WO2024177018A1 - Method for producing induced pluripotent stem cells - Google Patents
Method for producing induced pluripotent stem cells Download PDFInfo
- Publication number
- WO2024177018A1 WO2024177018A1 PCT/JP2024/005834 JP2024005834W WO2024177018A1 WO 2024177018 A1 WO2024177018 A1 WO 2024177018A1 JP 2024005834 W JP2024005834 W JP 2024005834W WO 2024177018 A1 WO2024177018 A1 WO 2024177018A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- container
- sealed container
- cells
- sealed
- containers
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12M—APPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
- C12M3/00—Tissue, human, animal or plant cell, or virus culture apparatus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
Definitions
- the present invention relates to a method for producing induced pluripotent stem cells and a method for producing differentiated cells.
- the present invention also relates to a cell production device for carrying out the production method, and a cell production method using the cell production device.
- iPS cells differentiated cells derived from induced pluripotent stem cells
- a treatment in which iPS cells are established from a patient's somatic cells (e.g., peripheral blood mononuclear cells) and various differentiated cells or organoids induced to differentiate from the iPS cells are then transplanted (autotransplant) into the patient has attracted attention as a treatment that can reduce the risk of rejection (Patent Document 1, Non-Patent Documents 1 and 2).
- obtaining iPS cells from cells (somatic cells) obtained from a living organism requires a long-term, multi-step process, such as contacting the somatic cells with reprogramming factors in a liquid medium, reducing the concentration of the reprogramming factors in the liquid medium, and then culturing the cells to establish iPS cells.
- obtaining various differentiated cells from the established iPS cells also requires a long-term, multi-step process, such as inducing differentiation into the desired cells and testing the quality of the cells.
- a common feature of such cell manufacturing devices is that, as shown in FIG. 42(a), materials required for the processes are supplied in sequence from a number of material supply bags X20 into a single culture vessel X10, and all processes are performed in sequence within the single culture vessel X10.
- a soft tube (hereinafter also referred to as "tube”) X30 extends from each of the many material supply bags X20, and each tube X30 passes through a pinch valve X40 that can be opened and closed, and then repeatedly merges to become one tube, which passes through a peristaltic pump X50 and is connected to the culture vessel X10.
- the pinch valve X40 is an electromagnetic valve that operates according to commands from a control unit (not shown). When the pinch valve presses the tube from the outside so as to crush it, the flow path in the tube is closed.
- the pinch valve and the peristaltic pump are controlled by a computer program, and the materials required for each process to be performed are sequentially sent into the culture vessel X10, and each process is performed sequentially.
- the flow path for the waste liquid is also configured so that the waste liquid (such as old culture liquid) that flows through the peristaltic pump passes through the tube and the pinch valve, and then is discharged into a waste liquid bag.
- the waste liquid may be disposed of as a liquid, but by using a hygroscopic waste liquid bag in which a hygroscopic material (e.g., water-absorbing resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbing pad, water-absorbing sheet, etc.) is enclosed in the waste liquid bag, the liquid components can be incinerated without separation, making disposal easier. With these configurations, all processes are performed in the single culture vessel X10.
- a hygroscopic material e.g., water-absorbing resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.
- an absorbent article containing the material e
- FIG. 42(b) the manner in which multiple tubes merge into one tube and are connected to a single sealed container X10 is as shown diagrammatically in FIG. 42(b).
- FIG. 42(b) the closer the flow path is to the sealed container X10, the more the flow path is shared by multiple material supply bags.
- pinch valves, peristaltic pumps, and waste liquid flow paths are omitted from the illustration.
- the conventional cell manufacturing devices described above are very expensive because of the complex valves and control circuits for switching the flow paths. Furthermore, the inventors' detailed studies have revealed that the conventional cell manufacturing devices described above have the following problems. Conventional cell manufacturing devices require multiple junction tubes, and the task of setting each section of the junction tubes in a pinch valve is complicated and time-consuming.
- the length of the flow path from the supply bag to the culture vessel X10 is necessarily long because it is necessary to secure the tube X30 section, making it difficult to transport small volumes of liquid, such as 100 ml or less. Because all steps are carried out within a single sealed container, production of other cells cannot begin in the cell production device until the last step is completed.
- the object of the present invention is to provide a new manufacturing method (a method for manufacturing iPS cells and a method for manufacturing differentiated cells) that can suppress or eliminate the above problems with conventional cell manufacturing devices, as well as a new cell manufacturing device and a cell manufacturing method using the same.
- a method for producing induced pluripotent stem cells comprising: sequentially moving cells in one direction from a first sealed container (A1) to an nth sealed container (An) by a feed mechanism among n (n ⁇ 3) sealed containers connected in series via a connecting pipeline; and sequentially carrying out a production process of induced pluripotent stem cells in each sealed container;
- Each of the sealed containers (A1 to An) has one or more openable/closable inlet/outlet ports,
- the connecting pipe is configured to be switchable between a communicating state and a non-communicating state, the feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to a communicating state,
- the process for producing the induced pluripotent stem cells comprises: A step (s1) of contacting a reprogramming factor with a somatic cell in a liquid medium in a first sealed container (A1); A step (s2) of reducing the concentration of the induced pluripotent stem cells
- the method further comprises a step (s4) of expanding the induced pluripotent stem cells p times (p ⁇ 1); p sealed containers (B1 to Bp) corresponding to the number of times of expansion culture are connected in series to the n-th sealed container (An) via a connecting pipeline;
- the artificial pluripotent stem cells are sequentially moved in one direction from the sealed container (An) to the sealed container (Bp) by the respective feeding mechanisms, and one expansion culture is carried out in each of the sealed containers (B1 to Bp), thereby carrying out a total of p expansion cultures;
- Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports,
- the connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
- the feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to the communicating state.
- the sealed container is A sealed container having a container body made of a flexible material; A sealed container having a container body made of a hard material; A sealed container having a container body made of a composite material of a flexible material and a hard material.
- the feed mechanism is A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container; A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container; a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline; a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container; A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
- a method for producing induced pluripotent stem cells comprising the steps of: A step of producing induced pluripotent stem cells by the production method according to any one of [1] to [4]; and a step (s5) of inducing differentiation of the produced induced pluripotent stem cells, a sealed container (C1) for carrying out the step (s5) is further connected to the rear end of the sealed container (X1) among the sealed containers used in the method for producing induced pluripotent stem cells via a connecting pipeline;
- the sealed container (C1) has one or more openable/closable inlet/outlet ports, and a material necessary for differentiation induction in step (s5) is supplied to the inside of the sealed container (C1) through the inlet/outlet ports;
- the artificial pluripotent stem cells are transferred from the sealed container (X1) to the sealed container (C1) by a transfer mechanism, and the step (s5) is carried out in the sealed container (C1);
- the method for producing the differentiated cells further comprises a step (s6) of removing undifferentiated cells after the differentiation induction step; a sealed container (D1) for carrying out the step (s6) is further connected to the rear end of the sealed container (X2) among the sealed containers used in the differentiation induction step via a connecting pipeline;
- the sealed container (D1) has one or more openable/closable inlet/outlet ports,
- the differentiated cells are transferred from the sealed container (X2) to the sealed container (D1) by a transfer mechanism, and the step (s6) is carried out in the sealed container (D1);
- the connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
- the feed mechanism is a mechanism for moving the contents from the sealed container (X2) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
- a cell manufacturing device comprising: A sealed container (A1) for carrying out a step (s1) of contacting a somatic cell with a reprogramming factor in a liquid medium; A sealed container (A2) for carrying out a step (s2) of reducing the concentration of the reprogramming factor in the liquid medium; and a sealed container (A3) for carrying out a step (s3) of culturing the somatic cells in the liquid medium to establish induced pluripotent stem cells;
- Each of the sealed containers (A1) to (A3) has one or more openable/closable inlet/outlet ports,
- the sealed containers (A1) to (A3) are connected in series in the order of the steps via a connecting pipe line that can be switched between a communicating state and a non-communicating state, or are capable of being connected in series in the order of the steps, and
- the cell manufacturing device comprises: a feed mechanism for moving the content of the sealed container (A1) to the sealed container (A2) through the connecting pipe
- the method further comprises: a step (s4) of expanding the induced pluripotent stem cells in a liquid medium p times (p ⁇ 1) in a number of sealed containers (B1 to Bp);
- Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports, (i) When the number of times of expansion culture, p, is 1, The p number of sealed containers is one sealed container (B1), the sealed container (B1) is connected to or can be connected to the sealed container (A3) via a connecting pipe that can be switched between a communicating state and a non-communicating state; the cell manufacturing apparatus has a transfer mechanism that transfers the content of the sealed container (A3) to the sealed container (B1) through the connecting pipeline that has been switched to a communicating state; (ii) When the number of times of the expansion culture, p, is 2 or more, The p number of sealed containers is two or more sealed containers (B1 to Bp), the sealed container (B1) is connected to or can be connected to the sealed
- each of the sealed containers (C1) to (Cq) has one or more openable/closable inlet/outlet ports, (i) When the q number of sealed containers is one sealed container (C1), the sealed container (C1) is connected or connectable to the sealed container (A3) or to the last sealed container (Bp) among the sealed containers (B1) to (Bp) via a connecting pipe that can be switched between a communicating state and a non-communicating state; the cell manufacturing apparatus has a transfer mechanism that transfers the contents of the sealed container (A3) or the sealed container (Bp) to the sealed container (C1) through the connecting pipeline that has been switched to a communicating state; (ii) When the q number of sealed containers is two or more sealed containers (C1) to (Cq), the sealed containers (C1) to (Cq) are
- the cell manufacturing apparatus according to [7] or [8]. [10] Further comprising a sealed container (D1) for carrying out a step (s6) of removing undifferentiated cells from the content of the last sealed container (Cq) among the sealed containers (C1) to (Cq),
- the sealed container (D1) has one or more openable/closable inlet/outlet ports, the sealed container (D1) is connected to or can be connected to the rearmost sealed container (Cq) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
- the cell manufacturing device comprises: A feed mechanism is provided for moving the contents of the rearmost sealed container (Cq) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
- the sealed container is A sealed container having a container body made of a flexible material; A sealed container having a container body made of a hard material; A sealed container having a container body made of a composite material of a flexible material and a hard material.
