intraarterial administration, intrathecal administration, intraperitoneal administration, rectal administration, vaginal administration, and transdermal administration.
  • implant, intraarterial administration, intravenous administration, and direct administration to an organ are more preferable. Are intravenous and direct administration to the organ.
  • the administration rate of the cell pharmaceutical composition of the present invention to the subject may vary depending on the patient's condition (weight, age, symptoms, physical condition, etc.) and the administration route of the non-alcoholic steatohepatitis therapeutic agent of the present invention.
  • it When administered to an adult, it is 50 mL / h to 1,000 mL / h, preferably 75 mL / h to 500 mL / h, and more preferably 100 mL / h to 250 mL / h.
  • the administration temperature of the cell pharmaceutical composition of the present invention to the subject may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.), the administration route of the cell pharmaceutical composition of the present invention, etc. ⁇ 45 ° C., preferably 15 ° C. to 37 ° C., more preferably room temperature to 37 ° C.
  • the cell pharmaceutical composition of the present invention can be administered to a subject using an infusion set.
  • an infusion set a wand disposable infusion tube set (manufactured by Yoshida Seisakusho Co., Ltd.), an infusion set (manufactured by Fortegro Medical Co., Ltd.), a terfusion (registered trademark) infusion set (manufactured by Terumo Corporation), a JMS infusion Set (manufactured by JMS Co., Ltd.), Sure plug infusion set (manufactured by Terumo Corporation), Infusion set (manufactured by Nipro Corporation), Top infusion set NP (manufactured by Top Co., Ltd.), Infusion set with filter (EX type ) (Manufactured by Toray Medical Co., Ltd.) can also be used.
  • EX type Manufactured by Toray Medical Co., Ltd.
  • the cell pharmaceutical composition of the present invention can be administered to a subject using an infusion tube.
  • an infusion tube Lectro Cass (manufactured by Samick International Co., Ltd.), JMS extension tube (manufactured by JMS Co., Ltd.), Saffy extension tube (manufactured by Terumo Corporation), extension tube (manufactured by Co., Ltd.) Top product), Connecting tube (Chuatsu) (Medicos Hirata Co., Ltd.), SAFTY AP tube (manufactured by Kawasumi Chemical Industry Co., Ltd.), Bioconnector 2 with extension tube (Toray Medical Co., Ltd.), Medicut Extension Tube Set B (Nippon Sherwood) Commercially available products such as Wand Disposable Infusion Tube Set (manufactured by Yoshida Seisakusho Co., Ltd.), Infusion Tube (manufactured by Forte Grow Medical Co., Ltd.), and the like can also be used.
  • the material of the infusion tube used when administering the cell pharmaceutical composition of the present invention to a subject includes polyvinyl chloride, thermoplastic elastomer, TPE thermoplastic elastomer, silicone, silicone rubber, polyethylene, polybutadiene, Teflon (registered trademark), Polyurethane, polypropylene, natural rubber, polyolefin, PVC (plasticizer: TOTM, DOA), plasticizer PVC, and mixtures thereof can be used.
  • the cell pharmaceutical composition of the present invention can be suitably used for the treatment of various diseases.
  • visceral diseases specifically heart disease, stomach / duodenal disease, small intestine / colon disease, liver disease, biliary disease, pancreatic disease, kidney disease, lung disease, mediastinal disease, diaphragm disease, pleural disease, peritoneal disease
  • CNS central nervous system
  • peripheral arterial diseases peripheral venous diseases.
  • Specific diseases include, for example, autoimmune hepatitis, fulminant hepatitis, chronic hepatitis, viral hepatitis, alcoholic hepatitis, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (Nonalcoholic steatohepatitis (NASH)), non-alcoholic fatty liver (NAFL), liver fibrosis, cirrhosis, liver cancer, fatty liver, drug allergic liver disorder, hemochromatosis, hemosiderosis, Wilson's disease, primary Hepatic diseases such as biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), biliary atresia, liver abscess, chronic active hepatitis, chronic persistent hepatitis; myocardial infarction, heart failure, arrhythmia, palpitations, cardiomyopathy, false blood Cardiomyopathy, angina, congenital heart disease, valvular heart disease, myocarditis,
  • biliary tract diseases such as acute cholecystitis, acute cholangitis, chronic cholecystitis, cholangiocarcinoma, gallbladder cancer
  • pancreatic diseases such as acute pancreatitis, chronic pancreatitis, pancreatic cancer
  • acute nephritis, chronic nephritis, acute renal failure chronic Renal diseases such as renal failure
  • Diaphragmatic diseases such as diaphragmatic hernia
  • Pleural diseases such as pleurisy, pyothorax, pleural tumor, cancerous pleurisy, pleural mesothelioma
  • Peritoneal diseases such as peritonitis, peritoneal tumor
  • Aseptic meningitis Guillain-Barre syndrome, amyotrophic lateral sclerosis (ALS), myasthenia gravis, mononeuropathy, polyneuropathy, spinal muscular atrophy, spinal disorder, acute transverse myelitis, spinal cord infarction (false) Hematologic spinal cord disorder), intracranial tumor, spine
  • Neurological diseases such as tumors; CNS disorders such as Alzheimer's disease, cognitive impairment, stroke, multiple sclerosis, Parkinson's disease; fibromyalplasia, peripheral arterial disease (PAD), obstructive thromboangitis (Burger's disease), Kawasaki Peripheral arterial disease such as KD disease; deep vein
  • liver diseases, heart diseases, lung diseases, neurological diseases, peripheral arterial diseases, immunodeficiency diseases that have been confirmed to have sufficient therapeutic effects by mesenchymal stem cells are preferred, among which liver fibrosis, Suitable for the treatment of cirrhosis, myocardial infarction, heart failure, pulmonary fibrosis, interstitial pneumonia, childhood cerebral palsy, amyotrophic lateral sclerosis (ALS), peripheral arterial disease (PAD), graft-versus-host disease (GVHD) And can be used more suitably for liver fibrosis, cirrhosis, myocardial infarction, heart failure, pulmonary fibrosis, and interstitial pneumonia. Moreover, it can use suitably for the cancer of each structure
  • the present invention includes (A) a cell and (B) a cell suspension solution, (B) the cell suspension solution contains Na + , Cl ⁇ , K + and Ca 2+ as an electrolyte, and a sugar Also included is a disease treatment kit that is an isotonic electrolyte without quality.
  • the disease treatment kit of the present invention is a kit containing the above-described cell pharmaceutical composition of the present invention, and may contain (A) cells, (B) cell suspension solutions, and other cell pharmaceutical compositions of the present invention. The description in the section of the cell pharmaceutical composition can be applied to the components. According to the disease treatment kit of the present invention, since the state of cells can be kept good and the survival rate can be maintained high over a long period of time, an excellent therapeutic effect can be obtained for various diseases. .
  • the disease treatment kit of the present invention can also be expressed as comprising the cell pharmaceutical composition, container and label of the present invention.
  • Suitable containers included in the disease treatment kit of the present invention are not particularly limited, and examples include cryotubes for freezing cells, bottles for cell suspension solutions, vials, test tubes, and dialysis bags. These containers may be formed from a variety of materials such as glass, metal, plastic, or combinations thereof.
  • the contents on the labels on these containers describe the contents of the cells, the solution for cell suspension, and the like.
  • the disease treatment kit of the present invention includes other additives, other drugs, diluents, filters, needles, syringes, and other packages that are desirable from a commercial and user standpoint, including package inserts that describe usage. Material can be included.
  • ⁇ Cell suspension solution> Also within the scope of the present invention is a solution for cell suspension for a cell pharmaceutical composition, which is an isotonic electrolyte containing Na + , Cl ⁇ , K + and Ca 2+ as an electrolyte and free of carbohydrates. is there.
  • an isotonic electrolyte solution containing Na + , Cl ⁇ , K + and Ca 2+ and not containing carbohydrates As a cell suspension solution for cell medicine injection / infusion, It is a finding newly found by the present inventors that the survival rate can be maintained high over a long period of time.
  • the description of (B) the cell suspension solution in the section of the cell pharmaceutical composition can be applied.
  • the present invention relates to a method for treating a disease in which (A) cells are suspended in (B) a cell suspension solution and administered to a patient, and (B) the cell suspension solution contains Na + , Cl as an electrolyte.
  • the cell suspension solution contains Na + , Cl as an electrolyte.
  • - includes a K + and Ca 2+, and does not contain carbohydrates, including methods of treatment wherein the isotonic electrolyte solution.
  • the disease method of the present invention is a treatment method using the above-described cell pharmaceutical composition of the present invention, and may contain (A) cells, (B) a cell suspension solution, and other cell pharmaceutical compositions of the present invention.
  • the description in the section of the cell pharmaceutical composition can be applied to the components.
  • Example 1 Preparation of adipose-derived mesenchymal stem cells
  • the subcutaneous adipose tissue obtained by the liposuction method was washed with physiological saline.
  • collagenase (Roche diagnostics) (solvent was physiological saline) was added, shaken at 37 ° C. for 90 minutes, and dispersed. Subsequently, the suspension was centrifuged at 800 g for 5 minutes to obtain a precipitate of stromal vascular cell groups.
  • a serum-free medium for mesenchymal stem cells (Rohto) is added to the cell precipitate, the cell suspension is centrifuged at 400 g for 5 minutes, and after removal of the supernatant, a serum-free medium for mesenchymal stem cells (Rohto)
  • the cells were seeded in a flask. Cells were cultured at 37 ° C. for several days in 5% CO 2 . Several days later, the culture was washed with PBS to remove residual blood cells and adipose tissue contained in the culture solution, and mesenchymal stem cells adhered to a plastic container were obtained.
  • adipose-derived mesenchymal stem cells were dispensed into a centrifuge tube and centrifuged at 400 g for 5 minutes to obtain cell precipitates. After removing the supernatant, an appropriate amount of a cell cryopreservation solution (STEM-CELLBANKER (Zenoac)) was added and suspended. The cell suspension was dispensed into a cryotube, stored at ⁇ 80 ° C. in a freezer, transferred to a gas phase on liquid nitrogen, and storage was continued.
  • a cell cryopreservation solution STEM-CELLBANKER (Zenoac)
  • Adipose tissue-derived mesenchymal stem cells were rapidly thawed in a hot water bath (37 ⁇ 1 ° C.), and then 62.5 ⁇ L of a cell suspension of adipose tissue-derived mesenchymal stem cells was added. Suspended by inversion and stored at room temperature. Immediately after preparation, 1 mL was taken from the middle layer with a 5 mL syringe with a 21 G injection needle immediately after preparation, 2 hours, 4 hours, and 7 hours, and transferred to a 1.5 mL tube. .
  • Example 2 In a 15 mL centrifuge tube (Sumitomo Bakelite Co., Ltd., product number: MS-56150), Ringer's solution (Ringer's solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73), bicarbonate Ringer's solution (Bikanate (registered trademark) infusion, Otsuka Pharmaceutical Co., Ltd.) After dispensing 5 mL each of the plant), in the same manner as in Example 1, the fat-derived mesenchymal stem cells that had been cryopreserved were rapidly thawed in a hot water bath (37 ⁇ 1 ° C.) and then the cell suspension of the fat-derived mesenchymal stem cells was The suspension was added in 62.5 ⁇ L aliquots.
  • Example 3 The fat-derived mesenchymal stem cells of Example 1 were replaced with bone marrow-derived mesenchymal stem cells (Lonza), and the same test was performed using Ringer's solution (Ringer solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73). The cell viability was calculated by measuring the number of living cells and the number of dead cells immediately after preparation, 2 hours later, 3 hours later and 7 hours later. The results are shown in Table 5 below.
  • Example 4 The fat-derived mesenchymal stem cells of Example 1 were replaced with umbilical cord-derived mesenchymal stem cells (Lifeline Cell Technology, LifeLine (registered trademark) _UCMSC, Lot. 160907), Ringer's solution (Ringer solution “Otsuka”, Otsuka Pharmaceutical Factory, Ltd., Lot : K4K73), the number of viable cells and the number of dead cells were measured immediately after preparation, 2 hours, 4 hours and 6 hours, and the cell viability was calculated in the same manner as in Example 1. . The results are shown in Table 6 below.
  • Example 5 The fat-derived mesenchymal stem cells of Example 1 were replaced with peripheral blood mononuclear cells (ACCUCELL (registered trademark) normal donor-derived PBMC, Precision Bioservices (PRECISION FOR MEDICINE), Lot. 13134-10), Ringer's solution ( The same test was conducted using Ringer's solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73), and the number of living cells and dead cells were measured immediately after preparation, 2 hours, 4 hours and 6 hours later. Cell viability was calculated. The results are shown in Table 7 below.
  • the cell pharmaceutical composition of the present invention can maintain a good cell state and maintain its survival rate in a high state for a long time, it can be expected to have an excellent therapeutic effect on various diseases.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Reproductive Health (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Dispersion Chemistry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention addresses the issue of providing a cell pharmaceutical composition capable of maintaining a cell state well and a high survival rate over the long term. This cell pharmaceutical composition contains cells (A) and a cell suspension solution (B). The cell suspension solution (B) is an electrolytic solution that includes Na+, Cl, K+, and Ca2+ as electrolytes, does not include carbohydrates, and is isotonic. The cell suspension solution (B) may further include at least one type of electrolyte selected from the group consisting of CH3COO, HCO3 , citrate ions, and lactate ions.