- the cell manufacturing apparatus according to any one of [7] to [10].
- the feed mechanism includes: A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container; A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container; a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline; a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container; A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
- the cell manufacturing apparatus according to any one of [7] to [11]. [13] Further comprising a substrate for arranging all of the sealed containers; The sealed containers are disposed on the substrate, and each sealed container is fixed to the substrate; The sealed containers are connected or connectable in the order of the steps via the connecting pipes that can be switched between a communicating state and a non-communicating state.
- a cell manufacturing apparatus according to any one of [7] to [12].
- the substrate can be folded in two around a folding center line, (i) in one region (e1) of two regions (e1) and (e2) on the substrate surface separated by the folding center line, a predetermined number of the sealed containers are arranged in order in one direction (d1) along the folding center line; (ii) in the other region (e2) of the two regions (e1) and (e2) of the substrate surface separated by the folding center line, the remaining sealed containers among the sealed containers are arranged in order along the folding center line in a direction (d2) opposite to the direction (d1); (iii) the rearmost sealed container among the sealed containers in the one region (e1) and the frontmost sealed container among the sealed containers in the other region (e2) are connected or in a connectable state by the connecting pipe line that can be switched between a communicating state and a non-communicating state;
- the cell manufacturing apparatus described in [13].
- a method for manufacturing a flexible sheet-type display device comprising: the two flexible sheets are bonded to each other while leaving the regions that become all of the above-mentioned sealed containers, the region that becomes one or more inlet/outlet ports, and the region that becomes the connecting pipeline at predetermined positions between the two flexible sheets as non-bonded regions; a pressing actuator for opening and closing the connection pipe line is provided on the outer surfaces of the two flexible sheets; the pressing actuator operates to take a pressing position in which the region that will become the connecting pipeline is pressed from the outside of the flexible sheet to bring it into a non-communicating state, and a non-pressing position in which the region that will become the connecting pipeline is not pressed to bring it into a communicating state, and by operation of the pressing actuator, the region that will become the connecting pipeline functions as a connecting pipeline that can be switched between a communicating state and a non-communicating state; The region that will become one or more inlet/outlet ports extends from the region that will become each sealed container to the outer periphery
- the outer peripheral shape of the two flexible sheets that are overlapped and joined to each other is a shape that can be folded in two around a folding center line, (i) In one of the two regions (e3) and (e4) separated by the folding center line, A predetermined number of regions that become sealed containers among the sealed containers are formed so as to be arranged in sequence in one direction (d3) along the folding center line, from an outer periphery of each of the regions that will become the predetermined number of sealed containers that is located farther from the folding center line, a region that will become the one or more inlet/outlet ports extends in a direction away from the folding center line and extends to an outer periphery of the two flexible sheets to form an open end, Between the regions that will become the predetermined number of sealed containers, a region that will become the connecting pipeline that connects the regions that will become the sealed containers is formed, (ii) In the other region (e4) of the two regions (e3) and (e
- the cell production method comprises at least a step (s3) of transferring the contents of the sealed container (A2) into the sealed container (A3) through the connecting pipeline that has been switched to a communicating state after completion of the step (s2), and establishing artificial pluripotent stem cells in the liquid medium within the sealed container (A3).
- the differentiation-inducing step (s5) is a step (s5a) of inducing differentiation of induced pluripotent stem cells into ectodermal cells, mesodermal cells, or endodermal cells. The method for producing differentiated cells described in [6].
- the method further comprises, after the step (s5a), a step (s5b) of inducing differentiation of the ectodermal cell, mesodermal cell, or endodermal cell,
- a sealed container (C2) for carrying out the step (s5b) is further connected to the rear of the sealed container (C1) via a connecting pipeline,
- the sealed container (C2) has one or more openable/closable inlet/outlet ports, and a material necessary for differentiation induction in step (s5b) is supplied to the inside of the sealed container (C2) through the inlet/outlet ports; the ectodermal cells, mesodermal cells, or endodermal cells are transferred from the sealed container (C1) to the sealed container (C2) by a transfer mechanism, and the step (s5b) is carried out in the sealed container (C2);
- the connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
- the feed mechanism is a mechanism for moving the contents from the sealed container (C1)
- the method for producing differentiated cells further comprises, after the step (s6) of removing undifferentiated cells, a step (s7) of removing a sample for testing differentiated cells;
- a sealed container (E1) for carrying out the step (s7) is further connected to the sealed container (D1) via a connecting pipeline,
- the sealed container (E1) has one or more openable/closable inlet/outlet ports, and the sample is taken out through the one or more inlet/outlet ports;
- the contents are transferred from the sealed container (D1) to the sealed container (E1) by the feeding mechanism,
- the connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
- the feed mechanism is a mechanism for moving the contents from the sealed container (D1) to the sealed container (E1) through the connecting pipe line switched to a communicating state.
- the sealed container will be simply referred to as a "container.”
- multiple containers are used to process cells, rather than a single container.
- Each of the multiple processes required to produce iPS cells or differentiated cells is associated with one of the multiple containers, and each process is carried out in the corresponding container.
- the process in each container is completed, the cells processed there are aseptically sent by a sending mechanism through a connecting pipeline to the next container, where the cells are processed in the next process.
- step s1 is carried out in the first container A1
- step s2 is carried out in the second container A2
- step s3 is carried out in the third container A3, and the target cells are obtained there.
- the above-described configuration provides the following effects.
- the complicated piping and control mechanism in the above-mentioned conventional manufacturing apparatus can be simplified, and the manufacturing apparatus can be constructed more inexpensively. Also, since each manufacturing process is completed by each corresponding container, and the order of connecting the containers determines the order of the manufacturing processes, it is possible to freely configure a series of manufacturing processes by changing the order of connecting the containers.
- the first step is completed in a first container, and the cells are sent to a second container for the second step, so that the management division for performing in-process tests such as sterility tests is clear, and it is possible to start a new first step in the first container. Therefore, the parallelism of the manufacturing process is increased, and the throughput per manufacturing device is improved.
- vessels can be easily added to add processes or removed to omit processes according to differences in the cells to be produced (for example, differences in differentiated cells), and each process can be increased or decreased like a block. Therefore, at the planning stage of the production process, it is possible to assemble the vessel connections according to the contents of the production plan.
- the cells since the cells are moved from container to container for each process, the cells can be moved together with the container separately from the device at each process, which provides the effect that the movable containers containing the cells have the mobility suitable for assembly line work on a manufacturing line.
- FIG. 1 is a block diagram for explaining the method for producing iPS cells according to the present invention.
- each container has a container body and a plug, and the container is shown in a cross-sectional view.
- Fig. 1 is also a block diagram showing an example of the configuration of a cell production device according to the present invention.
- a gap is drawn between the opening of the container body and the plug, but the plug seals the opening of the container body (the same applies to other figures).
- FIG. 2 is a diagram showing an example of the configuration of an input/output port in the present invention.
- FIG. 3 is a block diagram showing an example of the configuration of an inlet/outlet port in the present invention, in which the container is shown in cross section (the same is true for the other block diagrams).
- FIG. 4 is a block diagram showing an example of the configuration of a connecting pipeline according to the present invention.
- FIG. 5 is a block diagram showing an example of the configuration of a feed mechanism according to the present invention.
- FIG. 6 is a block diagram showing an example of the configuration of a feed mechanism according to the present invention, which is a diagram showing an example of the configuration that utilizes gravity.
- FIG. 7 is a block diagram showing another example of the configuration of the feed mechanism according to the present invention, which is a diagram showing an example of the configuration that utilizes magnetic force.
- FIG. 8 is a block diagram for explaining step s4 of expanding iPS cells in the method for producing iPS cells according to the present invention.
- each container has a container body and a stopper, and the container is shown in a cross-sectional view.
- Fig. 8 is also a block diagram showing an example of the configuration of the part for expanding iPS cells in the cell production device according to the present invention. In order to easily show the main parts, the assignment of symbols to input/output ports, etc. is omitted.
- Fig. 9 is a block diagram for explaining the method for producing differentiated cells according to the present invention, and is also a block diagram showing an example of the configuration of a cell production apparatus according to the present invention.
- FIG. 10 is a block diagram for explaining a preferred embodiment of the method for producing differentiated cells according to the present invention, and is a diagram for explaining step s6 of removing undifferentiated cells and step s7 of inspecting differentiated cells.
- Fig. 10 is also a block diagram showing an example of the configuration of a cell production apparatus according to the present invention.
- FIG. 11 is a block diagram for explaining the part of the cell manufacturing apparatus of the present invention that manufactures iPS cells.
- 12 is a diagram showing a part of the cell manufacturing device of the present invention where differentiation of iPS cells is induced. In order to clearly show the main parts, reference numerals for input/output ports and the like have been omitted.
- FIG. 10 is a block diagram for explaining a preferred embodiment of the method for producing differentiated cells according to the present invention, and is a diagram for explaining step s6 of removing undifferentiated cells and step s7 of inspecting differentiated cells.
- Fig. 10 is also a block diagram showing an
- FIG. 13 is a diagram showing an example of a preferred embodiment of the cell manufacturing apparatus according to the present invention.
- FIG. 14 is a diagram illustrating a state in which the substrate in the cell manufacturing apparatus shown in FIG. 13 is folded.
- FIG. 15 is a diagram showing another preferred embodiment of the cell manufacturing apparatus according to the present invention.
- FIG. 16 is a diagram showing an example of a preferred embodiment for arranging and fixing a plurality of containers on a substrate.
- FIG. 17 shows an embodiment of the substrate shown in FIG. 16 and its usage state.
- FIG. 18 is a diagram showing another preferred embodiment for arranging and fixing a plurality of containers on a substrate.
- FIG. 19 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 20 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 21 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 22 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 23 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 24 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 25 is a diagram showing the cell manufacturing device of the present invention produced in Example 1 and its usage state.
- FIG. 26 is a diagram showing the evaluation of iPS cells after expansion culture in Example 1 of the present invention.
- FIG. 27 is a diagram relating to the evaluation of iPS cells after establishment in Example 2 of the present invention.
- FIG. 28 is a diagram relating to the evaluation of iPS cells after expansion culture in Example 2 of the present invention.
- FIG. 29 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 30 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 31 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 32 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 33 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 34 is a diagram showing the cell manufacturing device of the present invention produced in Example 3 and its usage state.
- FIG. 35 is a diagram relating to the evaluation of iPS cells after expansion culture in Example 3 of the present invention.
- FIG. 36 is a diagram relating to the evaluation of iPS cells after expansion culture in Example 4 of the present invention.
- FIG. 37 is a diagram showing confirmation of expression of a marker protein (troponin T (TNNT2)) of cardiomyocytes in Example 5 of the present invention.
- FIG. 38 is a diagram showing induction of expression of marker proteins of cardiomyocytes/cardiac progenitor cells in Example 5 of the present invention.
- FIG. 39 is a diagram showing confirmation of expression of a marker protein (PDX-1) of pancreatic progenitor cells in Example 6 of the present invention.
- FIG. 40 is a diagram showing confirmation of expression of a marker protein (NKX6.1) of pancreatic progenitor cells in Example 6 of the present invention.
- FIG. 41 is a diagram showing induction of expression of marker proteins for pancreatic progenitor cells in Example 6 of the present invention.
- Fig. 42 is a diagram for explaining the configuration of a conventional cell manufacturing device
- Fig. 42(a) is a diagram showing an example of a commercially available cell manufacturing device
- Fig. 42(b) is a block diagram showing the features of the device in Fig. 42(a). In Fig. 42(b), pinch valves and peristaltic pumps are omitted.
- FIG. 1 is a block diagram for explaining the method for producing iPS cells according to the present invention, and is also a block diagram showing a schematic example of the configuration of a cell production device according to the present invention.
- the inlet/outlet ports of each sealed container and the connecting pipes between the sealed containers are configured to be openable and closable, so that the inside of each sealed container can be sealed. Therefore, the production method and production device of the present invention enable cell processing and cell culture in a closed system.
- the sealed container used in the manufacturing method and manufacturing apparatus of the present invention may be referred to simply as a "container" or, if necessary, as a "sealed container.”
- FIG. 1 indicate piping lines such as soft tubes.
- Fig. 1 in order to simply show the main parts of the configuration of the invention, connectors, on-off valves, etc. provided on the piping lines are omitted. The same is true for the examples in the other figures.
- the number of material supply sources (reference numerals G1 to Gn in FIG. 1) connected to each container is shown as only one, but the number is not limited.
- the mechanism for sending the material from the material supply source to the input/output port is not limited and may be one according to the material to be supplied.
- a feeding mechanism is provided for moving cells from a container to a container at the next stage through a connecting pipeline.
- Fig. 1 shows a feeding mechanism (indicated by symbols F1 to Fn in Fig. 1) provided as a pump on the connecting pipeline, but as described below, there are various modes of the feeding mechanism.
- the examples in the other figures are similar.
- production method (I) Method for producing induced pluripotent stem cells
- the manufacturing method (I) uses n containers (A1 to An) connected in series via connecting pipelines J1 to J(n-1).
- n is an integer equal to or greater than 3 (i.e., n ⁇ 3).
- Each container is provided with an inlet/outlet port (e.g., reference symbol h1 for container A1) that can be opened and closed, and each inlet/outlet port is connected to a necessary material supply source (G1 to Gn) so that a process associated with each container is performed.
- the content K1 contained in the container is a cell suspension, and the cells change or the composition of the cell suspension changes each time the content moves to the next container.
- the connecting pipeline (more detailed configuration will be described later) can be switched between a communicating state and a non-communicating state.
- the important point of this manufacturing method (I) is that the cells are sequentially moved in one direction from the first container A1 to the nth container An through the connecting pipelines J1 to J(n-1) by the feed mechanisms F1 to F(n-1), and the following steps s1 to s3 are sequentially carried out in each container.
- Step s1 A step of contacting somatic cells with reprogramming factors in a liquid medium in a first container A1.
- Step s2 A step of reducing the concentration of the reprogramming factor in the liquid medium in the second container A2 to the (n-1)th container A(n-1).
- Step s3 A step of culturing the somatic cells in a liquid medium in the n-th container An to establish iPS cells.
- step s2 is divided into multiple processing stages (multiple steps), and the multiple steps are carried out sequentially in containers A2 to A(n-1) to complete step s2.
- step s2 is completed, the cells are sent to container An, where step s3 is carried out.
- step s3 is carried out.
- the steps associated with each container are carried out sequentially in each container, and the final step s3 is completed in the last container An, and iPS cells are established in that container An.
- n containers (A1 to An) There is no particular limitation on the number of stages into which step s2 is divided, but in a normal processing operation, about 1 to 3 stages are preferable, and even a one-stage processing operation can preferably reduce the concentration of the reprogramming factor in the liquid medium. Therefore, the number n of containers in the production method (I) is not particularly limited, but is preferably about 3 to 5.
- step s2 is a one-stage processing operation, only container A2 is used in step s2, and the above n is 3, and in the production method (I), three containers (A1 to A3) connected in series via connecting pipes J1 and J2 are used.
- a connection that initially includes a parallel connection such as the merging of multiple vessels (A'11, A'12, A'13, . . . ) into one vessel (A2).
- a connection that includes a parallel connection along the way such as branching in parallel from one container A1 to multiple containers (A2a, A2b, A2c, ...) and then merging from those multiple containers into one container A3.
- a connection that includes a parallel connection at the end such as branching from one container A(n-1) to multiple containers (A'n1, A'n2, A'n3, ...) in parallel.
- each step is divided into two containers and carried out.
- the number of steps is three or more, the number of containers used does not necessarily have to be the same as the number of steps (i.e., one step does not have to correspond to one container), and the number of containers may be less than the number of steps.
- two containers may be used, and steps s1 and s2 may be carried out sequentially in container A1 and step s3 may be carried out in container A2, or step s1 may be carried out in container A1 and steps s2 and s3 may be carried out in container A2.
- the number of containers into which three or more steps are divided can be appropriately selected.
- one step s2 is further divided into multiple steps, and one container is associated with each divided step.
- the container can be one that can accommodate cells together with a liquid medium and process and culture the cells in a sterile environment.
- the container can be any of various sealed containers for culture used in conventional cell culture and cell production.
- the containers used in the present invention may be different from each other in terms of reducing costs, modularization, use of standardized and unified containers for tissue culture engineering evaluation, temperature control stability of the culture container, regulation of the amount of medium liquid supplied, management of the number of cultured cells, evaluation of medium components, correction of lot-to-lot differences, setting of oxygen and carbon dioxide supply values for the medium, speeding up the acceptance test of the container, unification of gas sterilization, gamma ray sterilization, ultraviolet sterilization, and packaging form of connected containers, and unification of packaging, transportation, and storage methods of the containers. It is preferable that all the containers have the same shape and specifications. If a sufficient number of inlet and outlet ports are provided for each container and unnecessary inlet and outlet ports are closed, the containers can be easily unified.