Description

細胞医薬組成物、疾患治療用キット及び細胞懸濁用溶液Cell pharmaceutical composition, disease treatment kit and cell suspension solution
 本発明は、細胞医薬組成物、疾患治療用キット及び細胞懸濁用溶液に関する。 The present invention relates to a cell pharmaceutical composition, a disease treatment kit and a cell suspension solution.
 細胞を含む医薬品を疾患治療に用いるための技術は年々進歩している。特にiPS細胞、造血幹細胞、間葉系幹細胞等の幹細胞、皮膚細胞、心筋細胞等については、基礎的な研究段階から開発段階へと移行して、現在では、実際に臨床の場で用いられているものも存在する。細胞による疾患の治療においては、細胞自体が有する機能を直接的又は間接的に疾患治療のために用いること、ダメージを受けた患者の細胞や組織の機能を、幹細胞から新たに分化させた細胞や臓器により補うこと等が期待されている。 ・ Technology for using drugs containing cells for disease treatment has been improving year by year. In particular, stem cells such as iPS cells, hematopoietic stem cells, mesenchymal stem cells, skin cells, cardiomyocytes, etc. have moved from the basic research stage to the development stage, and are currently used in clinical settings. There are also. In the treatment of diseases by cells, the functions of cells themselves are used directly or indirectly for disease treatment, and the functions of cells and tissues of damaged patients are newly differentiated from stem cells and It is expected to be supplemented by organs.
 例えば、間葉系幹細胞は、Friedenstein(1982)によって初めて骨髄から単離された多分化能を有する前駆細胞である(非特許文献1)。この間葉系幹細胞は、骨髄、臍帯、脂肪等の様々な組織に存在することが明らかにされており、間葉系幹細胞移植は、様々な難治性疾患に対する新しい治療方法として、期待されている(特許文献1~4)。最近では、脂肪組織、胎盤、臍帯、卵膜等の胎児付属物の間質細胞に同等の機能を有する細胞が存在することが知られている。従って、間葉系幹細胞を間質細胞(Mesenchymal Stromal Cell)と称することもある。 For example, mesenchymal stem cells are pluripotent progenitor cells isolated from bone marrow for the first time by Friedenstein (1982) (Non-patent Document 1). These mesenchymal stem cells have been revealed to exist in various tissues such as bone marrow, umbilical cord, and fat, and mesenchymal stem cell transplantation is expected as a new treatment method for various intractable diseases ( Patent Documents 1 to 4). Recently, it is known that cells having an equivalent function exist in stromal cells of fetal appendages such as adipose tissue, placenta, umbilical cord, and egg membrane. Therefore, the mesenchymal stem cell may be referred to as a stromal cell (Mesenchymal Stroma Cell).
特表2002-506831号公報JP 2002-506831 A 特表2000-508911号公報Special Table 2000-508911 特開2012-157263号公報JP 2012-157263 A 特表2012-508733号公報Special table 2012-508733 gazette
 間葉系幹細胞等の細胞を含む医薬品においては、安全性を確保する、細胞の投与を容易にすること等を目的として、細胞を溶液に懸濁して用いることがある。間葉系幹細胞等の細胞を懸濁液として、生体内へ点滴、注射等の方法によって移入・注入する場合において、懸濁液中の細胞の生存率が徐々に低下して十分な薬理作用が得られない、細胞同士が凝集してカニューレ中に詰まる、患者の肺静脈等に塞栓を形成する等の不都合が生じることが懸念されている。そこで、本発明は、細胞の生存率を長時間に渡って高く維持することができる細胞医薬組成物を提供することを課題とする。 In pharmaceuticals containing cells such as mesenchymal stem cells, the cells may be suspended in a solution for the purpose of ensuring safety and facilitating administration of the cells. When cells such as mesenchymal stem cells are transferred into a living body by infusion or injection as a suspension, the survival rate of the cells in the suspension is gradually reduced and sufficient pharmacological action is achieved. There are concerns that problems such as formation of emboli in the patient's pulmonary veins and the like that may not be obtained, cells may aggregate and clog in the cannula, and the like. Then, this invention makes it a subject to provide the cell pharmaceutical composition which can maintain the survival rate of a cell high over a long time.
 上記課題を解決するために鋭意研究した結果、本発明者らは、疾患治療用の細胞をリンゲル液、酢酸リンゲル液又は重炭酸リンゲル液といった、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液に懸濁して点滴注入等を行うと、投与中の細胞の生存率低下が顕著に抑制されることを見出し、本発明に到達した。本発明によれば、細胞を含む医薬組成物が、細胞の状態を良好に保ち、生存率を長時間に渡って高く維持することができるため、様々な疾患に対して優れた治療効果を奏することができる。すなわち本発明の要旨は、以下の通りである。 As a result of diligent research to solve the above problems, the inventors of the present invention include Na + , Cl , K + and Ca 2+ as electrolytes such as Ringer's solution, Ringer's acetate solution or Ringer's bicarbonate solution, as cells for disease treatment. In addition, the present inventors have found that when a drip infusion or the like is performed after suspending in an isotonic electrolyte solution that does not contain carbohydrates, a decrease in the survival rate of cells during administration is remarkably suppressed. According to the present invention, since a pharmaceutical composition containing cells can maintain a good state of cells and maintain a high survival rate over a long period of time, it has excellent therapeutic effects on various diseases. be able to. That is, the gist of the present invention is as follows.
[1](A)細胞、及び
 (B)細胞懸濁用溶液
を含有し、
 (B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、細胞医薬組成物。
[2](B)細胞懸濁用溶液が、CHCOO、HCO 、クエン酸イオン及び乳酸イオンからなる群より選択される少なくとも1種の電解質をさらに含む、[1]に記載の細胞医薬組成物。
[3](B)細胞懸濁用溶液が、リンゲル液、酢酸リンゲル液、重炭酸リンゲル液及び乳酸リンゲル液からなる群より選択される少なくとも1種である、[1]又は[2]に記載の細胞医薬組成物。
[4](A)細胞が、間葉系幹細胞又は末梢血単核球である、[1]から[3]のいずれかに記載の細胞医薬組成物。
[5]上記間葉系幹細胞が、脂肪由来、臍帯由来又は骨髄由来である、[4]に記載の細胞医薬組成物。
[6](A)細胞、及び
 (B)細胞懸濁用溶液
を含有し、
 (B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、疾患治療用キット。
[7]電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、細胞医薬組成物用の細胞懸濁用溶液。
[1] containing (A) cells, and (B) a cell suspension solution,
(B) The cell pharmaceutical composition, wherein the cell suspension solution is an isotonic electrolytic solution containing Na + , Cl , K + and Ca 2+ as electrolytes and not containing carbohydrates.
[2] The solution for cell suspension according to [1], wherein the solution for cell suspension further comprises at least one electrolyte selected from the group consisting of CH 3 COO , HCO 3 , citrate ions and lactate ions. Cell pharmaceutical composition.
[3] The cell pharmaceutical composition according to [1] or [2], wherein (B) the cell suspension solution is at least one selected from the group consisting of Ringer's solution, acetated Ringer's solution, bicarbonated Ringer's solution, and lactated Ringer's solution. object.
[4] The cell pharmaceutical composition according to any one of [1] to [3], wherein (A) the cells are mesenchymal stem cells or peripheral blood mononuclear cells.
[5] The cell pharmaceutical composition according to [4], wherein the mesenchymal stem cells are derived from fat, umbilical cord or bone marrow.
[6] containing (A) cells, and (B) a cell suspension solution,
(B) The disease treatment kit, wherein the cell suspension solution is an isotonic electrolyte solution containing Na + , Cl , K + and Ca 2+ as electrolytes and not containing carbohydrates.
[7] A cell suspension solution for a cell pharmaceutical composition, which is an isotonic electrolyte solution containing Na + , Cl , K + and Ca 2+ as an electrolyte and free of carbohydrates.
 本発明の細胞医薬組成物は、細胞の状態を良好に保ち、その生存率を長時間に渡って高い状態で維持することができるため、様々な疾患に対して優れた治療効果が期待できる。 Since the cell pharmaceutical composition of the present invention can maintain a good cell state and maintain its survival rate in a high state for a long time, it can be expected to have an excellent therapeutic effect on various diseases.
 本発明の細胞医薬組成物、疾患治療用キット、及び注射剤用の細胞懸濁用溶液について詳細に説明する。 The cell pharmaceutical composition, disease treatment kit, and cell suspension solution for injection of the present invention will be described in detail.
<細胞医薬組成物>
 本発明の細胞医薬組成物は、(A)細胞、及び(B)細胞懸濁用溶液を含有し、(B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液であることを特徴とする。なお、本発明において「細胞医薬組成物」とは、細胞を含有する医薬用の組成物であり、細胞が有する機能によって疾患に対する治療効果を奏するものをいう。本発明の細胞医薬組成物は、細胞を上記特定の溶液に懸濁することで、細胞の生存率を長時間に渡って高い状態で維持することができるため、様々な疾患に対して優れた治療効果を奏することが可能となる。本発明の細胞医薬組成物は、上記必須成分である(A)細胞及び(B)細胞懸濁用溶液に加えて、疾患に対する治療効果を有する他の薬剤を含有していてもよい。さらに、本発明の効果を損なわない限り、その他の成分を含有していてもよい。以下、本発明の細胞医薬組成物が含有する(A)細胞、(B)細胞懸濁用溶液、他の薬剤、その他の成分について詳細に説明する。
<Cellular pharmaceutical composition>
The cell pharmaceutical composition of the present invention contains (A) cells and (B) a cell suspension solution, and (B) the cell suspension solution contains Na + , Cl , K + and Ca 2+ as electrolytes. And an isotonic electrolyte solution that does not contain carbohydrates. In the present invention, the “cell pharmaceutical composition” refers to a pharmaceutical composition containing cells, which exhibits a therapeutic effect on diseases due to the functions of the cells. The cell pharmaceutical composition of the present invention is excellent for various diseases because the cell viability can be maintained in a high state for a long time by suspending the cells in the specific solution. A therapeutic effect can be achieved. In addition to the essential components (A) cells and (B) cell suspension solution, the cell pharmaceutical composition of the present invention may contain other drugs having a therapeutic effect on diseases. Furthermore, as long as the effects of the present invention are not impaired, other components may be contained. Hereinafter, (A) cells, (B) cell suspension solutions, other drugs, and other components contained in the cell pharmaceutical composition of the present invention will be described in detail.
[(A)細胞]
 本発明において(A)細胞とは、疾患の治療に対する効果を奏する細胞であれば特に限定されないが、例えば間葉系幹細胞、末梢血単核球(好中球、好酸球、好塩基球、リンパ球、単球等を含む)、赤血球、T細胞、NK細胞、NKT細胞、NKM細胞、LAK細胞、樹状細胞、繊維芽細胞、造血幹細胞、iPS細胞、ES細胞、骨髄細胞、心筋細胞、肝細胞、神経細胞、皮膚細胞、脂肪細胞、その他各組織を構成する細胞が挙げられる。これらのうち、後述する(B)細胞懸濁用溶液による生存率維持効果に優れるとい観点から、間葉系幹細胞、末梢血単核球、骨髄細胞が好ましい。
[(A) Cell]
In the present invention, the (A) cell is not particularly limited as long as it has an effect on the treatment of diseases. For example, mesenchymal stem cells, peripheral blood mononuclear cells (neutrophils, eosinophils, basophils, Lymphocytes, monocytes, etc.), red blood cells, T cells, NK cells, NKT cells, NKM cells, LAK cells, dendritic cells, fibroblasts, hematopoietic stem cells, iPS cells, ES cells, bone marrow cells, cardiomyocytes, Examples include hepatocytes, nerve cells, skin cells, fat cells, and other cells constituting each tissue. Among these, mesenchymal stem cells, peripheral blood mononuclear cells, and bone marrow cells are preferable from the viewpoint that (B) the cell suspension solution described later is excellent in viability maintenance effect.
(間葉系幹細胞)
 本発明において間葉系幹細胞とは、間葉系に属する一種以上、好ましくは二種以上、さらに好ましくは三種以上の細胞(骨細胞、心筋細胞、軟骨細胞、腱細胞、脂肪細胞など)への分化能を有し、当該能力を維持したまま増殖できる細胞を意味する。本発明において用いる間葉系幹細胞なる用語は、間質細胞と同じ細胞を意味し、両者を特に区別するものではない。また、単に間葉系細胞と表記される場合もある。間葉系幹細胞を含む組織としては、例えば、脂肪組織、臍帯、骨髄、臍帯血、子宮内膜、胎盤、羊膜、絨毛膜、脱落膜、真皮、骨格筋、骨膜、歯小嚢、歯根膜、歯髄、歯胚等が挙げられる。例えば脂肪組織由来間葉系幹細胞とは、脂肪組織に含有される間葉系幹細胞を意味し、脂肪由来間葉系幹細胞と称してもよい。これらのうち、各種疾患の治療に対する有効性の観点、入手容易性の観点等から、脂肪由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞、胎盤由来間葉系幹細胞、歯髄由来間葉系幹細胞が好ましく、脂肪由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞がより好ましい。
(Mesenchymal stem cells)
In the present invention, the mesenchymal stem cell refers to one or more cells belonging to the mesenchymal system, preferably two or more cells, more preferably three or more cells (bone cells, cardiomyocytes, chondrocytes, tendon cells, fat cells, etc.). It means a cell that has differentiation ability and can proliferate while maintaining the ability. The term mesenchymal stem cell used in the present invention means the same cell as the stromal cell, and does not particularly distinguish them. Moreover, it may be simply described as a mesenchymal cell. Examples of tissues containing mesenchymal stem cells include adipose tissue, umbilical cord, bone marrow, umbilical cord blood, endometrium, placenta, amniotic membrane, chorion, decidua, dermis, skeletal muscle, periosteum, dental follicle, periodontal ligament, Examples include dental pulp and tooth germ. For example, an adipose tissue-derived mesenchymal stem cell means a mesenchymal stem cell contained in an adipose tissue, and may be referred to as an adipose-derived mesenchymal stem cell. Among these, from the viewpoint of effectiveness for treatment of various diseases, from the viewpoint of availability, adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, placenta-derived mesenchymal stem cells, Dental pulp-derived mesenchymal stem cells are preferred, and adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and bone marrow-derived mesenchymal stem cells are more preferred.
 本発明における間葉系幹細胞は、処置される対象(被検体)と同種由来であってもよいし、異種由来であってもよい。本発明における間葉系幹細胞の種として、ヒト、ウマ、ウシ、ヒツジ、ブタ、イヌ、ネコ、ラビット、マウス、ラットが挙げられ、好ましくは処置される対象(被検体)と同種由来細胞である。本発明における間葉系幹細胞は、処置される対象(被検体)に由来、すなわち自家細胞(同種同系)であってもよいし、同種の別の対象に由来、すなわち他家細胞(同種異系)であってもよい。好ましくは他家細胞(同種異系)である。 The mesenchymal stem cells in the present invention may be derived from the same species as the subject to be treated (subject) or from a different species. Examples of the mesenchymal stem cell species in the present invention include humans, horses, cows, sheep, pigs, dogs, cats, rabbits, mice, and rats, preferably cells derived from the same species as the subject to be treated (subject). . The mesenchymal stem cells in the present invention may be derived from the subject (subject) to be treated, that is, autologous cells (allogeneic), or derived from another subject of the same species, ie, allogeneic cells (allogeneic). ). Preferred are allogeneic cells (allogeneic).
 間葉系幹細胞は同種異系の被験体に対しても拒絶反応を起こしにくいため、あらかじめ調製されたドナーの細胞を拡大培養して凍結保存したものを、本発明の細胞医薬組成物における(A)細胞としての間葉系幹細胞として使用することができる。そのため、自己の間葉系幹細胞を調製して用いる場合と比較して、商品化も容易であり、かつ安定して一定の効果を得られ易いという観点から、本発明における間葉系幹細胞は、同種異系であることがより好ましい。 Since mesenchymal stem cells are unlikely to cause rejection even for allogeneic subjects, cells prepared by pre-expanding donor cells that have been expanded and cryopreserved are used in the cell pharmaceutical composition of the present invention (A ) It can be used as a mesenchymal stem cell as a cell. Therefore, compared to the case of preparing and using autologous mesenchymal stem cells, commercialization is easy, and from the viewpoint that a certain effect can be easily obtained stably, the mesenchymal stem cells in the present invention, More preferably, it is allogeneic.
 本発明において間葉系幹細胞とは、間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%が間葉系幹細胞である。 In the present invention, the mesenchymal stem cell means an arbitrary cell population including the mesenchymal stem cell. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98 % Or 99% are mesenchymal stem cells.
 本発明において脂肪組織とは、脂肪細胞、及び微小血管細胞等を含む間質細胞を含有する組織を意味し、例えば、哺乳動物の皮下脂肪を外科的切除又は吸引して得られる組織である。脂肪組織は、皮下脂肪より入手され得る。後述する脂肪由来間葉系幹細胞の投与対象と同種動物から入手されることが好ましく、ヒトへ投与することを考慮すると、より好ましくは、ヒトの皮下脂肪である。皮下脂肪の供給個体は、生存していても死亡していてもよいが、本発明において用いる脂肪組織は、好ましくは、生存個体から採取された組織である。個体から採取する場合、脂肪吸引は、例えば、PAL(パワーアシスト)脂肪吸引、エルコーニアレーザー脂肪吸引、又は、ボディジェット脂肪吸引などが例示され、細胞の状態を維持するという観点から、超音波を用いないことが好ましい。 In the present invention, an adipose tissue means a tissue containing stromal cells including adipocytes and microvascular cells, and is, for example, a tissue obtained by surgical excision or aspiration of mammalian subcutaneous fat. Adipose tissue can be obtained from subcutaneous fat. It is preferably obtained from the same animal as the subject of administration of the adipose-derived mesenchymal stem cells described later, and more preferably human subcutaneous fat in consideration of administration to humans. An individual supplying subcutaneous fat may be alive or dead, but the adipose tissue used in the present invention is preferably a tissue collected from a living individual. In the case of collecting from an individual, liposuction is exemplified by PAL (power assist) liposuction, Erconia laser liposuction, or body jet liposuction. From the viewpoint of maintaining the state of cells, ultrasound is used. It is preferable not to use.