- a preferred embodiment of the container is as follows: (i) a sealed container having a container body made of a flexible material; (ii) A sealed container having a container body made of a hard material; (iii) A sealed container having a container body made of a composite material of a flexible material and a hard material.
- the container body has an opening (mouth), and the opening is closed with a lid, a stopper, or the like to form a sealed container.
- a cylindrical connector or the like may be attached to the opening, and the conduit of the connector may be closed with a stopcock or the like to seal the container body.
- the materials of the lid, stopper, and inlet/outlet port that close the opening of the container body may be appropriately selected with reference to conventionally known sealed containers for cell culture.
- the structure of the sealed container having a container body made of the flexible material (i) above is not particularly limited, but typical examples include flexible bags made of flexible films, such as conventionally known cell culture bags and infusion bags (MACS (registered trademark) GMP Cell Culture Bags (Miltenyi Biotec), Flexboy (registered trademark) bags (SARTORIUS)).
- the flexible film is a film that has flexibility to the extent that it can be deformed according to the amount of contents contained in the bag.
- the structure of the bag is not limited, but examples include a bag structure in which two rectangular or square flexible films are overlapped and the outer periphery parts other than the opening and the inlet/outlet port are fused (heat fusion, high-frequency fusion, etc.) or bonded to each other.
- the flexible film constituting the bag is preferably gas permeable, allowing O 2 and CO 2 necessary for cell culture to pass through.
- O 2 and CO 2 necessary for cell culture may be appropriately supplied through the inlet/outlet port described below.
- the flexible film is preferably made of a material that is industrially excellent in moldability, can withstand gamma ray sterilization, and is transparent so that the state of the culture medium inside can be observed. Additional structures such as holes for suspending the bag can be provided as appropriate with reference to conventionally known cell culture bags, etc.
- the above-mentioned sealed container used in the production method of the present invention is preferably non-cell-adhesive in order to enable suspension culture in the production method of the present invention described below.
- the sealed container may be one that has not been artificially treated (e.g., coated with an extracellular matrix, etc.) for the purpose of improving adhesion to cells.
- the flexible film that constitutes the bag can be made of known materials used for cell culture bags, such as polyethylene, polyethylene terephthalate, polypropylene, ethylene vinyl acetate copolymer (EVA), ultra-low density polyethylene (ULDPE)/ethyl vinyl alcohol (EVOH), ultra-low density polyethylene (ULDPE), polyolefin (PO), tank liner, polyvinylidene fluoride, polyethersulfone, etc.
- EVA ethylene vinyl acetate copolymer
- ULDPE ultra-low density polyethylene
- EVOH ultra-low density polyethylene
- PO polyolefin
- PO polyolefin
- PO polyolefin
- the flexible film can be a single layer film or a multilayer film made of these materials.
- the sealed container having a container body made of the above-mentioned (ii) hard material is not particularly limited, but examples thereof include flasks, flasks for tissue culture, vials, tubes (test tubes), bioreactors, jar fermenters, hollow fiber membrane bioreactors, and the like, which are difficult to deform due to the weight of the cell suspension even when the cell suspension is contained therein.
- the hard material known materials that have been used in cell culture containers in the past, such as glass and plastic materials (e.g., polycarbonate, polyester, polyamide, polystyrene, acrylonitrile-butadiene-styrene copolymer (ABS resin), polyethylene, polypropylene, polyvinyl chloride, biodegradable resins), can be used.
- the opening of the container body is sealed with a lid or a stopper, thereby forming a sealed container.
- a required number of tubes are preferably inserted through the lid or the stopper from the outside to the inside.
- the sealed container having a container body made of a hard material may be one designed and manufactured for the present invention, but a commercially available cell culture container equipped with an inlet/outlet port may also be used.
- G-Rex (registered trademark) 10N-CS and G-Rex 100N-CS manufactured by Wilson Wolf are cell culture devices that have a sufficient number of inlet/outlet ports on the lid and a gas-permeable membrane on the bottom of the container body that allows the permeation of O2 and CO2 necessary for cell culture, and can be preferably used as the sealed container in the present invention.
- An example of an airtight container having a container body made of a composite material of flexible and hard materials as described above in (iii) is one in which the framework is made of a hard material and the walls are made of a flexible film.
- Such containers have the three-dimensional characteristics of hard containers, but are also inexpensive and disposable.
- the sealed container that can be used in the present invention is a container that, when the input/output port including the opening for the connecting pipeline is closed, provides a sealed internal space that allows cell culture in a closed system.
- the above-mentioned "sealed internal space that allows for culture in a closed system” is a space whose walls, openings, and inlet/outlet ports are closed to the extent that microorganisms or viruses do not enter from the outside, that is, to the extent that sterility within the container is maintained.
- the space is airtightly sealed so that gas or contents do not leak from other unintended locations when, for example, gas is fed into the container and the contents are pushed out through a specified inlet/outlet port.
- a container having an inlet/outlet port provided with a porous filter that cannot pass bacteria or viruses but can pass fluids (especially gases) can allow outside air to pass through the porous filter and flow into the container, but the sterility of the container is maintained. Therefore, the inside of the container is the above-mentioned "sealed internal space that allows culture in a closed system.”
- the inlet/outlet port equipped with such a porous filter may be configured to be airtightly closed by a valve, stopcock, etc.
- the containers such as the above-mentioned G-Rex (registered trademark) 10N-CS and G-Rex 100N-CS manufactured by Wilson Wolf have a gas-permeable membrane at the bottom wall of the container body, which cannot be penetrated by bacteria or viruses, but is permeable by O2 gas molecules and CO2 gas molecules necessary for cell culture, and the inside of the container can be said to be an airtight sealed space. Therefore, it is possible to maintain the sterility of the inside of the container, and it is also possible to send gas into the container through the inlet/outlet port and push the contents out through a specified inlet/outlet port. Therefore, the inside of the container is the above-mentioned "sealed internal space that allows culture in a closed system.”
- the volume of the container is not particularly limited, and may be a large volume that allows industrially large amounts of iPS cells to be produced at once; however, for applications such as transplantation therapy using differentiated cells derived from autologous iPS cells, the volume is preferably about 1 to 1,000 ml, and more preferably about 10 to 100 ml.
- Input and output ports are provided for introducing materials into and removing materials from the container. As many input and output ports as necessary may be provided on each container for such purposes as: (i) Use as a supply port for delivering materials (such as somatic cells, liquid culture medium, and various reagents) required for the process carried out in each vessel from the outside into the vessel. (ii) Use as a pressure adjustment hole (gas vent hole, air supply hole, liquid outflow hole) to eliminate pressure changes when materials are sent into each container from the outside or when materials are removed from the container.
- materials such as somatic cells, liquid culture medium, and various reagents
- the number of inlet/outlet ports provided in one container does not necessarily have to be the number of the above applications.
- One inlet/outlet port may be used for multiple applications by using a valve or branch pipe for switching the flow path, or by attaching and detaching an external pipeline.
- the inlet/outlet port for supplying gas necessary for cell culture in (i) above can also be used as an inlet/outlet port for supplying gas when pushing out the contents from the container in (vi) above.
- the inlet/outlet port as a pressure adjustment hole in (ii) above can also be used as an inlet/outlet port for discharge in (iii) above.
- inlet/outlet ports In the case of replacing a liquid culture medium, when a new liquid culture medium is supplied into a container and an old liquid culture medium is pushed out from the container, two inlet/outlet ports are used. If one inlet/outlet port is used for multiple applications, it may be troublesome to attach and detach or switch multiple external pipelines to one inlet/outlet port, or the piping may become complicated. Therefore, the number of inlet/outlet ports may be provided for the number of applications, or one inlet/outlet port may be used for an appropriate number of applications. For these reasons, the number of inlet/outlet ports provided in one container is generally about 3 to 10.
- the number of inlet/outlet ports may vary from container to container, but in order to standardize container specifications to ensure compatibility and reduce costs, it is possible to provide the same number of inlet/outlet ports on all sealed containers and close unused inlet/outlet ports when using the container.
- the inlet/outlet port may be a simple through hole provided in the wall or lid of the container, or may be a short pipe material for a joint airtightly inserted into the through hole. It is preferable that the inlet/outlet port has an appropriate connector so that an external pipe or the tip of an external syringe can be firmly and leak-free connected to the inlet/outlet port and can be easily attached and detached. However, it may be difficult to directly fix a large number of connectors to the stopper of the container because the connectors may interfere with each other. Therefore, in the embodiment of FIG. 2, an inlet/outlet port configuration that solves this problem is provided. In the example of the figure, three inlet/outlet ports 11 are provided in each container 10.
- Each container 10 has a configuration in which the opening of the container body 10a is closed by a stopper 10b.
- Each inlet/outlet port 11 in the figure has a tube member 11a that penetrates the stopper 10b in the inward and outward directions.
- each tube member is a soft tube, but the tube member at the penetrating portion may be made of a hard material.
- a luer connector 11b is provided at the distal end (the end farthest from the plug) of the tube member 11a on the outside world side. This allows the connectors of the three input/output ports to avoid interference with each other, facilitating the attachment and detachment of each connector, and does not significantly affect the plug when force is applied during attachment and detachment.
- a clip for closing the conduit is attached to the central tube member of the three tube members.
- any type of pipe joint (also called a connector or coupling) can be used as the connector for the inlet/outlet port, such as a luer connector (old ISO 594-1, old ISO 594-2, etc.), a connector to prevent misconnection (ISO 80369), a sterile connection joint, a one-touch type sterile disconnect joint, a push-in joint for tubing, a one-touch joint, etc.
- a stopper configured to be pierced by a syringe needle, such as a rubber stopper for a vial, and a syringe needle or a chemoclav bag spike that pierces it, is also included in the openable/closable connector.
- the configuration for opening and closing the inlet/outlet port is not particularly limited, and examples include a cap for closing the connection end of a disengaged connector, a pinch valve for closing the conduit of a tube provided as an inlet/outlet port, a clip, a stopcock, and the like. Switching of flow paths using a three-way stopcock may also be used as a mechanism for opening and closing the inlet/outlet port.
- a configuration for opening and closing the inlet/outlet port includes a case where an external conduit that remains connected to the inlet/outlet port is configured to be openable and closable.
- a combination of the above-mentioned stopper and an injection needle that pierces it is also included in the configuration for opening and closing the inlet/outlet port.
- FIG. 3 is a block diagram illustrating the configuration of an inlet/outlet port inside a container, and the container is shown in cross section for the purpose of explanation.
- the container 10 has a configuration in which the opening of the container body 10a is closed with a plug 10b, and the container 10 is provided with three inlet/outlet ports 111, 112, and 113.
- Each inlet/outlet port has a configuration in which a tube member (soft tube) penetrates the plug 10b in the inward/outward direction, and the tube member also extends inside the container.
- a tube joint 11b1 is provided at the distal end of the tube member on the outside world side.
- a device 12 for opening and closing the pipeline is provided on the outside world side of the tube member.
- the outside world sides of the other inlet/outlet ports 112 and 113 are similarly configured, but are not shown.
- the pipe members of the inlet/outlet ports 111, 112, and 113 extend into the container, and are characterized by their lengths.
- the inside of the container of the tube member of the inlet/outlet port 111 does not reach the liquid level of the contents (such as a cell suspension). Since gas can be introduced into the container without stirring the contents, this is preferable because it allows the volume pressure to be adjusted using gas without being affected by the amount of liquid inside the container.
- the gas may be O2 or CO2 required for cell culture, or air for pushing out the contents inside the container.
- the inside of the container of the tube member of the inlet/outlet port 112 penetrates into the contents from the liquid surface to a surface layer of an appropriate depth.
- Such an inside of the container is preferable in that it can supply liquid culture medium into the container without generating strong convection in the liquid culture medium, etc., and therefore without stirring up the settled cells. Therefore, such an inside of the container is suitable for operations such as replacement of liquid culture medium in cell culture, dilution to reduce the concentration of reprogramming factors, and washing of cells.
- the inside of the container of the tube member of the inlet/outlet port 113 is inserted into the liquid contents from the liquid surface to a surface layer of an appropriate depth, similar to the inlet/outlet port 112.
- Such an inside of the container can be preferably used when the liquid (supernatant) near the liquid surface of the liquid medium is to be gently removed from the container.
- two inlet/outlet ports 112 and 113 as in the example of Fig. 3(a), depending on the orientation of these inlet/outlet ports, a vortex can be generated in the liquid medium, and rotational culture can be performed.
- Fig. 3(a) depending on the orientation of these inlet/outlet ports
- the inside of the vessel of the tube member of the inlet/outlet port 113 reaches near the bottom of the vessel body. If such an inside of the vessel is used as a supply port for liquid medium or gas, it can generate a strong vertical convection current and send new liquid medium or gas to the settled cells. If it is used as an outlet for the contents in the vessel or a port for a connecting pipe line described later, the contents can be discharged until the liquid level of the contents reaches near the bottom of the vessel.
- the connecting pipeline is for connecting containers to each other and has a configuration that can be switched between a communicating state and a non-communicating state.
- the communicating state refers to a state in which a fluid can move from a container to a container in the next stage through the connecting pipeline
- the non-communicating state refers to a state in which a fluid cannot move from one container to another.
- the form of the connecting pipeline is not particularly limited, but examples include the following forms. 4(a), a connecting pipe J1 is provided to connect a container A1 and a container A2.
- various opening and closing devices J1a such as open/close valves (including shutters provided to open and close the pipe), stopcocks, clips, etc.
- a connecting pipe J1 is provided to connect container A1 and container A2.
- a connector J1b is provided on the line or at both ends of the connecting pipe J1.
- the connector J1b is not a so-called opening and closing device, when the connector J1b is connected, the connecting pipe J1 switches to a connected state, and when the connector J1b is separated, the connecting pipe J1 switches to a non-connected state as shown in Fig. 4(b).
- a connector may be a connector for connecting to an input/output port.
- the container A1 is provided with a connection pipe J1 for connecting the container A1 and the container A2.
- An injection needle J1c is attached to the tip of the connection pipe J1.
- the injection needle J1c is not pierced into the stopper of the container A2, and therefore the container A1 and the container A2 are not connected by the connection pipe J1, but are in a connectable state (i.e., a pierceable state).
- the injection needle J1c pierces the stopper of the container A2, and the connection pipe J1 is switched from a non-communicating state to a communicating state.
- the opening of the container body is sealed with a single stopper, but a stopper for piercing the injection needle J1c may be locally provided at a predetermined position of the lid that seals the opening.
- the inner diameter of the injection needle J1c may be any diameter that allows cells or microcarriers to pass through.
- An injection needle with an inner diameter of about 23 gauge (G) or larger is preferably used, and more preferably, about 18 G is exemplified.
- a container when a container is "connectable" to another container via a connecting pipeline, it means that the containers are in a pre-connection state, such as the relationship between the containers via the injection needle and the stopper (there is no connection because the injection needle has not been pierced into the stopper, but will be connected when the injection needle is pierced into the stopper) or the relationship between containers via disengaged connectors (there is no connection because the connectors are not engaged, but will be connected when the connectors are engaged), in which case the disconnected connecting pipelines are in a connectable state and the containers are connected when the connecting pipelines are joined.
- a pre-connection state such as the relationship between the containers via the injection needle and the stopper (there is no connection because the injection needle has not been pierced into the stopper, but will be connected when the injection needle is pierced into the stopper) or the relationship between containers via disengaged connectors (there is no connection because the connectors are not engaged, but will be connected when the connectors are engaged), in which case the disconnected
- inlet/outlet ports to which the connecting pipelines are connected in Figures 4(a) to (c) are for illustrative purposes only, and the position and configuration of the inlet/outlet ports to which the connecting pipelines are connected can be designed as appropriate. Furthermore, even if the connecting pipeline itself does not have a configuration that allows it to be opened and closed, it is sufficient that the inlet/outlet ports connected to the connecting pipeline can be opened and closed, thereby allowing the connecting pipeline to be switched between a connected state and a non-connected state.
- the feed mechanism in the present invention is a mechanism for transferring the contents from each container to the next container through the above-mentioned connecting pipes switched to the communicating state.
- the "each container” here refers to a source container that has a container to which the contents should be transferred in the next stage (i.e., a container other than the last container).
- Appropriate inlet/outlet ports can be used to adjust the pressure inside the next container when the contents are being transferred (pressure relief such as by venting air). If the next container is an empty flexible bag such as a cell culture bag, the next container will expand as the contents are transferred, so pressure adjustment may not be necessary. The same applies to pressure adjustment inside the container when the contents of the source container are removed by suction from the outside (pressure relief such as by letting air in), and appropriate inlet/outlet ports can be used. If the source container is a flexible bag such as a cell culture bag, the bag will shrink as the contents are removed, so pressure adjustment may not be necessary.
- the feed mechanism include a mechanism selected from the group consisting of the mechanisms (i) to (vi) shown below, and may also be a mechanism that combines two or more mechanisms selected from the group.
- a mechanism for pushing the contents out of the container is a mechanism for adding a fluid (gas, liquid medium, etc.) to the source container A1 (supplied through an inlet/outlet port) in the configuration of FIG. 4(a) and pushing out and moving the contents of the source sealed container A1 to the next container A2.
- the inlet/outlet port for injecting the gas may be dedicated to the feed mechanism, or may also be used as an inlet/outlet port for injecting gas for cell culture.
- the feed gas may be any gas that does not adversely affect cells, such as air (preferably clean air passed through a filter (oxygen concentration of about 20%, carbon dioxide concentration of about 5%)) or O 2 or CO 2 necessary for cell culture. It is also possible to supply a liquid medium into the source container A1 and push out and move the contents to the next container A2. In this case, the contents in the container A1 and the supplied liquid medium are mixed and pushed out, so it can be understood that dilution and movement of the contents in the container A1 are performed simultaneously. Any external pump device can be used to supply fluid to the source container A1, or a mechanism for sending out the material from the material source can be used. In the above explanation, the movement from container A1 to A2 is shown as an example, but the same can also be applied to a mechanism for moving the contents to the subsequent containers A3 to An.