 本発明において臍帯とは、胎児と胎盤を結ぶ白い管状の組織であり、臍帯静脈、臍帯動脈、膠様組織(ウォートンジェリー;Wharton’s Jelly)、臍帯基質自体等から構成され、間葉系幹細胞を多く含む。臍帯は、本発明の細胞医薬組成物を使用する被験体(投与対象)と同種動物から入手されることが好ましく、本発明の細胞医薬組成物をヒトへ投与することを考慮すると、より好ましくは、ヒトの臍帯である。 In the present invention, the umbilical cord is a white tubular tissue that connects the fetus and the placenta, and is composed of umbilical vein, umbilical artery, collagenous tissue (Wharton's Jelly), umbilical matrix itself, etc., and mesenchymal stem cells Including many. The umbilical cord is preferably obtained from the same animal as the subject (administration subject) using the cell pharmaceutical composition of the present invention, and more preferably in consideration of administering the cell pharmaceutical composition of the present invention to a human. The human umbilical cord.
 本発明において骨髄とは、骨の内腔を満たしている柔組織のことをいい、造血器官である。骨髄中には骨髄液が存在し、その中に存在する細胞を骨髄細胞と呼ぶ。骨髄細胞には、赤血球、顆粒球、巨核球、リンパ球、脂肪細胞等の他、間葉系幹細胞、造血幹細胞、血管内皮前駆細胞等が含まれている。骨髄細胞は、例えば、ヒト腸骨、長管骨、又はその他の骨から採取することができる。 In the present invention, the bone marrow refers to a soft tissue filling the bone lumen, and is a hematopoietic organ. Bone marrow fluid is present in the bone marrow, and the cells present therein are called bone marrow cells. Bone marrow cells include erythrocytes, granulocytes, megakaryocytes, lymphocytes, adipocytes and the like, as well as mesenchymal stem cells, hematopoietic stem cells, vascular endothelial progenitor cells, and the like. Bone marrow cells can be collected, for example, from human iliac bone, long bone, or other bone.
 本発明において、脂肪由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞とは、それぞれ脂肪由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%が、脂肪由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞である。 In the present invention, each tissue-derived mesenchymal stem cell such as adipose-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell is a fat-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, It means any cell population containing mesenchymal stem cells derived from each tissue such as bone marrow-derived mesenchymal stem cells. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98 % Or 99% are mesenchymal stem cells derived from various tissues such as adipose-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, and bone marrow-derived mesenchymal stem cells.
 本発明における間葉系幹細胞は、成長特徴(例えば、継代から老化までの集団倍加能力、倍加時間)、核型分析(例えば、正常な核型、母体系統又は新生児系統)、フローサイトメトリー(例えば、FACS分析)による表面マーカー発現、免疫組織化学及び/又は免疫細胞化学(例えば、エピトープ検出)、遺伝子発現プロファイリング(例えば、遺伝子チップアレイ;逆転写PCR、リアルタイムPCR、従来型PCR等のポリメラーゼ連鎖反応)、miRNA発現プロファイリング、タンパク質アレイ、サイトカイン等のタンパク質分泌(例えば、血漿凝固解析、ELISA、サイトカインアレイ)、代謝産物(メタボローム解析)、本分野で知られている他の方法等によって、特徴付けられてもよい。 Mesenchymal stem cells according to the present invention have growth characteristics (eg, population doubling ability from aging to aging, doubling time), karyotype analysis (eg, normal karyotype, maternal line or neonatal line), flow cytometry ( For example, surface marker expression by FACS analysis, immunohistochemistry and / or immunocytochemistry (eg, epitope detection), gene expression profiling (eg, gene chip array; polymerase chain such as reverse transcription PCR, real-time PCR, conventional PCR, etc. Reaction), miRNA expression profiling, protein array, protein secretion such as cytokines (eg, plasma coagulation analysis, ELISA, cytokine array), metabolites (metabolome analysis), other methods known in the art, etc. May be.
(間葉系幹細胞の調製方法)
 間葉系幹細胞は、当業者に周知の方法により調製することができる。以下に、一つの例として、脂肪由来間葉系幹細胞の調製方法を説明する。脂肪由来間葉系幹細胞は、例えば米国特許第6,777,231号に記載の製造方法によって得られて良く、例えば、以下の工程(i)~(iii)を含む方法で製造することができる:
(i) 脂肪組織を酵素による消化により細胞懸濁物を得る工程;
(ii) 細胞を沈降させ、細胞を適切な培地に再懸濁する工程;ならびに
(iii) 細胞を固体表面で培養し、固体表面への結合を示さない細胞を除去する工程。
(Method for preparing mesenchymal stem cells)
Mesenchymal stem cells can be prepared by methods well known to those skilled in the art. Below, the preparation method of a fat-derived mesenchymal stem cell is demonstrated as an example. Adipose-derived mesenchymal stem cells may be obtained by the production method described in, for example, US Pat. No. 6,777,231, and can be produced, for example, by a method including the following steps (i) to (iii). :
(I) obtaining a cell suspension by digesting adipose tissue with an enzyme;
(Ii) sedimenting the cells and resuspending the cells in an appropriate medium; and (iii) culturing the cells on a solid surface and removing cells that do not show binding to the solid surface.
 工程(i)において用いる脂肪組織は、洗浄されたものを用いることが好ましい。洗浄は、生理学的に適合する生理食塩水溶液(例えばリン酸緩衝食塩水(PBS))を用いて、激しく攪拌して沈降させることによって行い得る。これは、脂肪組織に含まれる夾雑物 (デブリとも言い、例えば損傷組織、血液、赤血球など)を組織から除去するためである。したがって、洗浄及び沈降は一般に、上清からデブリが総体的に除去されるまで繰り返される。残存する細胞は、さまざまなサイズの塊として存在するので、細胞そのものの損傷を最小限に抑えながら解離させるため、洗浄後の細胞塊を、細胞間結合を弱めるか、又は破壊する酵素(例えば、コラゲナーゼ、ディスパーゼ又はトリプシンなど)で処理することが好ましい。このような酵素の量及び処理期間は、使用される条件に依存して変わるが、当技術分野で既知である。このような酵素処理に代えて、又は併用して、細胞塊を、機械的な攪拌、超音波エネルギー、熱エネルギーなどの他の処理法で分解することができるが、細胞の損傷を最小限に抑えるため、酵素処理のみで行うことが好ましい。酵素を用いた場合、細胞に対する有害な作用を最小限に抑えるために、適切な期間をおいた後に培地等を用いて酵素を失活させることが望ましい。 It is preferable to use a washed adipose tissue in the step (i). Washing may be performed by sedimentation with vigorous stirring using a physiologically compatible saline solution (eg, phosphate buffered saline (PBS)). This is to remove impurities (also called debris, such as damaged tissue, blood, and red blood cells) contained in the adipose tissue from the tissue. Accordingly, washing and sedimentation are generally repeated until the debris is totally removed from the supernatant. Since the remaining cells exist as lumps of various sizes, in order to dissociate them while minimizing damage to the cells themselves, the washed cell lumps are made to have an enzyme (eg, an enzyme that weakens or breaks cell-cell junctions). It is preferable to treat with collagenase, dispase or trypsin. The amount of such enzyme and the duration of treatment vary depending on the conditions used, but are known in the art. Instead of or in combination with such enzyme treatment, the cell mass can be broken down by other treatment methods such as mechanical agitation, ultrasonic energy, thermal energy, etc., but with minimal cell damage In order to suppress it, it is preferable to carry out only by enzyme treatment. When an enzyme is used, in order to minimize harmful effects on cells, it is desirable to deactivate the enzyme using a medium or the like after a suitable period of time.
 工程(i)により得られる細胞懸濁物は、凝集状の細胞のスラリー又は懸濁物、ならびに各種夾雑細胞、例えば赤血球、平滑筋細胞、内皮細胞、及び線維芽細胞を含む。従って、続いて凝集状態の細胞とこれらの夾雑細胞を分離、除去してもよいが、後述する工程(iii)での接着及び洗浄により、除去可能であることから、当該分離、除去は割愛してもよい。夾雑細胞を分離、除去する場合、細胞を上清と沈殿に強制的に分ける遠心分離によって達成しえる。得られた夾雑細胞を含む沈殿は、生理学的に適合する溶媒に懸濁させる。懸濁状の細胞には、赤血球を含む恐れがあるが、後述する個体表面への接着による選択により、赤血球は除外されるため、溶解する工程は必ずしも必要ではない。赤血球を選択的に溶解する方法として、例えば、塩化アンモニウムによる溶解による高張培地又は低張培地中でのインキュベーションなど、当技術分野で周知の方法を使用することができる。溶解後、例えば濾過、遠心沈降、又は密度分画によって溶解物を所望の細胞から分離してもよい。 The cell suspension obtained by the step (i) includes a slurry or suspension of aggregated cells, and various kinds of contaminated cells such as erythrocytes, smooth muscle cells, endothelial cells, and fibroblasts. Therefore, the cells in the aggregated state and these contaminated cells may be separated and removed, but they can be removed by adhesion and washing in step (iii) to be described later. May be. When contaminating cells are separated and removed, this can be achieved by centrifugation that forcibly separates the cells into a supernatant and a precipitate. The resulting precipitate containing contaminating cells is suspended in a physiologically compatible solvent. Suspended cells may contain erythrocytes, but erythrocytes are excluded by selection by adhesion to the individual surface described later, and thus a lysis step is not always necessary. As a method for selectively lysing erythrocytes, for example, a method well known in the art such as incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride can be used. After lysis, the lysate may be separated from the desired cells, for example, by filtration, centrifugation, or density fractionation.
 工程(ii)において、懸濁状の細胞において、間葉系幹細胞の純度を高めるために、1回もしくは連続して複数回洗浄し、遠心分離し、培地に再懸濁してもよい。この他にも、細胞を、細胞表面マーカープロファイルを基に、又は細胞のサイズ及び顆粒性を基に分離してもよい。 In step (ii), in order to increase the purity of the mesenchymal stem cells in the suspended cells, the cells may be washed once or continuously several times, centrifuged, and resuspended in the medium. Alternatively, cells may be separated based on cell surface marker profile or based on cell size and granularity.
 再懸濁において用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されないが、このような培地は、基礎培地に、血清を添加する、及び/又は、アルブミン、トランスフェリン、脂肪酸、インスリン、亜セレン酸ナトリウム、コレステロール、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオールグリセロール等の1つ以上の血清代替物を添加して作製してもよい。これらの培地には、必要に応じて、さらに脂質、アミノ酸、タンパク質、多糖、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等の物質を添加してもよい。 The medium used in the resuspension is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes basal medium added with serum and / or albumin, transferrin, fatty acid, One or more serum substitutes such as insulin, sodium selenite, cholesterol, collagen precursor, trace elements, 2-mercaptoethanol, 3′-thiolglycerol, and the like may be added. In addition to these media, substances such as lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular compounds, antibiotics, antioxidants, pyruvate, buffers, inorganic salts, etc. are added as necessary. May be.
 上記基礎培地としては、例えば、IMDM培地、Medium 199培地、Eagle’s Minimum Essential Medium(EMEM)培地、αMEM培地、Dulbecco’s modified Eagle’s Medium(DMEM)培地、Ham’s F12培地、RPMI 1640培地、Fischer’s培地、MCDB201培地及びこれらの混合培地等が挙げられる。 Examples of the basal medium include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, αMEM medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, Ham's F16 medium. Examples include a medium, Fischer's medium, MCDB201 medium, and a mixed medium thereof.
 上記血清としては、例えば、ヒト血清、ウシ胎児血清(FBS)、ウシ血清、仔ウシ血清、ヤギ血清、ウマ血清、ブタ血清、ヒツジ血清、ウサギ血清、ラット血清等が挙げられるがこれらに限定されない。血清を用いる場合、基礎培地に対して、5v/v%から15v/v%、好ましくは、10v/v%を添加してもよい。 Examples of the serum include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like. . When using serum, 5 v / v% to 15 v / v%, preferably 10 v / v% may be added to the basal medium.
 上記脂肪酸としては、リノール酸、オレイン酸、リノレイン酸、アラキドン酸、ミリスチン酸、パルミトイル酸、パルミチン酸、及びステアリン酸等が例示されるが、これらに限定されない。脂質は、フォスファチジルセリン、フォスファチジルエタノールアミン、フォスファチジルコリン等が例示されるが、これらに限定されない。アミノ酸は、例えば、L-アラニン、L-アルギニン、L-アスパラギン酸、L-アスパラギン、L-システイン、L-シスチン、L-グルタミン酸、L-グルタミン、L-グリシンなどを含むがこれらに限定されない。タンパク質は、例えば、エコチン、還元型グルタチオン、フィブロネクチン及びβ2-ミクログロブリン等が例示されるが、これらに限定されない。多糖は、グリコサミノグリカンが例示され、グリコサミノグリカンのうち特に、ヒアルロン酸、ヘパラン硫酸等が例示されるが、これらに限定されない。増殖因子は、例えば、血小板由来増殖因子(PDGF)、塩基性線維芽細胞成長因子(bFGF)、トランスフォーミング増殖因子ベータ(TGF-β)、肝細胞増殖因子(HGF)、上皮成長因子(EGF)、結合組織増殖因子(CTGF)、血管内皮細胞増殖因子(VEGF)等が例示されるが、これらに限定されない。本発明において得られる脂肪由来間葉系幹細胞を細胞移植に用いるという観点から、血清等の異種由来成分を含まない(ゼノフリー)培地を用いることが好ましい。このような培地は、例えば、PromoCell社、Lonza社、Biological Industries社、Veritas社、R&D Systems社、Corning社及びRohto社などから間葉系幹細胞(間質細胞)用として予め調製された培地として提供されている。 Examples of the fatty acid include, but are not limited to, linoleic acid, oleic acid, linolenic acid, arachidonic acid, myristic acid, palmitoyl acid, palmitic acid, and stearic acid. Examples of the lipid include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, phosphatidylcholine, and the like. Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like. Examples of proteins include, but are not limited to, ecotin, reduced glutathione, fibronectin and β2-microglobulin. Examples of the polysaccharide include glycosaminoglycans, and among the glycosaminoglycans, hyaluronic acid, heparan sulfate and the like are particularly exemplified, but the polysaccharide is not limited thereto. Growth factors include, for example, platelet derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF-β), hepatocyte growth factor (HGF), epidermal growth factor (EGF) Examples include, but are not limited to, connective tissue growth factor (CTGF), vascular endothelial growth factor (VEGF), and the like. From the viewpoint of using the adipose-derived mesenchymal stem cells obtained in the present invention for cell transplantation, it is preferable to use a (xenofree) medium that does not contain components derived from different species such as serum. Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from, for example, PromoCell, Lonza, Biological Industries, Veritas, R & D Systems, Corning, and Roto. Has been.
 続いて、工程(iii)では、工程(ii)で得られた細胞懸濁液中の細胞を分化させずに固体表面上で、上述の適切な細胞培地を使用して、適切な細胞密度及び培養条件で培養する。本発明において、「固体表面」とは、本発明における脂肪由来間葉系幹細胞の結合・接着を可能とする任意の材料を意味する。特定の態様では、このような材料は、その表面への哺乳類細胞の結合・接着を促すように処理されたプラスチック材料である。固体表面を有する培養容器の形状は特に限定されないが、シャーレやフラスコなどが好適に用いられる。非結合状態の細胞及び細胞の破片を除去するために、インキュベーション後に細胞を洗浄する。 Subsequently, in step (iii), the appropriate cell density and the appropriate cell medium are used on the solid surface without differentiating the cells in the cell suspension obtained in step (ii). Culture under culture conditions. In the present invention, the “solid surface” means any material capable of binding / adhering the adipose-derived mesenchymal stem cells in the present invention. In a particular embodiment, such a material is a plastic material that has been treated to promote the attachment and adhesion of mammalian cells to its surface. The shape of the culture vessel having a solid surface is not particularly limited, but a petri dish or a flask is preferably used. The cells are washed after incubation to remove unbound cells and cell debris.
 本発明では、最終的に固体表面に結合・接着した状態で留まる細胞を、脂肪由来間葉系幹細胞の細胞集団として選択することができる。 In the present invention, cells that finally remain attached and adhered to the solid surface can be selected as a cell population of adipose-derived mesenchymal stem cells.
 選択された細胞について、本発明における脂肪由来間葉系幹細胞であることを確認するために、表面抗原についてフローサイトメトリー等を用いて従来の方法で解析してもよい。さらに、各細胞系列に分化する能力について検査してもよく、このような分化は、従来の方法で行うことができる。 In order to confirm that the selected cells are the adipose-derived mesenchymal stem cells in the present invention, the surface antigen may be analyzed by a conventional method using flow cytometry or the like. Further, the ability to differentiate into each cell line may be examined, and such differentiation can be performed by conventional methods.
 本発明における間葉系幹細胞は、上述の通り調製することができるが、次の特性を持つ細胞として定義してもよい;
(1)標準培地での培養条件で、プラスチックに接着性を示す、
(2)表面抗原CD44、CD73、CD90が陽性であり、CD31、CD45が陰性であり、及び
(3)培養条件にて骨細胞、脂肪細胞、軟骨細胞に分化可能。
Mesenchymal stem cells in the present invention can be prepared as described above, but may be defined as cells having the following characteristics;
(1) It exhibits adhesiveness to plastic under the culture conditions in a standard medium.
(2) Surface antigens CD44, CD73, CD90 are positive, CD31, CD45 are negative, and (3) Differentiated into bone cells, adipocytes, and chondrocytes under culture conditions.
(末梢血単核球)
 本発明において末梢血単核球細胞とは、ヒト又は動物の末梢血から取得される、リンパ球、好中球、好酸球、好塩基球、単球を含む分画をいう。末梢血単核球は、末梢血からFicoll-hypaque(登録商標)等を用いた密度勾配遠心法により分離することができる。本発明における(A)細胞としての末梢血単核球は、末梢血から分離した状態の細胞でもよいし、それらを必要に応じて、各種因子、低分子化合物、抗体等と共に培養等することで、増殖・活性化させたものであってもよい。
(Peripheral blood mononuclear cells)
In the present invention, peripheral blood mononuclear cells refer to fractions containing lymphocytes, neutrophils, eosinophils, basophils, and monocytes, which are obtained from human or animal peripheral blood. Peripheral blood mononuclear cells can be separated from peripheral blood by density gradient centrifugation using Ficoll-hypaque (registered trademark) or the like. Peripheral blood mononuclear cells as cells (A) in the present invention may be cells separated from peripheral blood, or they may be cultured with various factors, low molecular compounds, antibodies, etc. as necessary. It may be proliferated and activated.
((A)細胞の凍結保存)
 本発明における(A)細胞は、各種疾患に対する治療効果を備えていれば、適宜、凍結保存及び融解を繰り返した細胞であってもよい。本発明において、凍結保存は、当業者に周知の凍結保存液で(A)細胞を懸濁し、冷却することによって行い得る。懸濁は、細胞を、必要に応じてトリプシンなどの剥離剤によって剥離し、凍結保存容器に移し、適宜処理した後、凍結保存液を加えることによって行い得る。
((A) Cryopreservation of cells)
The (A) cell in the present invention may be a cell that has been appropriately cryopreserved and thawed as long as it has a therapeutic effect on various diseases. In the present invention, cryopreservation can be performed by suspending (A) cells in a cryopreservation solution well known to those skilled in the art and cooling. Suspension can be performed by detaching the cells with a release agent such as trypsin as necessary, transferring them to a cryopreservation container, treating them appropriately, and adding a cryopreservation solution.
 凍結保存液は、凍害防御剤として、DMSO(Dimethyl sulfoxide)を含有していてもよいが、DMSOは、細胞毒性を有することから、DMSO含有量を減らすことが好ましい。なお、DMSOは間葉系幹細胞に対しては、分化誘導する特性を有することも知られている。DMSOの代替物として、グリセロール、プロピレングリコール又は多糖類が例示される。DMSOを用いる場合、5%~20%の濃度、好ましくは5%~10%の濃度、より好ましくは10%の濃度を含有する。この他にも、WO2007/058308に記載の添加剤を含んでもよい。このような凍結保存液として、例えば、バイオベルデ社、日本ジェネティクス株式会社、リプロセル社、ゼノアック社、コスモ・バイオ社、コージンバイオ株式会社、サーモフィッシャーサイエンティフィック社等から提供されている凍結保存液を用いてもよい。 The cryopreservation solution may contain DMSO (Dimethylsulfoxide) as a frost damage protective agent. However, since DMSO is cytotoxic, it is preferable to reduce the DMSO content. DMSO is also known to have differentiation-inducing properties for mesenchymal stem cells. As an alternative to DMSO, glycerol, propylene glycol or polysaccharides are exemplified. When DMSO is used, it contains a concentration of 5% to 20%, preferably a concentration of 5% to 10%, more preferably a concentration of 10%. In addition, additives described in WO2007 / 058308 may be included. As such a cryopreservation solution, for example, cryopreservation provided by Bioverde, Nippon Genetics, Reprocell, Xenoac, Cosmo Bio, Kojin Bio, Thermo Fisher Scientific, etc. A liquid may be used.
 上述の懸濁した細胞を凍結保存する場合、-80℃~-100℃の間の温度(例えば、-80℃)で保管することで良く、当該温度に達成しえる任意のフリーザーを用いて行い得る。特に限定されないが、急激な温度変化を回避するため、プログラムフリーザーを用いて、冷却速度を適宜制御してもよい。冷却速度は、凍結保存液の成分によって適宜選択しても良く、凍結保存液の製造者指示に従って行われ得る。 When storing the above-mentioned suspended cells in a frozen state, it may be stored at a temperature between −80 ° C. and −100 ° C. (eg, −80 ° C.), and any freezer capable of achieving the temperature is used. obtain. Although not particularly limited, in order to avoid a sudden temperature change, the cooling rate may be appropriately controlled using a program freezer. The cooling rate may be appropriately selected depending on the components of the cryopreservation solution, and may be performed according to the manufacturer's instructions for the cryopreservation solution.
 保存期間は、上記条件で凍結保存した細胞が融解した後、凍結前と同等の性質を保持している限り、特に上限は限定されないが、例えば、1週間以上、2週間以上、3週間以上、4週間以上、2か月以上、3か月以上、4か月以上、5か月以上、6か月以上、1年以上、又はそれ以上が挙げられる。より低い温度で保存することで細胞障害を抑制することができるため、液体窒素上の気相(約-150℃以下から-180℃以下)へ移して保存してもよい。液体窒素上の気相で保存する場合、当業者に周知の保存容器を用いて行うことができる。特に限定されないが、例えば、2週間以上保存する場合、液体窒素上の気相で保存することが好ましい。 The upper limit of the storage period is not particularly limited as long as the cells cryopreserved under the above conditions are thawed and retain the same properties as before freezing, for example, 1 week or more, 2 weeks or more, 3 weeks or more, 4 weeks or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, 6 months or more, 1 year or more, or more. Since cell damage can be suppressed by storing at a lower temperature, it may be transferred to a gas phase on liquid nitrogen (from about −150 ° C. to −180 ° C.). When storing in a gas phase on liquid nitrogen, it can be performed using a storage container well known to those skilled in the art. Although not particularly limited, for example, when storing for 2 weeks or more, it is preferable to store in a gas phase on liquid nitrogen.
 融解した(A)細胞は、次の凍結保存までに適宜培養してもよい。例えば、間葉系幹細胞の培養は、上述した間葉系幹細胞を培養できる培地を用いて行われ、特に限定されないが、約30~40℃、好ましくは約37℃の培養温度で、CO含有空気の雰囲気下で行われてもよい。CO濃度は、約2~5%、好ましくは約5%である。培養において、培養容器に対して適切なコンフルエンシー(例えば、培養容器に対して、50%から80%を細胞が占有することが挙げられる)に達した後に、細胞をトリプシンなどの剥離剤によって剥離し、別途用意した培養容器に適切な細胞密度で播種して培養を継続してもよい。細胞を播種する際において、典型的な細胞密度として、100細胞/cm~100,000細胞/cm、500細胞/cm~50,000細胞/cm、1,000~10,000細胞/cm、2,000~10,000細胞/cmなどが例示される。特定の態様では、細胞密度は2,000~10,000細胞/cmである。適切なコンフルエンシーに達するまでの期間が、3日間から7日間となるように調整することが好ましい。培養中、必要に応じて、適宜、培地を交換してもよい。 The thawed (A) cells may be appropriately cultured before the next cryopreservation. For example, the mesenchymal stem cells are cultured using the above-described medium capable of culturing mesenchymal stem cells, and are not particularly limited, but contain CO 2 at a culture temperature of about 30 to 40 ° C., preferably about 37 ° C. It may be performed in an air atmosphere. The CO 2 concentration is about 2-5%, preferably about 5%. In culture, after reaching the appropriate confluency for the culture container (for example, cells may occupy 50% to 80% of the culture container), the cells are detached with a release agent such as trypsin. Alternatively, the culture may be continued by seeding in a separately prepared culture vessel at an appropriate cell density. When seeding cells, typical cell densities are 100 cells / cm 2 to 100,000 cells / cm 2 , 500 cells / cm 2 to 50,000 cells / cm 2 , 1,000 to 10,000 cells. / Cm 2 , 2,000 to 10,000 cells / cm 2, etc. are exemplified. In a particular embodiment, the cell density is between 2,000 and 10,000 cells / cm 2 . It is preferable to adjust the period until reaching appropriate confluency from 3 days to 7 days. During culture, the medium may be changed as necessary.
 凍結保存した細胞の融解は、当業者に周知の方法によって行い得る。例えば、37℃の恒温槽内又は湯浴中にて静置又は振とうすることによって行う方法が例示される。 Thawing of cryopreserved cells can be performed by methods well known to those skilled in the art. For example, the method performed by standing or shaking in a 37 degreeC thermostat or a hot water bath is illustrated.
((A)細胞の形態)
 本発明の細胞医薬組成物が含有する(A)細胞は、いずれの状態の細胞であってもよく、例えば培養中の細胞を剥離して回収された細胞でもよいし、凍結保存液中に凍結された状態の細胞でもよい。拡大培養して得られる同ロットの細胞を小分けして凍結保存したものを使用すると、安定して同様の作用効果が得られる点、取扱い性に優れる点等において好ましい。
((A) Cell morphology)
The cell (A) contained in the cell pharmaceutical composition of the present invention may be a cell in any state, for example, a cell recovered by detaching a cell in culture, or frozen in a cryopreservation solution It may be a cell in a state of being formed. Use of the same lot of cells obtained by expanding and culturing in the same lot is preferable in that the same action and effect can be stably obtained and the handling property is excellent.
 凍結保存状態の(A)細胞は、使用直前に融解し、凍結保存液に懸濁したまま後述する(B)細胞懸濁用溶液に直接混合してもよい。また、遠心分離等の方法により凍結保存液を除去してから(B)細胞懸濁用溶液に懸濁してもよい。 The (A) cells in the cryopreservation state may be thawed immediately before use and directly mixed with the cell suspension solution (B) described later while being suspended in the cryopreservation solution. Alternatively, the cryopreservation solution may be removed by a method such as centrifugation and then suspended in the (B) cell suspension solution.
 本発明の(A)細胞の用量(投与量)は、患者の状態(体重、年齢、症状、体調等)、及び剤形等によって異なりうるが、十分な治療効果を奏する観点からは、その量は多い方が好ましい傾向にあり、一方、副作用の発現を抑制する観点からはその量は少ない方が好ましい傾向にある。通常、成人に投与する場合には、細胞数として、1x10~1x1012個/回、好ましくは1x10~1x1011個/回、更に好ましくは1x10~1x1010個/回、特に好ましくは5x10~1x10個/回である。なお、本用量を1回量として、複数回投与してもよく、本用量を複数回に分けて投与しても良い。 The dose (dosage) of (A) cells of the present invention may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.), dosage form, etc., but from the standpoint of sufficient therapeutic effect, the amount From the viewpoint of suppressing the occurrence of side effects, a smaller amount tends to be preferable. Usually, when administered to an adult, the number of cells is 1 × 10 3 to 1 × 10 12 cells / time, preferably 1 × 10 4 to 1 × 10 11 cells / time, more preferably 1 × 10 5 to 1 × 10 10 cells / time, particularly preferably 5 × 10 5. 6 to 1 × 10 9 pieces / time. In addition, this dose may be administered once as a single dose, or the dose may be divided into multiple doses.
 本発明の(A)細胞の用量(投与量)は、患者の状態(体重、年齢、症状、体調等)、及び本発明の組成物の剤形等によって異なりうるが、通常、成人に投与する場合には、細胞数として、1x10~5x1010個/kg、好ましくは1x10~5x10個/kg、更に好ましくは1x10~5x10個/kg、特に好ましくは1x10~5x10個/kgである。なお、本用量を1回量として、複数回投与してもよく、本用量を複数回に分けて投与しても良い。 The dose (dosage) of the cell (A) of the present invention may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.) and the dosage form of the composition of the present invention, but is usually administered to an adult. In this case, the number of cells is 1 × 10 to 5 × 10 10 cells / kg, preferably 1 × 10 2 to 5 × 10 9 cells / kg, more preferably 1 × 10 3 to 5 × 10 8 cells / kg, particularly preferably 1 × 10 4 to 5 × 10 7 cells / kg. It is. In addition, this dose may be administered once as a single dose, or the dose may be divided into multiple doses.
[(B)細胞懸濁用溶液]
 本発明の(B)細胞懸濁用溶液は、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である。(B)細胞懸濁用溶液は、CHCOO、HCO 、クエン酸イオン及び乳酸イオンからなる群より選択される少なくとも1種の電解質をさらに含んでいてもよい。(B)細胞懸濁用溶液は、生体における細胞外液補液効果を有する等張性電解質輸液製剤ともいえ、具体的には、リンゲル液、酢酸リンゲル液重炭酸リンゲル液及び乳酸リンゲル液からなる群より選択される少なくとも1種を好ましいものとして挙げられる。このような組成の(B)細胞懸濁用溶液を採用することで、本発明の細胞医薬組成物においては(A)細胞の状態を良好に保ち、その生存率を長時間に渡って高い状態で維持することができる。
[(B) Cell suspension solution]
The (B) cell suspension solution of the present invention is an isotonic electrolyte solution containing Na + , Cl , K + and Ca 2+ as electrolytes and not containing carbohydrates. (B) The cell suspension solution may further contain at least one electrolyte selected from the group consisting of CH 3 COO , HCO 3 , citrate ions and lactate ions. (B) The solution for cell suspension can be said to be an isotonic electrolyte infusion preparation having an extracellular fluid replacement effect in a living body, and is specifically selected from the group consisting of Ringer's solution, Ringer's acetate solution, Ringer's bicarbonate bicarbonate solution, and Ringer's lactate solution At least one of them is preferred. By adopting the (B) cell suspension solution having such a composition, in the cell pharmaceutical composition of the present invention, (A) the state of the cells is kept good, and the survival rate is high over a long period of time. Can be maintained.
 (B)細胞懸濁用溶液としては、糖質を含まず、かつ電解質としてNa、K、Ca2+、Clを含み、Naが140mEq/L~160mEq/L、Kが1mEq/L~10mEq/L、Ca2+が1mEq/L~10mEq/L、Clが140mEq/L~170mEq/Lであるリンゲル液を好ましいものとして挙げることができる。上記リンゲル液は各電解質を上記濃度範囲として調製することができるが、リンゲル液「オーツカ」(日局リンゲル液、株式会社大塚製薬工場)、リンゲル液「フソー」(日局リンゲル液、扶桑薬品工業株式会社)等の市販品を用いることもできる。 (B) The cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Cl as electrolytes, Na + is 140 mEq / L to 160 mEq / L, and K + is 1 mEq / L. A Ringer solution having L to 10 mEq / L, Ca 2+ of 1 mEq / L to 10 mEq / L, and Cl of 140 mEq / L to 170 mEq / L can be mentioned as a preferable example. The above Ringer solution can be prepared with each electrolyte in the above concentration range, such as Ringer solution “Otsuka” (Nippon Ringer solution, Otsuka Pharmaceutical Factory), Ringer solution “Fuso” (Nippon Ringer solution, Fuso Pharmaceutical Co., Ltd.), etc. Commercial products can also be used.
 (B)細胞懸濁用溶液としては、糖質を含まず、かつ電解質としてNa、K、Ca2+、Cl、CHCOOを含み、Naが120mEq/L~140mEq/L、Kが1mEq/L~10mEq/L、Ca2+が1mEq/L~10mEq/L、Clが90mEq/L~130mEq/L、CHCOOが10mEq/L~40mEq/Lである酢酸リンゲル液を好ましいものとして挙げることができる。上記酢酸リンゲル液は各電解質を上記濃度範囲として調製することができるが、ヴィーン(登録商標)F輸液(興和株式会社)、ソリューゲン(登録商標)F注(共和クリティケア株式会社)、ソルアセト(登録商標)F輸液(テルモ株式会社)等の市販品を用いることもできる。 (B) The cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Cl , CH 3 COO as electrolytes, and Na + is 120 mEq / L to 140 mEq / L, An acetic Ringer solution in which K + is 1 mEq / L to 10 mEq / L, Ca 2+ is 1 mEq / L to 10 mEq / L, Cl is 90 mEq / L to 130 mEq / L, and CH 3 COO is 10 mEq / L to 40 mEq / L. It can be mentioned as a preferable one. The above-mentioned Ringer's solution can be prepared by adjusting the concentration of each electrolyte to the above-mentioned concentration range. Commercial products such as F infusion (Terumo Corporation) can also be used.
 (B)細胞懸濁用溶液としては、糖質を含まず、かつ電解質としてNa、K、Ca2+、Mg2+、Cl、HCO 、クエン酸イオン(Citrate3+)を含み、それぞれの電解質濃度は、Naが120mEq/L~145mEq/L、Kが1mEq/L~10mEq/L、Ca2+が1mEq/L~10mEq/L、Mg2+が0.1mEq/L~10mEq/L、Clが95mEq/L~135mEq/L、HCO が10mEq/L~40mEq/L、クエン酸イオン(Citrate3+)が1mEq/L~10mEq/Lである重炭酸リンゲル液を好ましいものとして挙げることができる。上記重炭酸リンゲル液は各電解質を上記濃度範囲として調製することができるが、ビカネイト(登録商標)輸液(株式会社大塚製薬工場)、ビカーボン(登録商標)輸液(エイワイファーマ株式会社)等の市販品を用いることもできる。 (B) The cell suspension solution does not contain carbohydrates, and contains Na + , K + , Ca 2+ , Mg 2+ , Cl , HCO 3 , citrate ions (Citrate 3+ ) as electrolytes, The electrolyte concentrations of Na + are 120 mEq / L to 145 mEq / L, K + is 1 mEq / L to 10 mEq / L, Ca 2+ is 1 mEq / L to 10 mEq / L, and Mg 2+ is 0.1 mEq / L to 10 mEq / L. Preferred is a Ringer's solution of bicarbonate, wherein Cl is 95 mEq / L to 135 mEq / L, HCO 3 is 10 mEq / L to 40 mEq / L, and citrate ion (Citrate 3+ ) is 1 mEq / L to 10 mEq / L. Can do. The bicarbonate Ringer's solution can be prepared with each electrolyte in the above-mentioned concentration range. However, commercially available products such as Bicanate (registered trademark) infusion (Otsuka Pharmaceutical Factory), Bicarbon (registered trademark) infusion (Ai Pharma Co., Ltd.), etc. Can also be used.
 (B)細胞懸濁用溶液としては、糖質を含まず、かつ電解質としてNa、K、Ca2+、Cl、乳酸イオン(Lactate-)を含み、それぞれの電解質濃度は、Naが120mEq/L~140mEq/L、Kが1mEq/L~10mEq/L、Ca2+が1mEq/L~10mEq/L、Clが90mEq/L~130mEq/L、Lactateが15mEq/L~45mEq/Lである乳酸リンゲル液を好ましいものとして挙げることができる。上記乳酸リンゲル液は各電解質を上記濃度範囲として調製することができるが、ソルラクト(登録商標)輸液(テルモ株式会社)、ニソリ(登録商標)輸液(マイラン製薬株式会社)、ハルトマン液「コバヤシ」(共和クリティケア株式会社)、ハルトマン輸液「NP」(ニプロ株式会社)、ハルトマン輸液pH8「NP」(ニプロ株式会社)、ラクテック(商標登録)注(株式会社大塚製薬工場)、ラクトリンゲル液“フソー”(扶桑薬品工業株式会社)等の市販品を用いることもできる。 The (B) a solution for cell suspension, free of carbohydrates, and Na +, K +, Ca 2+ as the electrolyte, Cl -, lactate (Lactate -) include respective electrolyte concentration, Na + is 120 mEq / L to 140 mEq / L, K + 1 mEq / L to 10 mEq / L, Ca 2+ 1 mEq / L to 10 mEq / L, Cl 90 mEq / L to 130 mEq / L, Lactate 15 mEq / L to 45 mEq / L A preferred example is Lactated Ringer's solution which is L. The above Lactated Ringer solution can be prepared with each electrolyte in the above-mentioned concentration range. Kriticare Co., Ltd.), Hartmann Infusion "NP" (Nipro Corporation), Hartmann Infusion pH 8 "NP" (Nipro Corporation), Lactec (registered trademark) Note (Otsuka Pharmaceutical Factory), Lactolin Gel Solution "Fuso" (Fuso Pharmaceutical) Commercial products such as Kogyo Co., Ltd. can also be used.
[他の薬剤]
 本発明の細胞医薬組成物は、1又は2以上の、疾患に対する治療効果を有する他の薬剤を含有してもよい。他の薬剤としては、肝疾患治療薬、心疾患治療薬、炎症性腸疾患治療薬、呼吸器用薬、神経系用薬、循環器用薬、脳循環改善薬、免疫抑制薬として用いることができる任意の薬剤が挙げられる。
[Other drugs]
The cell pharmaceutical composition of the present invention may contain one or more other drugs having a therapeutic effect on a disease. Other drugs that can be used as liver disease drugs, heart disease drugs, inflammatory bowel disease drugs, respiratory drugs, nervous system drugs, cardiovascular drugs, cerebral circulation improving drugs, immunosuppressive drugs Of these drugs.