- air preferably clean air passed through a filter (oxygen concentration of about 20%, carbon dioxide concentration of about 5%)
- O 2 or CO 2 necessary for cell culture.
- FIG. 5(a) A mechanism for pushing the contents out of the container (2) As shown in FIG. 5(a), this mechanism reduces the volume of the source container A1, and pushes out and moves the contents K1 to the next container (not shown).
- the container A1 is a flexible cell culture bag lying on the installation surface. The container A1 is pressed from the outside by a pressing head member 1 attached to the tip of a pressing device such as an air cylinder, and compressed to reduce its volume, and the contents K1 are pushed out through an input/output port 11 and a connecting pipe line J1 to the next container (not shown).
- the movement from the container A1 to the next container is shown as an example, but the mechanism may also be applied to a mechanism for moving contents to the following containers A3 to An.
- the dashed arrow in the figure indicates the contents to be pushed out.
- the container may be pressed automatically using a pressing device or the like, or may be pressed manually.
- a flexible bag is used as the container, but a container whose volume can be changed to push out the contents, such as one whose entire container has a syringe structure, may also be used.
- a mechanism for moving contents through a pump device As illustrated in FIG. 1, this mechanism moves the contents K1 of the source container A1 to the next container A2 by a pump device F1 provided in a connecting pipe J1 between two containers A1 and A2.
- the pump device that can be used for this mechanism is not particularly limited, but is preferably a pump device that can send the contents (suspension) without damaging the cells, and examples of such pump devices include a peristaltic pump and a syringe pump.
- the pump device may be operated manually or may be an automatic device equipped with a drive source and a control unit. In the example of FIG.
- a syringe pump is connected as the pump device F1 to a T-shaped pipe F1c inserted into the connecting pipe J1.
- Check valves F1a and F2b are connected before and after the T-shaped pipe F1c so that the contents are moved in one direction from the container A1 to the container A2 by the suction and discharge operation of the syringe pump.
- the movement from container A1 to A2 is shown as an example, but the mechanism may also be applied to a mechanism for moving the contents to subsequent containers A3, A4, etc.
- a stopcock may be used instead of the check valve.
- the arrows drawn on the check valves F1a and F2b indicate that the flow is restricted to one direction.
- the check valves F1a and F2b can be omitted, and the contents flow from container A1 to the syringe pump when the syringe pump is sucking, and the contents flow from the syringe pump to container A2 when the syringe pump is discharging.
- (iv) Mechanism for sucking contents from the next container This mechanism is a mechanism for sucking and moving the contents K1 of the source container A1 to the next container A2 by applying suction force from the next container A2 to the source container A1 in the configuration of FIG. 4(a), for example.
- the suction force for example, the air in the container A2 is sucked out of the container through the inlet/outlet port of the container A2, and thereby the suction force is applied to the contents K1 of the container A1 through the connecting pipe J1.
- the inlet/outlet port for sucking out the air to the outside of the container may be dedicated to the feed mechanism, or may also be used as an inlet/outlet port for injecting gas for cell culture.
- a container capable of sucking the contents K1 of the container A1 through the connecting pipe J1, such as one in which the entire next container A2 has a syringe structure may be used.
- the movement from container A1 to container A2 is shown as an example, but the present invention may also be applied to a mechanism for moving the contents to subsequent containers A3 to An.
- This mechanism is configured by the positional relationship of two containers and the connection of connecting pipes so that gravity can be utilized.
- this mechanism is a mechanism in which a next-stage container A2 is placed below a source container A1, and the contents of the source container are moved by gravity by descending to the next-stage container A2.
- the container A2 is placed directly below the container A1, a connecting pipe J1 is connected to the bottom surface of the container A1, and an opening/closing device J1a is provided on the connecting pipe J1.
- the opening/closing device J1a opens, the connecting pipe J1 is switched to a communicating state, and the contents in the container A1 fall into the next-stage container A2 through the connecting pipe J1 by gravity.
- a configuration in which the contents flow from container A1 into container A2 in the next stage by the principle of a siphon is also a mechanism for moving the contents by utilizing gravity.
- container A1 and container A2 are adjacent to each other, and the internal space of container A1 and the internal space of container A2 are isolated from each other by an openable and closable partition plate J1a1 of a shutter device J1a.
- the area J1 opened and closed by the partition plate J1a1 corresponds to the connection pipeline that can be switched between a connected state and a non-connected state in the present invention.
- the partition plate J1a1 is in a closed position.
- container A1 and container A2 become one internal space.
- container A2 is located below container A1, and the bottom surfaces of container A1 and container A2 form a single inclined surface so that cells M can slide down from container A1 to container A2.
- the partition plate J1a1 can be stopped in a state where it is retracted into the main body of the shutter device J1a by any amount.
- the connecting pipe is slightly open.
- the partition plate J1a1 is in the closed position.
- the partition plate J1a1 opens (i.e., the connecting pipe J1 switches to a connected state), and the contents in the container A1 move by gravity into the next container A2. Also, as shown in FIG. 6(b), if a liquid culture medium is also contained in the container A2, the cells M in the container A1 will slowly descend in the liquid culture medium by gravity and enter the next container A2.
- the partition plate J1a1 may be fully opened, but in terms of mainly moving the settled cells, as shown in FIG. 6(b), it is preferable to open it less than halfway to the full opening, or to the extent that the settled cells can move. This makes it possible to prevent the liquid medium in container A1 from mixing with the liquid medium in container A2.
- this mode of mainly cell movement also corresponds to the movement of contents.
- the movement from container A1 to A2 is shown as an example, but the mechanism may also be applied to move contents to subsequent containers.
- (vi) Mechanism for moving cells using magnetic force This mechanism uses a magnetic microcarrier and a magnet to move cells. Cells are attached to the magnetic microcarrier, and the microcarrier is pulled by magnetic force using a magnet. This allows the microcarrier and the cells attached thereto to be moved to the next vessel A2.
- the vessel A1 and the vessel A2 are adjacent to each other, and the openable and closable partition plate J1a1 of the shutter device J1a separates and communicates the vessel A1 and the vessel A2.
- the bottom surface of the vessel A2 does not need to be located lower than the bottom surface of the vessel A1.
- the bottom surfaces of the vessels A1 and A2 are one horizontal surface at the same level. If gravity is also used to move the cells, it is preferable that the bottom surfaces of the vessel A1 and the vessel A2 form one inclined surface, similar to the example of FIG. 6(b).
- step s1 When step s1 is performed in container A1, the partition plate J1a1 is in the closed position.
- step s1 is completed, as shown in FIG. 7, the partition plate J1a1 opens by an appropriate amount, and container A1 and container A2 communicate with each other.
- FIG. 7 shows a state in which the external magnet F1 moves and pulls the microcarrier with cells attached to it to container A2.
- the liquid culture medium if liquid culture medium is also contained in container A2, the liquid culture medium does not move violently even when the partition plate J1a1 opens, and therefore the microcarrier M1 with cells attached moves gently through the liquid culture medium by magnetic force and enters the next container A2.
- FIG. 7 shows an example of movement from container A1 to A2, but it may also be applied to a mechanism for moving contents to a subsequent container.
- the magnetic microcarrier that can be used in the present invention is not particularly limited, but may be, for example, a magnetic particle in which the surface of a magnetizable material such as ⁇ Fe 2 O 3 or Fe 3 O 4 known per se is coated with carboxydextran or crosslinked agarose, etc., and functional groups such as amino groups, carboxyl groups, hydroxyl groups, and epoxy groups are introduced, and a physiologically active substance such as an antibody that recognizes cells is bound via these functional groups, thereby expressing adsorptivity to surface antigens of target cells.
- a magnetizable material such as ⁇ Fe 2 O 3 or Fe 3 O 4 known per se
- functional groups such as amino groups, carboxyl groups, hydroxyl groups, and epoxy groups
- microcarriers examples include Corning Synthemax vitronectin substrate (Corning) and Atelocollagen Microspheres (KOKEN).
- examples of commercially available products include Global Eukaryotic Microcarriers TM (Global Cell Solutions).
- the magnet may be a permanent magnet or an electromagnet.
- An electromagnet is preferable for moving magnetic microcarriers because it can generate a magnetic field only when necessary (particularly when moving in the feed direction).
- the shape of the magnet (particularly the outer shape and area of the surface facing the container), the strength of the magnetic field, the moving speed, etc. can be appropriately determined so as to tow the microcarriers.
- the magnet moves from vessel to vessel in a predetermined course so that the microcarriers move from vessel to vessel.
- the magnet may be stationary and the vessels A1 and A2 may be moved to move the magnet relatively to the vessels.
- the magnet may be moved manually or automatically (moved by a driving source).
- electromagnets may be arranged along the direction in which the microcarriers are moved, and the electromagnets may be activated in sequence in the direction of movement, shifting the magnetic field as if the magnet were moving, thereby attracting the microcarriers.