 肝疾患治療薬としては、例えば、B型肝炎治療薬(ラミブジン、アデホビル、エンテカビル、テノホビル等)、インターフェロン製剤(インターフェロンα、インターフェロンα-2b、インターフェロンβ、ペグインターフェロンα-2a、ペグインターフェロンα-2b等)、C型肝炎治療薬(リバビリン、テラピレビル、シメプレビル、バニプレビル、ダクラタスビル、アスナプレビル、ソホスブビル等)、副腎皮質ステロイド(プレドニゾロン、メチルプレドニゾロンコハク酸エステルナトリウム等)、抗凝固剤(乾燥濃縮人アンチトロンビンIII、ガベキサートメシル酸塩、トロンボモデュリンα等)、解毒剤(エデト酸カルシウム二ナトリウム水和物、グルタチオン、ジメチカプロール、チオ硫酸ナトリウム水和物、スガマデスクナトリウム等)、人血清アルブミン、肝臓抽出エキス、ウルソデオキシコール酸、グリチルリチン酸、アザチオプリン、ベザフィーブラート、アミノ酸(グリシン、L-システイン、L-イソロイシン、L-ロイシン、L-バリン、L-トレオニン、L-セリン、L-アラニン、L-メチオニン、L-フェニルアラニン、L-トリプトファン、L-リシン、L-ヒスチジン、L-アルギニン及びこれらの塩等)、ビタミン(トコフェロール、フラビンアデニンジヌクレオチド、リン酸チアミンジスルフィド、ピリドキシン、シアノコバラミン及びこれらの塩等)、抗生物質(スルバクタムナトリウム、セフォペラゾンナトリウム、メロペネム水和物、塩酸バンコマイシン等)等が挙げられる。 Examples of therapeutic agents for liver diseases include hepatitis B therapeutic agents (lamivudine, adefovir, entecavir, tenofovir, etc.), interferon preparations (interferon α, interferon α-2b, interferon β, peginterferon α-2a, peginterferon α-2b, etc. Etc.), hepatitis C drugs (ribavirin, terapyrevir, simeprevir, vaniprevir, daclatasvir, asunaprevir, sofosbuvir, etc.), corticosteroids (prednisolone, methylprednisolone sodium succinate, etc.), anticoagulants (dry concentrated human antithrombin III) , Gabexate mesylate, thrombomodulin α, etc.), antidote (calcium edetate disodium hydrate, glutathione, dimethicaprol, sodium thiosulfate hydrate, Sugamasekuna Human serum albumin, liver extract, ursodeoxycholic acid, glycyrrhizic acid, azathioprine, bezafibrate, amino acids (glycine, L-cysteine, L-isoleucine, L-leucine, L-valine, L-threonine, L-serine, L-alanine, L-methionine, L-phenylalanine, L-tryptophan, L-lysine, L-histidine, L-arginine and their salts, vitamins (tocopherol, flavin adenine dinucleotide, thiamine phosphate) Disulfide, pyridoxine, cyanocobalamin and their salts), antibiotics (sulbactam sodium, cefoperazone sodium, meropenem hydrate, vancomycin hydrochloride, etc.).
 心疾患治療薬としては、例えば、ACE阻害薬、アンギオテンシンII受容体拮抗薬、β遮断薬、抗血小板薬、ワーファリン、カルシウム拮抗薬、硝酸薬、利尿剤、HMG-CoA還元酵素阻害薬、アンカロン等が挙げられる。 Examples of therapeutic agents for heart diseases include ACE inhibitors, angiotensin II receptor antagonists, β-blockers, antiplatelet agents, warfarin, calcium antagonists, nitrates, diuretics, HMG-CoA reductase inhibitors, ancalons, etc. Is mentioned.
 炎症性腸疾患治療薬としては、例えば、サラゾスルファピリジン、メサラジン等が挙げられる。 Examples of the therapeutic agent for inflammatory bowel disease include salazosulfapyridine and mesalazine.
 呼吸器用薬としては、例えば、ジモルホラミン、ドキサプラム塩酸塩水和物、シベレスタットナトリウム水和物、ピルフェニドン、肺サーファクタント、ドルナーゼ アルファ等が挙げられる。 Examples of the respiratory medicine include dimorpholine, doxapram hydrochloride hydrate, cyberestat sodium hydrate, pirfenidone, pulmonary surfactant, and Dornase alpha.
 神経系用薬としては、例えば、エダラボン、インターフェロンベータ-1a、インターフェロンベータ-1b、フィンゴリモド塩酸塩、リルゾール、タルチレリン水和物等が挙げられる。 Examples of the nervous system drug include edaravone, interferon beta-1a, interferon beta-1b, fingolimod hydrochloride, riluzole, tartilelin hydrate and the like.
 循環器用薬としては、例えば、ヘプロニカート、ミドドリン塩酸塩、アメジニウムメチルメチル硫酸塩、エチレフリン塩酸塩、フェニレフリン塩酸塩等が挙げられる。 Examples of circulatory drugs include hepronicart, midodrine hydrochloride, amedinium methylmethyl sulfate, ethylephrine hydrochloride, phenylephrine hydrochloride, and the like.
 脳循環改善薬としては、例えば、イフェンプロジル酒石酸塩、ニセルゴリン、イプジラスト、ジヒドロエルゴトキシンメシル酸塩、ニゾフェノンフマル酸塩、ファスジル塩酸塩水和物等が挙げられる。 Examples of the cerebral circulation improving drug include ifenprodil tartrate, nicergoline, ipdilast, dihydroergotoxin mesylate, nizophenone fumarate, fasudil hydrochloride hydrate and the like.
 免疫抑制薬としては、例えば、シクロスポリン、アザチオプリン、ミゾリビン、バシリキシマブ、タクロリムス水和物、グスペリムス塩酸塩、ミコフェノール酸モフェチル、エベロリムス等が挙げられる。 Examples of the immunosuppressant include cyclosporine, azathioprine, mizoribine, basiliximab, tacrolimus hydrate, gusperimus hydrochloride, mycophenolate mofetil, everolimus and the like.
 上記他の薬剤を本発明の細胞医薬組成物が含有する場合、保存時には、(A)細胞、(B)細胞懸濁用溶液とは別の容器中に保存されていてもよいし、いずれかに配合される形で含有されていてもよい。疾患の種類、治療方法、患者の状態等により、他の薬剤と、(A)細胞及び(B)細胞懸濁用溶液とを同時に投与してもよいし、一定の間隔をあけて投与してもよい。 When the cell pharmaceutical composition of the present invention contains the above-mentioned other drug, at the time of storage, it may be stored in a container different from (A) the cell and (B) the cell suspension solution. It may be contained in a form blended with. Depending on the type of disease, treatment method, patient condition, etc., other drugs and (A) cells and (B) cell suspension solutions may be administered simultaneously, or at regular intervals. Also good.
 本発明の細胞医薬組成物は、本発明の効果を損なわない範囲であれば、上記(A)細胞及び(B)細胞懸濁用溶液以外に、その用途や形態に応じて、常法に従い、薬学的に許容される担体や添加物等のその他の成分を含有させてもよい。このような担体や添加物は、(B)細胞懸濁用溶液に含有させてもよいし、(B)細胞懸濁用溶液とは別に含有させてもよい。このような担体や添加物としては、例えば、等張化剤、増粘剤、糖類、糖アルコール類、防腐剤(保存剤)、殺菌剤又は抗菌剤、pH調節剤、安定化剤、キレート剤、油性基剤、ゲル基剤、界面活性剤、懸濁化剤、結合剤、賦形剤、滑沢剤、崩壊剤、発泡剤、流動化剤、分散剤、乳化剤、緩衝剤、溶解補助剤、抗酸化剤、甘味剤、酸味剤、着色剤、呈味剤、香料又は清涼化剤等が挙げられるが、これらに限定されない。 If the cell pharmaceutical composition of the present invention is within a range not impairing the effects of the present invention, in addition to the above (A) cell and (B) cell suspension solution, according to its use and form, according to a conventional method, Other components such as pharmaceutically acceptable carriers and additives may be included. Such carriers and additives may be contained in the (B) cell suspension solution, or may be contained separately from the (B) cell suspension solution. Examples of such carriers and additives include isotonic agents, thickeners, sugars, sugar alcohols, preservatives (preservatives), bactericides or antibacterial agents, pH regulators, stabilizers, chelating agents. , Oily base, gel base, surfactant, suspending agent, binder, excipient, lubricant, disintegrant, foaming agent, fluidizer, dispersant, emulsifier, buffer, solubilizer , Antioxidants, sweeteners, sour agents, colorants, flavoring agents, fragrances or refreshing agents, but are not limited thereto.
 本発明の細胞医薬組成物は、目的に応じて種々の形態、例えば、注射剤(輸液剤、埋め込み注射剤、持続性注射、用時調製型の注射剤を含む)、透析用剤、貼付剤、パップ剤等の形態で利用できる。本発明の細胞医薬組成物は噴霧により、患部に適用することもでき、本発明の細胞医薬組成物は噴霧した後に患部でゲル化もしくはシート化される形態でも利用できる。本発明の細胞医薬組成物は上記(A)細胞をシート状または立体構造体とした後に、患部に適用することもできる。 The cell pharmaceutical composition of the present invention can be used in various forms depending on the purpose, for example, injections (including infusions, implantable injections, continuous injections, injections prepared at the time of use), dialysis agents, patches. It can be used in the form of a poultice. The cell pharmaceutical composition of the present invention can also be applied to the affected area by spraying, and the cell pharmaceutical composition of the present invention can also be used in the form of gelation or sheeting at the affected area after spraying. The cell pharmaceutical composition of the present invention can also be applied to the affected area after the (A) cell is made into a sheet or a three-dimensional structure.
 本発明の細胞医薬組成物は、(A)細胞、(B)細胞懸濁用溶液、他の薬剤、その他の成分が別々の容器に封入されて保管され、使用時にこれらを混合して用いてもよい。なお、保管の際、上記(A)細胞、(B)細胞懸濁用溶液、他の薬剤、その他の成分はそれぞれに適した条件で保管されていればよく、例えば凍結条件、冷蔵条件、室温条件等いずれであってもよい。 In the cell pharmaceutical composition of the present invention, (A) cells, (B) cell suspension solutions, other drugs, and other components are sealed and stored in separate containers, and are used by mixing them at the time of use. Also good. In addition, at the time of storage, the above (A) cells, (B) cell suspension solution, other drugs, and other components may be stored under conditions suitable for each, for example, freezing conditions, refrigerated conditions, room temperature Any of conditions etc. may be sufficient.
 本発明の細胞医薬組成物のpHは、医薬上、薬理学的に(製薬上)又は生理学的に許容される範囲内であれば特に限定されるものではないが、一例として、2.0~9.0、好ましくは2.5~8.5、より好ましくは4.0~8.0となる範囲が挙げられる。 The pH of the cell pharmaceutical composition of the present invention is not particularly limited as long as it is within a pharmaceutically, pharmacologically (pharmaceutical) or physiologically acceptable range. The range is 9.0, preferably 2.5 to 8.5, and more preferably 4.0 to 8.0.
 本発明の細胞医薬組成物の浸透圧については、生体に許容される範囲内であれば、特に制限されない。本発明の細胞医薬組成物の浸透圧比の一例として、好ましくは0.6~1.5、より好ましくは0.7~1.2、さらに好ましくは0.8~1.0となる範囲が挙げられる。浸透圧の調整は上述した電解質等を用いて、当該技術分野で既知の方法で行うことができる。浸透圧比は、第十五改正日本薬局方に基づき286mOsm(0.9w/v%塩化ナトリウム水溶液)の浸透圧に対する試料の浸透圧の比とし、浸透圧は日本薬局方記載の浸透圧測定法(氷点降下法)を参考にして測定する。なお、浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)は、塩化ナトリウム(日本薬局方標準試薬)を500~650℃で40~50分間乾燥した後、デシケーター(シリカゲル)中で放冷し、その0.900gを正確に量り、精製水に溶かし正確に100mLとして調製するか、市販の浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)を用いる。 The osmotic pressure of the cell pharmaceutical composition of the present invention is not particularly limited as long as it is within the range acceptable to the living body. An example of the osmotic pressure ratio of the cell pharmaceutical composition of the present invention is preferably in the range of 0.6 to 1.5, more preferably 0.7 to 1.2, and still more preferably 0.8 to 1.0. It is done. The osmotic pressure can be adjusted by a method known in the technical field using the above-described electrolyte or the like. The osmotic pressure ratio is the ratio of the osmotic pressure of the sample to the osmotic pressure of 286 mOsm (0.9 w / v% sodium chloride aqueous solution) based on the 15th revised Japanese Pharmacopoeia. Measure by referring to the freezing point method. The standard solution for osmotic pressure ratio measurement (0.9 w / v% sodium chloride aqueous solution) was prepared by drying sodium chloride (Japanese Pharmacopoeia standard reagent) at 500 to 650 ° C. for 40 to 50 minutes, and then in a desiccator (silica gel). The mixture is allowed to cool and 0.900 g is accurately weighed and dissolved in purified water to make exactly 100 mL, or a commercially available standard solution for osmotic pressure ratio measurement (0.9 w / v% sodium chloride aqueous solution) is used.
 本発明の細胞医薬組成物における(A)細胞の濃度、即ち(A)細胞を(B)細胞懸濁用溶液に懸濁して調製し投与に用いる際の濃度は、細胞の種類、細胞懸濁用溶液によって異なりうるが、通常、1x10~2.5x10個/mL、好ましく1x10~2.5x10個/mL、より好ましくは1x10~2.5x10個/mLである。 The concentration of (A) cells in the cell pharmaceutical composition of the present invention, that is, the concentration when (A) the cells are prepared by suspending them in the (B) cell suspension solution and used for administration is the cell type, cell suspension Although it may vary depending on the solution used, it is usually 1 × 10 2 to 2.5 × 10 8 cells / mL, preferably 1 × 10 3 to 2.5 × 10 7 cells / mL, more preferably 1 × 10 4 to 2.5 × 10 6 cells / mL.
 本発明の細胞医薬組成物の好ましい一形態としては、ヒト間葉系幹細胞を細胞懸濁用溶液中に1×10~2.5×10個/mLの密度で含んでなり、細胞懸濁用溶液が125~150mEq/Lのナトリウムイオン、100~160mEq/Lの塩素イオン、3~5mEq/Lのカリウムイオン及び2~6mEq/Lのカルシウムイオンを含み、生理食塩水に対する浸透圧比が0.8~1.0であり、pHが4.0~8.0のものである。 As a preferred form of the cell pharmaceutical composition of the present invention, human mesenchymal stem cells are contained in a cell suspension solution at a density of 1 × 10 4 to 2.5 × 10 6 cells / mL, The turbid solution contains 125 to 150 mEq / L sodium ion, 100 to 160 mEq / L chloride ion, 3 to 5 mEq / L potassium ion and 2 to 6 mEq / L calcium ion, and has an osmotic pressure ratio of 0 to physiological saline. And a pH of 4.0 to 8.0.
 本発明の細胞医薬組成物の対象への投与経路は、皮下投与、筋肉内投与、静脈内投与、動脈内投与、髄腔内投与、腹腔内投与、経直腸投与、経腟投与、経皮投与、インプラント、臓器への直接投与等が挙げられるが、本発明の細胞医薬組成物の有効性の観点から、好ましくはインプラント、動脈内投与、静脈内投与及び臓器への直接投与であり、更に好ましくは静脈内投与及び臓器への直接投与である。 The administration route to the subject of the cell pharmaceutical composition of the present invention includes subcutaneous administration, intramuscular administration, intravenous administration, intraarterial administration, intrathecal administration, intraperitoneal administration, rectal administration, vaginal administration, and transdermal administration. From the viewpoint of the effectiveness of the cell pharmaceutical composition of the present invention, implant, intraarterial administration, intravenous administration, and direct administration to an organ are more preferable. Are intravenous and direct administration to the organ.
 本発明の細胞医薬組成物の対象への投与速度は、患者の状態(体重、年齢、症状、体調等)、及び本発明の非アルコール性脂肪肝炎治療剤の投与経路等によって異なりうるが、通常、成人に投与する場合には、50mL/h~1,000mL/hであり、75mL/h~500mL/hであることが好ましく、100mL/h~250mL/hであることがより好ましい。 The administration rate of the cell pharmaceutical composition of the present invention to the subject may vary depending on the patient's condition (weight, age, symptoms, physical condition, etc.) and the administration route of the non-alcoholic steatohepatitis therapeutic agent of the present invention. When administered to an adult, it is 50 mL / h to 1,000 mL / h, preferably 75 mL / h to 500 mL / h, and more preferably 100 mL / h to 250 mL / h.
 本発明の細胞医薬組成物の対象への投与温度は、患者の状態(体重、年齢、症状、体調等)、及び本発明の細胞医薬組成物の投与経路等によって異なりうるが、通常、4℃~45℃であり、15℃~37℃であることが好ましく、室温~37℃であることがより好ましい。 The administration temperature of the cell pharmaceutical composition of the present invention to the subject may vary depending on the patient's condition (body weight, age, symptoms, physical condition, etc.), the administration route of the cell pharmaceutical composition of the present invention, etc. ˜45 ° C., preferably 15 ° C. to 37 ° C., more preferably room temperature to 37 ° C.
 本発明の細胞医薬組成物は、輸液セットを用いて、対象への投与を行う事ができる。具体的に輸液セットとしては、ワンド・ディスポーザブル輸液チューブセット(株式会社吉田製作所製)、輸液セット(フォルテグロウメディカル株式会社製)、テルヒュージョン(登録商標)輸液セット(テルモ株式会社製)、JMS輸液セット(株式会社ジェイ・エム・エス製)、シュアプラグ輸液セット(テルモ株式会社製)、輸液セット(ニプロ株式会社製)、トップ輸液セットNP(株式会社トップ製)、フィルター付き輸液セット(EXタイプ)(東レ・メディカル株式会社製)等の市販品を用いることもできる。 The cell pharmaceutical composition of the present invention can be administered to a subject using an infusion set. Specifically, as an infusion set, a wand disposable infusion tube set (manufactured by Yoshida Seisakusho Co., Ltd.), an infusion set (manufactured by Fortegro Medical Co., Ltd.), a terfusion (registered trademark) infusion set (manufactured by Terumo Corporation), a JMS infusion Set (manufactured by JMS Co., Ltd.), Sure plug infusion set (manufactured by Terumo Corporation), Infusion set (manufactured by Nipro Corporation), Top infusion set NP (manufactured by Top Co., Ltd.), Infusion set with filter (EX type ) (Manufactured by Toray Medical Co., Ltd.) can also be used.
 本発明の細胞医薬組成物は、輸液チューブを用いて、対象への投与を行う事ができる。具体的に輸液チューブとしては、レクトロ・キャス(株式会社サミック・インターナショナル製)、JMSエキステンションチューブ(株式会社ジェイ・エム・エス製)、サフィード延長チューブ(テルモ株式会社製)、延長チューブ(株式会社トップ製)、コネクティングチューブ(チュウアツ)(株式会社メディコスヒラタ)、サフティAPチューブ(川澄化学工業株式会社製)、延長チューブ付ビオネクター2(東レ・メディカル株式会社)、メディカットエクステンションチューブセット B(日本シャーウッド株式会社製)、ワンド・ディスポーザブル輸液チューブセット(株式会社吉田製作所製)、輸液チューブ(フォルテグロウメディカル株式会社製)等の市販品を用いることもできる。 The cell pharmaceutical composition of the present invention can be administered to a subject using an infusion tube. Specifically, as an infusion tube, Lectro Cass (manufactured by Samick International Co., Ltd.), JMS extension tube (manufactured by JMS Co., Ltd.), Saffy extension tube (manufactured by Terumo Corporation), extension tube (manufactured by Co., Ltd.) Top product), Connecting tube (Chuatsu) (Medicos Hirata Co., Ltd.), SAFTY AP tube (manufactured by Kawasumi Chemical Industry Co., Ltd.), Bioconnector 2 with extension tube (Toray Medical Co., Ltd.), Medicut Extension Tube Set B (Nippon Sherwood) Commercially available products such as Wand Disposable Infusion Tube Set (manufactured by Yoshida Seisakusho Co., Ltd.), Infusion Tube (manufactured by Forte Grow Medical Co., Ltd.), and the like can also be used.