- the cells may move as a cell suspension together with a liquid medium or microcarriers, or the cells may move primarily in the liquid medium, or the microcarriers and the cells attached thereto may move primarily in the liquid medium.
- Step s1 A step of contacting somatic cells with reprogramming factors in a liquid medium in a first container A1.
- Step s2 A step of reducing the concentration of the reprogramming factor in the liquid medium in the second container A2 to the (n-1)th container A(n-1).
- Step s3 A step of culturing the somatic cells in a liquid medium in the n-th container An to establish iPS cells.
- the culture in the production method (I) of the present invention is a suspension culture.
- suspension culture means culture carried out under conditions that maintain the state in which cells or cell aggregates are suspended in the culture medium, that is, culture under conditions that do not allow the formation of strong cell-substratum junctions between the cells or cell aggregates and the culture vessel.
- induced pluripotent stem cells are cells obtained by reprogramming mammalian somatic cells or undifferentiated stem cells through the introduction of specific factors (reprogramming factors). Induced pluripotent stem cells can differentiate into tissues and cells with various different forms and functions in the body, and have the ability to differentiate into cells of any lineage of the three germ layers (endoderm, mesoderm, and ectoderm).
- induced pluripotent stem cells including human iPSCs established by Yamanaka et al. by introducing four factors, Oct3/4, Sox2, Klf4, and c-Myc, into human fibroblasts (Takahashi K, Yamanaka S., et al. Cell, (2007) 131: 861-872.), Nanog-iPSCs established by selecting using the expression of Nanog as an indicator after the introduction of the above four factors (Okita, K., Ichisaka, T., and Yamanaka, S. (2007). Nature 448, 313-317.), iPS cells produced by a method that does not contain c-Myc (Nakagawa M, Yamanaka S., et al.
- iPS cells established by introducing six factors using a virus-free method (Okita K et al. Nat. Methods 2011 May;8(5):409-12, Okita K et al. Stem Cells. 31(3):458-66.) and the like can also be used.
- iPSC lines As induced pluripotent stem cell lines, various iPSC lines established by NIH, RIKEN, Kyoto University, etc. can be used.
- human iPSC lines include RIKEN's HiPS-RIKEN-1A line, HiPS-RIKEN-2A line, HiPS-RIKEN-12A line, Nips-B2 line, etc., and Kyoto University's AdiPS cells, 253G1 line, 253G4 line, 1201C1 line, 1205D1 line, 1210B2 line, 1383D2 line, 1383D6 line, 201B7 line, 409B2 line, 454E2 line, 585A1 line, 585B2 line, 606A1 line, 610B1 line, 648A1 line, 1231A3 line, FfI-01s04 line, etc.
- the induced pluripotent stem cells may be cells derived from a patient with a genetic disease.
- Cells induced to differentiate from pluripotent stem cells derived from a patient with a genetic disease can serve as a disease model that reflects the pathology of the disease, and are therefore suitable for screening therapeutic or preventive drugs for the disease.
- pluripotent stem cells derived from a patient with a genetic disease can be genetically repaired by genome editing using the CRISPR-Cas system or the like, and then differentiated into the desired cells, making it possible to use the cells as a therapeutic drug for the disease.
- somatic cells refers to cells other than reproductive cells that make up an animal. Somatic cells are not particularly limited, but include fetal (offspring) somatic cells, neonatal (offspring) somatic cells, and mature healthy or diseased somatic cells, as well as primary culture cells, passaged cells, and established cell lines. Specifically, somatic cells may be, for example, floating cells (e.g., blood cells, etc.) or adhesive cells, with floating cells being preferred.
- Somatic cells used in the manufacturing method of the present invention include, but are not limited to, for example, fibroblasts such as skin cells, skin cells, visual cells, brain cells, hair cells, oral mucosa, lung cells, liver cells, gastric mucosa cells, intestinal cells, spleen cells, pancreatic cells, kidney cells, neural stem cells, mesenchymal stem cells derived from wisdom teeth, etc., tissue stem cells, tissue progenitor cells, blood cells (e.g., hematopoietic stem cells, peripheral blood mononuclear cells (PBMCs) (including T cells and non-T cells), umbilical cord blood cells, etc.), epithelial cells, endothelial cells (e.g., vascular endothelial cells), muscle cells, etc.
- fibroblasts such as skin cells, skin cells, visual cells, brain cells, hair cells, oral mucosa, lung cells, liver cells, gastric mucosa cells, intestinal cells, spleen cells, pancreatic cells, kidney cells
- the cells when blood cells (e.g., peripheral blood mononuclear cells) are used as somatic cells, the cells are obtained by centrifuging whole blood (density gradient centrifugation).
- whole blood density gradient centrifugation
- the layer containing peripheral blood mononuclear cells which is the fraction layer obtained by centrifugation, can be moved directly to the first container A1 without coming into contact with the outside air.
- the species of origin of the somatic cells is not particularly limited, and may be, for example, cells from rodents such as rats, mice, hamsters, guinea pigs, etc.; lagomorphs such as rabbits; ungulates such as pigs, cows, goats, sheep, etc.; carnivores such as dogs and cats; tarsiers, long-tailed macaques, cynomolgus monkeys, rhesus monkeys, Japanese macaques, gibbons, spider monkeys, capuchin monkeys, orangutans, gorillas, chimpanzees, humans (all in the Stratorhinidae suborder), etc.; and primates such as lemurs, aye-ayes, and lorises (all in the Strangorhinidae suborder). Humans are preferred as the species of origin of the somatic cells.
- cells includes “cell populations.”
- a cell population may be composed of one type of cell, or may be composed of two or more types of cells.
- reprogramming factors include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, ESrrb, Nr5a2, Tbx3, and Glis1, and these reprogramming factors may be used alone or in combination.
- Combinations of reprogramming factors include those described in WO2007/069666, WO2008/118820, WO2009/007852, WO2009/032194, WO2009/058413, WO2009/057831, WO2009/075119, WO2009/079007, WO2009/091659, WO2009/101084, WO2009/101407, WO2009/102983, WO2009/114949, WO2009/117439, WO2009/126250, WO2009/126251, WO 2009/126655, WO2009/157593, WO2010/009015, WO2010/033906, WO2010/033920, WO2010/042800, WO2010/050626, WO2010/056831, WO2010/0689 55, WO2010/098419, WO2010/102267, WO2010/111409, WO2010/111422, WO2010/115050, WO2010/124290, WO2010/147395, WO2010/147
- Combinations of reprogramming factors include, for example, the following: (1) Oct3/4, Klf4, Sox2, c-Myc (Here, Sox2 can be replaced by Sox1, Sox3, Sox15, Sox17 or Sox18. Klf4 can be replaced by Klf1, Klf2 or Klf5. Furthermore, c-Myc can be replaced by T58A (active mutant), N-Myc or L-Myc.
- L-Myc is preferred for clinical use.
- the reprogramming factor is introduced into the somatic cells by contacting the reprogramming factor with the somatic cells in a liquid medium in a first container A1.
- the reprogramming factor introduced into the somatic cells may be in the form of a protein, or in the form of a nucleic acid (RNA or DNA) encoding the protein or an expression vector containing the nucleic acid.
- the immunogenic RNA introduced into the cells may activate the cell's defense mechanism, so RNA to circumvent the defense mechanism (e.g., E3 mRNA, K3L mRNA, B18 mRNA derived from vaccinia virus, a combination of these mRNAs, etc.) may be introduced into the somatic cells.
- RNA to circumvent the defense mechanism e.g., E3 mRNA, K3L mRNA, B18 mRNA derived from vaccinia virus, a combination of these mRNAs, etc.
- miRNAs or mimics thereof e.g., miR-302a-3p or mimic thereof, miR-302b-3p or mimic thereof, miR-302c-3p or mimic thereof, miR-302d-3p or mimic thereof, miR-367-3p or mimic thereof, combinations of these miRNAs or mimics, etc.
- miR-302a-3p or mimic thereof miR-302b-3p or mimic thereof, miR-302c-3p or mimic thereof, miR-302d-3p or mimic thereof, miR-367-3p or mimic thereof, combinations of these miRNAs or mimics, etc.
- the miRNA described above when introduced into somatic cells, it may be in the form of natural miRNA, miRNA with chemical modification of nucleic acid, miRNA mimic, or DNA encoding miRNA or an expression vector containing said DNA, but is preferably a miRNA mimic.
- a miRNA mimic is in the form of double-stranded RNA, and is typically composed of a guide strand made of natural RNA and a passenger strand with chemical modification (e.g., 2'-O methyl modification).
- the guide strand exhibits RNAi activity, while the passenger strand does not exhibit RNAi activity, so it mimics natural miRNA in cells.
- Expression vectors include, for example, viral vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, and Sendai virus, as well as plasmid vectors, episomal vectors, artificial chromosome vectors, and transposon vectors (piggyBac, piggyBat, TolII).