 本発明の細胞医薬組成物を対象へ投与する際に用いる輸液チューブの材質としては、ポリ塩化ビニル、熱可塑性エラストマー、TPE 熱可塑性エラストマー、シリコーン、シリコーンゴム、ポリエチレン、ポリブタジエン、テフロン(登録商標)、ポリウレタン、ポリプロピレン、天然ゴム、ポリオレフィン、PVC(可塑剤:TOTM、DOA)、無可塑剤PVC及びこれらの混合物を用いることができる。 The material of the infusion tube used when administering the cell pharmaceutical composition of the present invention to a subject includes polyvinyl chloride, thermoplastic elastomer, TPE thermoplastic elastomer, silicone, silicone rubber, polyethylene, polybutadiene, Teflon (registered trademark), Polyurethane, polypropylene, natural rubber, polyolefin, PVC (plasticizer: TOTM, DOA), plasticizer PVC, and mixtures thereof can be used.
 本発明の細胞医薬組成物は、種々の疾患の治療に好適に用いることができる。例えば、内臓疾患、具体的には、心疾患、胃・十二指腸疾患、小腸・大腸疾患、肝疾患、胆道疾患、膵疾患、腎疾患、肺疾患、縦隔膜疾患、横隔膜疾患、胸膜疾患、腹膜疾患、神経疾患、中枢神経系(CNS)障害、末梢動脈疾患、末梢静脈疾患に対して用いることが好ましい。 The cell pharmaceutical composition of the present invention can be suitably used for the treatment of various diseases. For example, visceral diseases, specifically heart disease, stomach / duodenal disease, small intestine / colon disease, liver disease, biliary disease, pancreatic disease, kidney disease, lung disease, mediastinal disease, diaphragm disease, pleural disease, peritoneal disease It is preferably used for neurological diseases, central nervous system (CNS) disorders, peripheral arterial diseases, and peripheral venous diseases.
 具体的疾患としては、例えば、自己免疫性肝炎、劇症肝炎、慢性肝炎、ウイルス性肝炎、アルコール性肝炎、非アルコール性脂肪性肝疾患(nonalcoholic fatty liver disease(NAFLD))、非アルコール性脂肪肝炎(nonalcoholic steatohepatitis(NASH))、非アルコール性脂肪肝(nonalcoholic fatty liver (NAFL))、肝線維症、肝硬変、肝癌、脂肪肝、薬剤アレルギー性肝障害、ヘモクロマトーシス、ヘモジデローシス、ウィルソン病、原発性胆汁性肝硬変(PBC)、原発性硬化性胆管炎(PSC)、胆道閉鎖、肝膿瘍、慢性活動性肝炎、慢性持続性肝炎等の肝疾患;心筋梗塞、心不全、不整脈、動悸、心筋症、虚血性心筋症、狭心症、先天性心疾患、心臓弁膜症、心筋炎、家族性肥大型心筋症、拡張型心筋症、急性冠症候群、アテローム血栓症、再狭窄等の心疾患;急性胃炎、慢性胃炎、胃・十二指腸潰瘍、胃癌、十二指腸癌等の胃・十二指腸疾患;虚血性腸炎、炎症性腸疾患、潰瘍性大腸炎、Crohn病、単純性潰瘍、腸管ベーチェット病、小腸癌、大腸癌等の小腸・大腸疾患;急性胆嚢炎、急性胆管炎、慢性胆嚢炎、胆管癌、胆嚢癌等の胆道疾患;急性膵炎、慢性膵炎、膵癌等の膵疾患;急性腎炎、慢性腎炎、急性腎不全、慢性腎不全等の腎疾患;肺炎、肺気腫、肺線維症、間質性肺炎、特発性間質性肺炎、剥離性間質性肺炎、急性間質性肺炎、非特異的間質性肺炎、薬物誘発性肺疾患、好酸球性肺疾患、肺高血圧症、肺結核、肺結核後遺症、急性呼吸窮迫症候群、嚢胞性線維症、慢性閉塞性肺疾患、肺塞栓症、肺膿症、塵肺、嚥下性肺炎肺線維症、急性上気道感染症、慢性下気道感染症、気胸、肺胞上皮に傷害が見られる疾患、リンパ管平滑筋種、リンパ性間質性肺炎、肺胞蛋白症、肺ランゲルハンス細胞肉芽腫症等の肺疾患;縦隔腫瘍、縦隔の嚢胞性疾患、縦隔炎等の縦隔膜疾患;横隔膜ヘルニア等の横隔膜疾患;胸膜炎、膿胸、胸膜腫瘍、がん性胸膜炎、胸膜中皮腫等の胸膜疾患;腹膜炎、腹膜腫瘍等の腹膜疾患;小児脳性麻痺を含む脳性麻痺症候群、無菌性髄膜炎、ギランーバレー症候群、筋萎縮性側索硬化症(ALS)、重症筋無力症、モノニューロパシー、多発ニューロパシー、脊髄性筋萎縮症、脊椎障害、急性横断性脊髄炎、脊髄梗塞(虚血性脊髄障害)、頭蓋内腫瘍、脊椎腫瘍等の神経疾患;Alzheimer病、認知障害、脳卒中、多発性硬化症、Parkinson病等のCNS障害;線維筋性異形成、末梢動脈疾患(PAD)、閉塞性血栓血管炎(ビュルガー病)、川崎病(KD)等の末梢動脈疾患;深部静脈血栓症、慢性静脈不全、静脈炎後症候群、表在性静脈血栓症等の末梢静脈疾患;移植片対宿主病(GVHD)、続発性免疫不全症、原発性免疫不全疾患、B細胞の欠損、T細胞不全、BおよびT細胞複合欠損、食細胞欠損、古典経路における補体欠損、MBL経路における補体欠損、代替経路における補体欠損、補体調節蛋白欠損、補体レセプター欠損等の免疫不全疾患が挙げられる。 Specific diseases include, for example, autoimmune hepatitis, fulminant hepatitis, chronic hepatitis, viral hepatitis, alcoholic hepatitis, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (Nonalcoholic steatohepatitis (NASH)), non-alcoholic fatty liver (NAFL), liver fibrosis, cirrhosis, liver cancer, fatty liver, drug allergic liver disorder, hemochromatosis, hemosiderosis, Wilson's disease, primary Hepatic diseases such as biliary cirrhosis (PBC), primary sclerosing cholangitis (PSC), biliary atresia, liver abscess, chronic active hepatitis, chronic persistent hepatitis; myocardial infarction, heart failure, arrhythmia, palpitations, cardiomyopathy, false blood Cardiomyopathy, angina, congenital heart disease, valvular heart disease, myocarditis, familial hypertrophic cardiomyopathy, dilated cardiomyopathy, acute coronary syndrome, atherothrombosis, restenosis, etc .; acute gastritis, chronic Gastritis, gastric / duodenal ulcer, gastric cancer, duodenal cancer and other gastric / duodenal diseases; ischemic enteritis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, simple ulcer, intestinal Behcet's disease, small intestine cancer, colon cancer, etc. Small intestine / colon disease; biliary tract diseases such as acute cholecystitis, acute cholangitis, chronic cholecystitis, cholangiocarcinoma, gallbladder cancer; pancreatic diseases such as acute pancreatitis, chronic pancreatitis, pancreatic cancer; acute nephritis, chronic nephritis, acute renal failure, chronic Renal diseases such as renal failure; pneumonia, emphysema, pulmonary fibrosis, interstitial pneumonia, idiopathic interstitial pneumonia, exfoliative interstitial pneumonia, acute interstitial pneumonia, nonspecific interstitial pneumonia, drug induction Lung disease, eosinophilic lung disease, pulmonary hypertension, pulmonary tuberculosis, pulmonary tuberculosis sequelae, acute Sucking syndrome, cystic fibrosis, chronic obstructive pulmonary disease, pulmonary embolism, pulmonary abscess, pneumoconiosis, swallowing pneumonia pulmonary fibrosis, acute upper respiratory tract infection, chronic lower respiratory tract infection, pneumothorax, alveolar epithelium Injured diseases, lymphatic smooth muscle species, lymphoid interstitial pneumonia, alveolar proteinosis, pulmonary Langerhans cell granulomatosis, etc .; mediastinal tumor, mediastinal cystic disease, mediastinitis, etc. Mediastinal diseases; Diaphragmatic diseases such as diaphragmatic hernia; Pleural diseases such as pleurisy, pyothorax, pleural tumor, cancerous pleurisy, pleural mesothelioma; Peritoneal diseases such as peritonitis, peritoneal tumor; Aseptic meningitis, Guillain-Barre syndrome, amyotrophic lateral sclerosis (ALS), myasthenia gravis, mononeuropathy, polyneuropathy, spinal muscular atrophy, spinal disorder, acute transverse myelitis, spinal cord infarction (false) Hematologic spinal cord disorder), intracranial tumor, spine Neurological diseases such as tumors; CNS disorders such as Alzheimer's disease, cognitive impairment, stroke, multiple sclerosis, Parkinson's disease; fibromyalplasia, peripheral arterial disease (PAD), obstructive thromboangitis (Burger's disease), Kawasaki Peripheral arterial disease such as KD disease; deep vein thrombosis, chronic venous insufficiency, peripheral venous disease such as post-phlebitis syndrome, superficial venous thrombosis; graft-versus-host disease (GVHD), secondary immune deficiency Primary immune deficiency disease, B cell deficiency, T cell deficiency, B and T cell complex deficiency, phagocytic deficiency, complement deficiency in classical pathway, complement deficiency in MBL pathway, complement deficiency in alternative pathway, complement Examples include immunodeficiency diseases such as regulatory protein deficiency and complement receptor deficiency.
 これらのうち、間葉系幹細胞による治療効果が十分に得られることが確認されている肝疾患、心疾患、肺疾患、神経疾患、末梢動脈疾患、免疫不全疾患が好ましく、中でも、肝線維症、肝硬変、心筋梗塞、心不全、肺線維症、間質性肺炎、小児脳性麻痺、筋萎縮性側索硬化症(ALS)、末梢動脈疾患(PAD)、移植片対宿主病(GVHD)の治療により好適に用いることができ、肝線維症、肝硬変、心筋梗塞、心不全、肺線維症、間質性肺炎にさらに好適に用いることができる。また、末梢血単核球による治療効果が十分に得られることが確認されている各組織の癌に好適に用いることができる。 Among these, liver diseases, heart diseases, lung diseases, neurological diseases, peripheral arterial diseases, immunodeficiency diseases that have been confirmed to have sufficient therapeutic effects by mesenchymal stem cells are preferred, among which liver fibrosis, Suitable for the treatment of cirrhosis, myocardial infarction, heart failure, pulmonary fibrosis, interstitial pneumonia, childhood cerebral palsy, amyotrophic lateral sclerosis (ALS), peripheral arterial disease (PAD), graft-versus-host disease (GVHD) And can be used more suitably for liver fibrosis, cirrhosis, myocardial infarction, heart failure, pulmonary fibrosis, and interstitial pneumonia. Moreover, it can use suitably for the cancer of each structure | tissue where it is confirmed that the therapeutic effect by a peripheral blood mononuclear cell is fully acquired.
<疾患治療用キット>
 本発明は、(A)細胞、及び(B)細胞懸濁用溶液を含有し、(B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、疾患治療用キットも含む。本発明の疾患治療用キットは、上述した本発明の細胞医薬組成物を含むキットであり、(A)細胞、(B)細胞懸濁用溶液、その他本発明の細胞医薬組成物が含有し得る成分については細胞医薬組成物の項における説明を適用できる。本発明の疾患治療用キットによれば、細胞の状態を良好に保ち、生存率を長時間に渡って高く維持することができるため、様々な疾患に対して優れた治療効果を奏することができる。
<Disease treatment kit>
The present invention includes (A) a cell and (B) a cell suspension solution, (B) the cell suspension solution contains Na + , Cl , K + and Ca 2+ as an electrolyte, and a sugar Also included is a disease treatment kit that is an isotonic electrolyte without quality. The disease treatment kit of the present invention is a kit containing the above-described cell pharmaceutical composition of the present invention, and may contain (A) cells, (B) cell suspension solutions, and other cell pharmaceutical compositions of the present invention. The description in the section of the cell pharmaceutical composition can be applied to the components. According to the disease treatment kit of the present invention, since the state of cells can be kept good and the survival rate can be maintained high over a long period of time, an excellent therapeutic effect can be obtained for various diseases. .
 また、本発明の疾患治療用キットは、本発明の細胞医薬組成物、容器及びラベルを含むものであると表現することもできる。本発明の疾患治療用キットが含む適切な容器としては、特に限定されないが、例えば、細胞凍結用のクライオチューブ、細胞懸濁用溶液用のボトル、バイアル、試験管、透析バック等が挙げられる。これらの容器は、ガラス、金属、プラスチック又はこれらの組み合わせ等の多様な材料から形成されていてもよい。これらの容器上のラベルには、内容物である細胞、細胞懸濁用溶液等を説明する内容が記載されている。 The disease treatment kit of the present invention can also be expressed as comprising the cell pharmaceutical composition, container and label of the present invention. Suitable containers included in the disease treatment kit of the present invention are not particularly limited, and examples include cryotubes for freezing cells, bottles for cell suspension solutions, vials, test tubes, and dialysis bags. These containers may be formed from a variety of materials such as glass, metal, plastic, or combinations thereof. The contents on the labels on these containers describe the contents of the cells, the solution for cell suspension, and the like.
 本発明の疾患治療用キットは、その他の添加剤、その他の薬剤、希釈剤、フィルター、針、シリンジ、使用法を記載した添付文書を含めた、商業的、及び利用者の観点から望ましい他の材料を包含することができる。 The disease treatment kit of the present invention includes other additives, other drugs, diluents, filters, needles, syringes, and other packages that are desirable from a commercial and user standpoint, including package inserts that describe usage. Material can be included.
<細胞懸濁用溶液>
 電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、細胞医薬組成物用の細胞懸濁用溶液も本発明の範囲内である。細胞医薬品の注射・点滴用の細胞懸濁用溶液として、Na、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液を採用することで、細胞の生存率を長時間に渡って高く維持することができることは、本発明者らが新たに見出した知見である。細胞懸濁用溶液の詳細については、細胞医薬組成物の項における(B)細胞懸濁用溶液の説明を適用できる。
<Cell suspension solution>
Also within the scope of the present invention is a solution for cell suspension for a cell pharmaceutical composition, which is an isotonic electrolyte containing Na + , Cl , K + and Ca 2+ as an electrolyte and free of carbohydrates. is there. By adopting an isotonic electrolyte solution containing Na + , Cl , K + and Ca 2+ and not containing carbohydrates as a cell suspension solution for cell medicine injection / infusion, It is a finding newly found by the present inventors that the survival rate can be maintained high over a long period of time. For the details of the cell suspension solution, the description of (B) the cell suspension solution in the section of the cell pharmaceutical composition can be applied.
<疾患の治療方法>
 本発明は、(A)細胞を、(B)細胞懸濁用溶液に懸濁して患者に投与する疾患の治療方法であって、(B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液であることを特徴とする治療方法も含む。本発明の治療方法によると、細胞の生存率を長時間に渡って高く維持することができるため、様々な疾患に対して優れた治療効果を奏することができる。本発明の疾患方法は、上述した本発明の細胞医薬組成物を用いた治療方法であり、(A)細胞、(B)細胞懸濁用溶液、その他本発明の細胞医薬組成物が含有し得る成分については細胞医薬組成物の項における説明を適用できる。
<Disease treatment method>
The present invention relates to a method for treating a disease in which (A) cells are suspended in (B) a cell suspension solution and administered to a patient, and (B) the cell suspension solution contains Na + , Cl as an electrolyte. - includes a K + and Ca 2+, and does not contain carbohydrates, including methods of treatment wherein the isotonic electrolyte solution. According to the treatment method of the present invention, it is possible to maintain a high cell survival rate over a long period of time, and therefore, an excellent therapeutic effect can be achieved for various diseases. The disease method of the present invention is a treatment method using the above-described cell pharmaceutical composition of the present invention, and may contain (A) cells, (B) a cell suspension solution, and other cell pharmaceutical compositions of the present invention. The description in the section of the cell pharmaceutical composition can be applied to the components.
 以下に、実施例及び試験例を挙げて本発明を詳細に説明するが、本発明はこれらの実施例等によって限定されるものではない。 Hereinafter, the present invention will be described in detail with reference to Examples and Test Examples, but the present invention is not limited to these Examples and the like.
[実施例1]
(脂肪由来間葉系幹細胞の調製)
 ヒトドナーから同意を得た後、脂肪吸引法で得た皮下脂肪組織を生理食塩液で洗浄した。細胞外基質の破壊、及び細胞の単離を達成するために、コラゲナーゼ(Roche diagnostics社)(溶媒は生理食塩液)を添加し、37℃で90分間振倒し、分散した。続いて、この上記懸濁液を800gで5分間、遠心分離して間質血管細胞群の沈殿を得た。上記細胞の沈殿に間葉系幹細胞用無血清培地(Rohto社)を加え、当該細胞懸濁液を400gで5分間遠心分離し、上清除去後に間葉系幹細胞用無血清培地(Rohto社)に再懸濁し、フラスコに細胞を播種した。細胞を37℃で数日間、5%CO中で培養した。数日後に培養物をPBSで洗浄して、培養液中に含まれていた血球や脂肪組織の残存等を除去し、プラスチック容器に接着している間葉系幹細胞を得た。
[Example 1]
(Preparation of adipose-derived mesenchymal stem cells)
After obtaining consent from a human donor, the subcutaneous adipose tissue obtained by the liposuction method was washed with physiological saline. In order to achieve destruction of the extracellular matrix and isolation of the cells, collagenase (Roche diagnostics) (solvent was physiological saline) was added, shaken at 37 ° C. for 90 minutes, and dispersed. Subsequently, the suspension was centrifuged at 800 g for 5 minutes to obtain a precipitate of stromal vascular cell groups. A serum-free medium for mesenchymal stem cells (Rohto) is added to the cell precipitate, the cell suspension is centrifuged at 400 g for 5 minutes, and after removal of the supernatant, a serum-free medium for mesenchymal stem cells (Rohto) The cells were seeded in a flask. Cells were cultured at 37 ° C. for several days in 5% CO 2 . Several days later, the culture was washed with PBS to remove residual blood cells and adipose tissue contained in the culture solution, and mesenchymal stem cells adhered to a plastic container were obtained.
 得られた脂肪由来間葉系幹細胞を遠沈管に分注し、400gで5分間、遠心分離し細胞の沈殿を得た。上清を除去した後、細胞凍結保存液(STEM-CELLBANKER(ゼノアック社))を適量加え懸濁した。当該細胞懸濁液を、クライオチューブに分注した後、フリーザー内で-80度にて保存後、液体窒素上の気相に移し、保存を継続した。 The obtained adipose-derived mesenchymal stem cells were dispensed into a centrifuge tube and centrifuged at 400 g for 5 minutes to obtain cell precipitates. After removing the supernatant, an appropriate amount of a cell cryopreservation solution (STEM-CELLBANKER (Zenoac)) was added and suspended. The cell suspension was dispensed into a cryotube, stored at −80 ° C. in a freezer, transferred to a gas phase on liquid nitrogen, and storage was continued.
 15mL遠沈管(住友ベークライト株式会社,品番:MS-56150)に、リンゲル液(リンゲル液「オーツカ」、株式会社大塚製薬工場、Lot:K4K73)、KN2号輸液(2号液(脱水補給液))(株式会社大塚製薬工場、Lot:K6E92)、KN3号輸液(3号液(維持液))(株式会社大塚製薬工場、Lot:K6D96)、KN4号輸液(4号液(術後回復液))(株式会社大塚製薬工場、Lot:K6D80)及びフィジオ(登録商標)70(2.5%ブドウ糖加酢酸リンゲル液)(株式会社大塚製薬工場、Lot:M6D91)をそれぞれ5mLずつ分注した後に、凍結保存された脂肪組織由来間葉系幹細胞を湯浴(37±1℃)で急速融解後、脂肪組織由来間葉系幹細胞の細胞懸濁液を62.5μLずつ加えた。転倒混和により懸濁した後、室温で保管し、調製直後、2時間後、4時間後、及び7時間後に21Gの注射針付5mLシリンジで中層より1mL分取し、1.5mLチューブに移した。細胞懸濁液10μLに対して、トリパンブルー(Trypan Blue Stain(0.4%);Life technologies、15250-061)10μLを加えて、生細胞及び死細胞を区別して位相差顕微鏡(OLYMPUS、品番:CKX41SF)にて計測を行った。なお、細胞のカウントにはディスポーザブル細胞計算盤(WAKEN、品番:WC2-100)を用い、18区画カウントを5回行い、最大の数値と最少の数値を除いた3回の計測値の平均値を用いて、下記式により細胞生存率を算出した。なお、4時間後において細胞生存率が70%を下回ったものについては、7時間後の計測を行わなかった。各輸液の組成を下記表1に、細胞生存率の結果を下記表2に示した。
 
細胞生存率(%)=生細胞数/総細胞数×100
 
In a 15 mL centrifuge tube (Sumitomo Bakelite Co., Ltd., product number: MS-56150), Ringer solution (Ringer solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73), KN2 infusion solution (No. 2 solution (dehydration supplement)) (stock) Company Otsuka Pharmaceutical Factory, Lot: K6E92), KN3 Infusion (No.3 Liquid (Maintenance Solution)) (Otsuka Pharmaceutical Factory, Lot: K6D96), KN4 Infusion (No.4 Liquid (Postoperative Recovery Solution)) (Stock) Company Otsuka Pharmaceutical Factory, Lot: K6D80) and Physio 70 (2.5% glucose-acetated Ringer's solution) (Otsuka Pharmaceutical Factory, Lot: M6D91) were each dispensed in 5 mL, and stored frozen. Adipose tissue-derived mesenchymal stem cells were rapidly thawed in a hot water bath (37 ± 1 ° C.), and then 62.5 μL of a cell suspension of adipose tissue-derived mesenchymal stem cells was added. Suspended by inversion and stored at room temperature. Immediately after preparation, 1 mL was taken from the middle layer with a 5 mL syringe with a 21 G injection needle immediately after preparation, 2 hours, 4 hours, and 7 hours, and transferred to a 1.5 mL tube. . To 10 μL of the cell suspension, 10 μL of trypan blue (Trypan Blue Stain (0.4%); Life technologies, 15250-061) was added to distinguish between live and dead cells, and a phase contrast microscope (OLYMPUS, product number: Measurement was performed with CKX41SF). In addition, the cell count is performed using a disposable cell counter (WAKEN, product number: WC2-100), 18 counts are performed 5 times, and the average value of the 3 measured values excluding the maximum value and the minimum value is calculated. The cell viability was calculated by the following formula. For cells whose cell viability fell below 70% after 4 hours, measurement after 7 hours was not performed. The composition of each infusion solution is shown in Table 1 below, and the results of cell viability are shown in Table 2 below.

Cell viability (%) = number of living cells / total number of cells × 100
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002
 KN2号輸液及びフィジオ(登録商標)70に脂肪由来間葉系幹細胞を懸濁した場合に、4時間後の細胞生存率が70%を下回った。KN3号輸液及びKN4号輸液に脂肪由来間葉系幹細胞を懸濁した場合に、7時間後において細胞生存率が70%を下回った。これに対して、リンゲル液に脂肪由来間葉系幹細胞を懸濁した場合には、7時間後においても細胞生存率が80%以上であった。以上の結果から、リンゲル液に脂肪由来間葉系幹細胞を懸濁することにより、細胞生存率を顕著に高い状態で長時間に渡って維持できることがわかった。 When fat-derived mesenchymal stem cells were suspended in KN2 infusion and Physio (registered trademark) 70, the cell viability after 4 hours was less than 70%. When fat-derived mesenchymal stem cells were suspended in KN3 infusion and KN4 infusion, cell viability fell below 70% after 7 hours. In contrast, when fat-derived mesenchymal stem cells were suspended in Ringer's solution, the cell viability was 80% or more even after 7 hours. From the above results, it was found that by suspending fat-derived mesenchymal stem cells in Ringer's solution, the cell viability can be maintained for a long time in a significantly high state.
[実施例2]
 15mL遠沈管(住友ベークライト株式会社、品番:MS-56150)に、リンゲル液(リンゲル液「オーツカ」、株式会社大塚製薬工場、Lot:K4K73)、重炭酸リンゲル液(ビカネイト(登録商標)輸液、株式会社大塚製薬工場)をそれぞれ5mLずつ分注した後に、実施例1と同様に、凍結保存された脂肪由来間葉系幹細胞を湯浴(37±1℃)で急速融解後脂肪由来間葉系幹細胞の細胞懸濁液を62.5μLずつ加えた。転倒混和により懸濁した後、室温で保管し、調製直後、1時間後、2時間後及び4時間後の生細胞数及び死細胞数を実施例1と同様に測定して細胞生存率を算出し、調製直後に対しての細胞生存率を算出した。各リンゲル液の組成を下記表3に、細胞生存率の結果を下記表4に示した。
[Example 2]
In a 15 mL centrifuge tube (Sumitomo Bakelite Co., Ltd., product number: MS-56150), Ringer's solution (Ringer's solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73), bicarbonate Ringer's solution (Bikanate (registered trademark) infusion, Otsuka Pharmaceutical Co., Ltd.) After dispensing 5 mL each of the plant), in the same manner as in Example 1, the fat-derived mesenchymal stem cells that had been cryopreserved were rapidly thawed in a hot water bath (37 ± 1 ° C.) and then the cell suspension of the fat-derived mesenchymal stem cells was The suspension was added in 62.5 μL aliquots. Suspend by inversion and store at room temperature. Calculate the cell viability by measuring the number of living and dead cells immediately after preparation, 1 hour, 2 hours and 4 hours in the same manner as in Example 1. The cell viability with respect to immediately after preparation was calculated. The composition of each Ringer's solution is shown in Table 3 below, and the results of cell viability are shown in Table 4 below.
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004
 リンゲル液だけでなく、重炭酸リンゲル液に脂肪由来間葉系幹細胞を懸濁した場合には、4時間後を含むすべての保存時間において、細胞生存率が90%程度と高かった。以上の結果から、リンゲル液及び重炭酸リンゲル液に脂肪由来間葉系幹細胞を懸濁することにより、細胞生存率を顕著に高い状態で長時間に渡って維持できることがわかった。 When not only Ringer's solution but also fat-derived mesenchymal stem cells were suspended in bicarbonate Ringer's solution, the cell viability was as high as about 90% in all the storage times including 4 hours later. From the above results, it was found that by suspending fat-derived mesenchymal stem cells in Ringer's solution and bicarbonated Ringer's solution, cell viability can be maintained for a long time in a significantly high state.
[実施例3]
 実施例1の脂肪由来間葉系幹細胞を骨髄由来間葉系幹細胞(ロンザ社製)に代え、リンゲル液(リンゲル液「オーツカ」、株式会社大塚製薬工場、Lot:K4K73)を用いて、同様に試験を行い、調製直後、2時間後、3時間後及び7時間後の生細胞数及び死細胞数を測定して細胞生存率を算出した。結果を下記表5に示す。
[Example 3]
The fat-derived mesenchymal stem cells of Example 1 were replaced with bone marrow-derived mesenchymal stem cells (Lonza), and the same test was performed using Ringer's solution (Ringer solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73). The cell viability was calculated by measuring the number of living cells and the number of dead cells immediately after preparation, 2 hours later, 3 hours later and 7 hours later. The results are shown in Table 5 below.
Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000005
 リンゲル液に骨髄由来間葉系幹細胞を懸濁した場合にも、7時間後において細胞生存率が95%以上であった。以上の結果より、リンゲル液に骨髄由来間質細胞を懸濁することにより、細胞生存率を顕著に高い状態で長時間に渡って維持できることがわかった。 Even when bone marrow-derived mesenchymal stem cells were suspended in Ringer's solution, the cell viability was 95% or more after 7 hours. From the above results, it was found that by suspending bone marrow-derived stromal cells in Ringer's solution, the cell viability can be maintained for a long time in a significantly high state.
[実施例4]
 実施例1の脂肪由来間葉系幹細胞を臍帯由来間葉系幹細胞(Lifeline Cell Technology、LifeLine(登録商標)_UCMSC、Lot.160907)に代え、リンゲル液(リンゲル液「オーツカ」、株式会社大塚製薬工場、Lot:K4K73)を用いて同様に試験を行い、調製直後、2時間後、4時間後及び6時間後の生細胞数及び死細胞数を測定し、実施例1と同様に細胞生存率を算出した。結果を下記表6に示す。
[Example 4]
The fat-derived mesenchymal stem cells of Example 1 were replaced with umbilical cord-derived mesenchymal stem cells (Lifeline Cell Technology, LifeLine (registered trademark) _UCMSC, Lot. 160907), Ringer's solution (Ringer solution “Otsuka”, Otsuka Pharmaceutical Factory, Ltd., Lot : K4K73), the number of viable cells and the number of dead cells were measured immediately after preparation, 2 hours, 4 hours and 6 hours, and the cell viability was calculated in the same manner as in Example 1. . The results are shown in Table 6 below.
Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006
 リンゲル液に臍帯由来間質細胞を懸濁した場合にも、6時間後において細胞生存率が90%以上であった。従って、リンゲル液は、臍帯由来間葉系幹細胞についても脂肪由来間葉系幹細胞と同様に、細胞生存率を顕著に高い状態で長時間に渡って維持できることがわかった。 Even when umbilical cord-derived stromal cells were suspended in Ringer's solution, the cell viability was 90% or more after 6 hours. Therefore, it was found that the Ringer's solution can maintain the cell viability for a long time in a state of significantly high umbilical cord-derived mesenchymal stem cells as well as the fat-derived mesenchymal stem cells.
[実施例5]
 実施例1の脂肪由来間葉系幹細胞を末梢血単核球細胞(ACCUCELL(登録商標)正常ドナー由来PBMC、Precision Bioservices社(PRECISION FOR MEDICINE社)製、Lot.13134-10)に代え、リンゲル液(リンゲル液「オーツカ」、株式会社大塚製薬工場、Lot:K4K73)を用いて同様に試験を行い、調製直後、2時間後、4時間後及び6時間後の生細胞数及び死細胞数を測定して細胞生存率を算出した。結果を下記表7に示す。
[Example 5]
The fat-derived mesenchymal stem cells of Example 1 were replaced with peripheral blood mononuclear cells (ACCUCELL (registered trademark) normal donor-derived PBMC, Precision Bioservices (PRECISION FOR MEDICINE), Lot. 13134-10), Ringer's solution ( The same test was conducted using Ringer's solution “Otsuka”, Otsuka Pharmaceutical Factory, Lot: K4K73), and the number of living cells and dead cells were measured immediately after preparation, 2 hours, 4 hours and 6 hours later. Cell viability was calculated. The results are shown in Table 7 below.
Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007
 リンゲル液に末梢血単核球細胞を懸濁した場合にも、6時間後において細胞生存率が80%程度であった。したがって、リンゲル液は、末梢血単核球細胞についても脂肪由来間葉系幹細胞と同様に、細胞生存率を顕著に高い状態で長時間に渡って維持できることがわかった。 Even when peripheral blood mononuclear cells were suspended in Ringer's solution, the cell viability was about 80% after 6 hours. Therefore, it was found that the Ringer's solution can maintain the cell survival rate of peripheral blood mononuclear cells over a long period of time with a remarkably high state as in the case of fat-derived mesenchymal stem cells.
 本発明の細胞医薬組成物は、細胞の状態を良好に保ち、その生存率を長時間に渡って高い状態で維持することができるため、様々な疾患に対して優れた治療効果が期待できる。 Since the cell pharmaceutical composition of the present invention can maintain a good cell state and maintain its survival rate in a high state for a long time, it can be expected to have an excellent therapeutic effect on various diseases.

Claims (7)

  1.  (A)細胞、及び
     (B)細胞懸濁用溶液
    を含有し、
     (B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、細胞医薬組成物。
    (A) cells, and (B) a cell suspension solution,
    (B) The cell pharmaceutical composition, wherein the cell suspension solution is an isotonic electrolytic solution containing Na + , Cl , K + and Ca 2+ as electrolytes and not containing carbohydrates.
  2.  (B)細胞懸濁用溶液が、CHCOO、HCO 、クエン酸イオン及び乳酸イオンからなる群より選択される少なくとも1種の電解質をさらに含む、請求項1に記載の細胞医薬組成物。 (B) The cell pharmaceutical composition according to claim 1, wherein the cell suspension solution further comprises at least one electrolyte selected from the group consisting of CH 3 COO , HCO 3 , citrate ions and lactate ions. object.
  3.  (B)細胞懸濁用溶液が、リンゲル液、酢酸リンゲル液、重炭酸リンゲル液及び乳酸リンゲル液からなる群より選択される少なくとも1種である、請求項1又は2に記載の細胞医薬組成物。 (B) The cell pharmaceutical composition according to claim 1 or 2, wherein the solution for cell suspension is at least one selected from the group consisting of Ringer's solution, acetated Ringer's solution, bicarbonated Ringer's solution and lactated Ringer's solution.
  4.  (A)細胞が、間葉系幹細胞又は末梢血単核球である、請求項1から3のいずれか1項に記載の細胞医薬組成物。 (A) The cell pharmaceutical composition according to any one of claims 1 to 3, wherein the cells are mesenchymal stem cells or peripheral blood mononuclear cells.
  5.  上記間葉系幹細胞が、脂肪由来、臍帯由来又は骨髄由来である、請求項4に記載の細胞医薬組成物。 The cell pharmaceutical composition according to claim 4, wherein the mesenchymal stem cells are derived from fat, umbilical cord or bone marrow.
  6.  (A)細胞、及び
     (B)細胞懸濁用溶液
    を含有し、
     (B)細胞懸濁用溶液が、電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、疾患治療用キット。
    (A) cells, and (B) a cell suspension solution,
    (B) The disease treatment kit, wherein the cell suspension solution is an isotonic electrolyte solution containing Na + , Cl , K + and Ca 2+ as electrolytes and not containing carbohydrates.
  7.  電解質としてNa、Cl、K及びCa2+を含み、かつ糖質を含まない、等張性の電解液である、細胞医薬組成物用の細胞懸濁用溶液。 A solution for cell suspension for a cell pharmaceutical composition, which is an isotonic electrolyte containing Na + , Cl , K + and Ca 2+ as an electrolyte and not containing a carbohydrate.
PCT/JP2017/044993 2016-12-28 2017-12-14 Cell pharmaceutical composition, disease treatment kit, and cell suspension solution WO2018123628A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2018559040A JPWO2018123628A1 (en) 2016-12-28 2017-12-14 Cell pharmaceutical composition, disease treatment kit and cell suspension solution
JP2022165115A JP2022179749A (en) 2016-12-28 2022-10-14 Cell pharmaceutical composition, disease treatment kit, and cell suspension solution

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2016-256633 2016-12-28
JP2016256633 2016-12-28

Publications (1)

Publication Number Publication Date
WO2018123628A1 true WO2018123628A1 (en) 2018-07-05

Family

ID=62707371

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2017/044993 WO2018123628A1 (en) 2016-12-28 2017-12-14 Cell pharmaceutical composition, disease treatment kit, and cell suspension solution

Country Status (2)

Country Link
JP (2) JPWO2018123628A1 (en)
WO (1) WO2018123628A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009057537A1 (en) * 2007-11-02 2009-05-07 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition containing human mesenchymal stem cell
JP2009242265A (en) * 2008-03-28 2009-10-22 Jcr Pharmaceuticals Co Ltd Therapeutic agent for atopic dermatitis
JP2014501778A (en) * 2011-01-10 2014-01-23 ステムニオン,インコーポレイテッド Methods for promoting healing of connective tissue damage and disorders
WO2016204230A1 (en) * 2015-06-17 2016-12-22 仁幸 小林 Stem cell administration method, method for improving symptoms of race horses or show horses, injection container and stem cell injection set

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009213448A (en) * 2008-03-12 2009-09-24 Institute Of Physical & Chemical Research Method of making mesenchymal cell line and use thereof
JP5993113B2 (en) * 2011-01-31 2016-09-14 株式会社 バイオミメティクスシンパシーズ Human adipose tissue-derived mesenchymal stem cells for the treatment of Alzheimer's disease
JP2013252126A (en) * 2012-05-08 2013-12-19 Otsuka Pharmaceut Factory Inc Dextran-containing mammalian cell suspension for prevention of pulmonary embolism formation
JP5432322B2 (en) * 2012-05-08 2014-03-05 株式会社大塚製薬工場 Mammalian cell suspension for prevention of pulmonary embolism containing trehalose
EP3015545B1 (en) * 2013-06-28 2018-12-26 Otsuka Pharmaceutical Factory, Inc. Trehalose and dextran-containing solution for transplanting mammalian cells
JP6191694B2 (en) * 2013-08-01 2017-09-06 株式会社ツーセル Cartilage injury therapeutic agent and method for producing the same

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009057537A1 (en) * 2007-11-02 2009-05-07 Jcr Pharmaceuticals Co., Ltd. Pharmaceutical composition containing human mesenchymal stem cell
JP2009242265A (en) * 2008-03-28 2009-10-22 Jcr Pharmaceuticals Co Ltd Therapeutic agent for atopic dermatitis
JP2014501778A (en) * 2011-01-10 2014-01-23 ステムニオン,インコーポレイテッド Methods for promoting healing of connective tissue damage and disorders
WO2016204230A1 (en) * 2015-06-17 2016-12-22 仁幸 小林 Stem cell administration method, method for improving symptoms of race horses or show horses, injection container and stem cell injection set

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Ringer's Solution Otsuka- Ringer's Solution Fuso", PHARMACEUTICAL PACKAGE INSERT. JAPANSES PHAMACOPOEIA, 2009 *

Also Published As

Publication number Publication date
JPWO2018123628A1 (en) 2019-10-31
JP2022179749A (en) 2022-12-02

Similar Documents

Publication Publication Date Title
WO2018159432A1 (en) Mesenchymal stem cells and pharmaceutical composition
PT2120977E (en) Treatment of inflammatory diseases using placental stem cells
JP7082372B2 (en) Pulmonary fibrosis treatment agent, PTPRR expression promoter and pulmonary fibrosis treatment kit
KR20130092394A (en) Treatment of sarcoidosis using placental stem cells
JP6960120B2 (en) Liver disease therapeutic agents and methods for treating liver disease
JP2024023760A (en) Mesenchymal stem cell and therapeutic agent for neuropathy
JP2019156739A (en) Mesenchymal stem cell, disease therapeutic agent, and microparticle
WO2018116732A1 (en) Therapeutic agent for non-alcoholic steatohepatitis, and kit for treatment of non-alcoholic steatohepatitis
JP7108541B2 (en) Kit for preparing therapeutic agent for disease, therapeutic agent for disease, and method for preparing therapeutic agent for disease
EP3381458B1 (en) Therapeutic agent for liver disease containing stromal cells derived from adipose tissue and method for producing the same
JP2023103416A (en) Therapeutic agent for dilated cardiomyopathy
JP7136700B2 (en) Cell pharmaceutical composition, disease treatment kit and cell suspension solution
JP2023024665A (en) Mesenchymal stem cells and therapeutic agent for liver disease
WO2018123628A1 (en) Cell pharmaceutical composition, disease treatment kit, and cell suspension solution
JP7189659B2 (en) Cell pharmaceutical composition for treating disease, kit for treating disease, method for preparing cell pharmaceutical composition
WO2017141831A1 (en) Therapeutic agent containing fat-tissue derived stromal cells for ischemic disease and manufacturing method therefor

Legal Events

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

Ref document number: 17886473

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018559040

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17886473

Country of ref document: EP

Kind code of ref document: A1