- viral vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, and Sendai virus
- plasmid vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, and Sendai virus
- plasmid vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, and Sendai virus
- plasmid vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus,
- Promoters used in expression vectors include, for example, EF1 ⁇ promoter, ACTB promoter, UbqC promoter, PGK promoter, CAG promoter, SR ⁇ promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (moloney murine leukemia virus) LTR, HIV LTR, and HSV-TK (herpes simplex virus thymidine kinase) promoter.
- the promoter is preferably a pol III promoter (e.g., SNR6, SNR52, SCR1, RPR1, U3, U6, H1 promoter, etc.).
- RNA such as mRNA and miRNA can be produced by chemical synthesis or by in vitro transcription (IVT). Alternatively, it can be expressed in an organism (Escherichia coli or cultured mammalian cells) and then purified.
- Chemical synthesis methods include, for example, methods using nucleoside phosphoramidites and solid-phase supports.
- modified nucleotides can be introduced into any base site in an RNA sequence by using chemically modified nucleoside phosphoramidites (for example, 2'-O-methylated phosphoramidites in the sugar moiety).
- Nucleic acids e.g., the above-mentioned mRNA, miRNA, DNA encoding these, etc.
- expression vectors containing the nucleic acids, or proteins e.g., reprogramming factors
- methods include calcium phosphate-mediated transfection, electroporation, liposome transfection, lipofection, gene gun, microinjection, viral vector method, virus-like particle method, Agrobacterium method, agroinfiltration method, PEG-calcium method, sonoporation method, lipid nanoparticle method, etc.
- the culture in the production method (I) of the present invention may be cultured under feeder-free conditions and/or xeno-free conditions for all or part of the period. From the viewpoint of clinical use, it is preferable that the production method of the present invention is carried out under feeder-free and xeno-free conditions for the entire period.
- feeder-free means a medium or culture conditions that do not contain other cell types (i.e., feeder cells) that play a supporting role and are used to prepare the culture conditions for the cells to be cultured.
- xeno-free means a medium or culture conditions that do not contain components derived from organisms different from the organism species of the cells to be cultured.
- the basal medium used in the production method (I) of the present invention is not particularly limited, and examples thereof include Essential 8 medium (CTS TM Essential 8 TM Medium, Essential 8 TM Medium, Essential 8 TM Flex Medium, Essential 6 TM Medium) (Thermo Fisher Scientific), StemFit (registered trademark) AK02 medium (Ajinomoto Co., Inc.), StemFit (registered trademark) AK03 medium (Ajinomoto Co., Inc.), StemFit (registered trademark) Basic03 medium, CTS (registered trademark) KnockOut SR XenoFree Medium (Gibco), mTeSR1 medium, TeSR1 medium (Stem Cell Technologies), Iscove's modified Dulbecco's
- suitable media include Improved MEM (GE Healthcare), and Improved MEM (Thermo Fisher Scientific).
- RPMI-1640 medium Eagle MEM (EMEM), Dulbecco's modified MEM, Glasgow's MEM (GMEM), ⁇ -MEM, 199 medium, IMDM, DMEM, Hybridoma Serum free medium, KnockOut TM DMEM, Advanced TM medium (e.g., Advanced MEM, Advanced RPMI, Advanced DMEM/F-12), Ham's Medium F-12, Ham's Medium F-10, Ham's Medium F12K, DMEM/F-12, ATCC-CRCM30, DM-160, DM-201, BME, Fischer, McCoy's 5A, Leibovitz's L-15, RITC80-7, MCDB105, MCDB107, MCDB131, MCDB15 3.
- EMEM Eagle MEM
- GMEM Glasgow's MEM
- ⁇ -MEM 199 medium
- IMDM Glasgow's MEM
- DMEM Hybridoma Serum free medium
- KnockOut TM DMEM Advanced
- MCDB201 NCTC109, NCTC135, Waymouth's Medium (e.g. Waymouth's MB752/1), CMRL medium (e.g. CMRL-1066), Williams' medium E, Brinster's BMOC-3 um, E8 Medium, StemPro 34, MesenPRO RS (Thermo Fisher Scientific), ReproFF2, Primate ES Cell Medium, ReproStem (ReproCELL Co., Ltd.), ProculAD (Rohto Pharmaceutical Co., Ltd.), MSCBM-CD, MSCGM-CD (Lonza), EX-CELL302 medium (SAFC) or EX-CELL-CD-CHO (SAFC), ReproMed TM iPSC Medium (ReproCELL Co., Ltd.), and mixtures thereof.
- Waymouth's Medium e.g. Waymouth's MB752/1
- CMRL medium e.g. CMRL-1066
- Williams' medium E Brinster's BMOC-3 um
- E8 Medium Stem
- the basal medium used in the production method (I) of the present invention can be supplemented with physiologically active substances and nutritional factors necessary for cell survival or proliferation, if necessary.
- physiologically active substances and nutritional factors necessary for cell survival or proliferation may be added to the medium in advance, or may be added during cell culture.
- the method of adding them during culture may be in any form, such as one solution or a mixed solution of two or more types, and may be continuous or intermittent addition.
- physiologically active substances include insulin, IGF-1, transferrin, albumin, coenzyme Q10, various cytokines (interleukins (IL-2, IL-7, IL-15, etc.), stem cell factor (SCF), activin, etc.), various hormones, and various growth factors (leukemia inhibitory factor (LIF), basic fibroblast growth factor (bFGF), TGF- ⁇ , etc.).
- Nutritional factors include sugars, amino acids, vitamins, hydrolysates, lipids, and the like. Examples of the sugar include glucose, mannose, fructose, and the like, and one or more of them may be used in combination.
- amino acids examples include L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, and L-valine, and these may be used alone or in combination of two or more.
- vitamins examples include d-biotin, D-pantothenic acid, choline, folic acid, myo-inositol, niacinamide, pyrodoxal, riboflavin, thiamine, cyanocobalamin, and DL- ⁇ -tocopherol, and these may be used alone or in combination of two or more kinds.
- Hydrolysates include those derived from hydrolyzed soybeans, wheat, rice, peas, corn, cottonseed, yeast extracts, and the like.
- the lipids include cholesterol, linoleic acid, and linolenic acid.
- antibiotics such as kanamycin, streptomycin, penicillin, or hygromycin may be added to the medium as necessary.
- an acidic substance such as sialic acid is added to the medium, it is desirable to adjust the pH of the medium to a neutral range suitable for cell growth, between pH 5 and 9, preferably between pH 6 and 8.
- the medium may be a serum-containing medium (e.g., fetal bovine serum (FBS), human serum, horse serum) or a serum-free medium. From the viewpoint of preventing contamination with components derived from different kinds of animals, it is preferable that the medium does not contain serum, or that serum derived from the same kind of animal as the cells to be cultured is used.
- serum-free medium means a medium that does not contain unconditioned or unpurified serum.
- the serum-free medium may contain purified blood-derived components or animal tissue-derived components (e.g., growth factors).
- the medium may or may not contain serum substitutes, as well as serum.
- Serum substitutes include, for example, albumin substitutes such as albumin, lipid-rich albumin, and recombinant albumin, vegetable starch, dextran, protein hydrolysates, transferrin or other iron transporters, fatty acids, insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thioglycerol, or equivalents thereof.
- Specific examples of serum substitutes include those prepared by the method described in WO98/30679, as well as commercially available products such as Knockout Serum Replacement [KSR] (Life Technologies), Chemically-defined Lipid Concentrated (Life Technologies), and Glutamax (Life Technologies).
- KSR Knockout Serum Replacement
- biologically derived factors include platelet-rich plasma (PRP) and culture supernatant components of human mesenchymal stem cells.
- the medium may contain a scaffold material (hereinafter also referred to as "scaffold") used for suspension culture of cells, and the scaffold material refers to a material or substrate that functions as a scaffold for cells in cell culture.
- the scaffold material is not particularly limited as long as it can be used for suspension culture of cells as described above (in other words, it may be free in the medium), but examples include those that contain or are made of synthetic resin, and those that are made of flexible materials such as collagen.
- the scaffold material may contain or be made of atelocollagen, and specifically, atelocollagen molded into a shape suitable for use as a scaffold material.
- the scaffold material is a material other than nanofibers.
- Synthetic resin refers to a material whose main component is a polymer (hereinafter, also simply referred to as "polymer”) obtained by polymerizing (including polycondensation) a polymerizable monomer (hereinafter, also simply referred to as “monomer”).
- polymer a polymer obtained by polymerizing (including polycondensation) a polymerizable monomer (hereinafter, also simply referred to as “monomer”).
- the above polymer also includes copolymers of one or more polymerizable monomers.
- the scaffold material may be one whose main component is an inorganic material such as glass or silicone.
- polymers examples include polymers made of one or more polymerizable monomers of (un)saturated hydrocarbons, aromatic hydrocarbons, (un)saturated fatty acids, aromatic carboxylic acids, (un)saturated ketones, aromatic ketones, (un)saturated alcohols, aromatic alcohols, (un)saturated amines, aromatic amines, (un)saturated thiols, aromatic thiols, and organosilicon compounds.
- these polymers may be used alone or in combination of two or more.
- the two or more polymers may be mixed and used, or the skeletons of the two or more polymers may be chemically bonded to form a polymer.
- Scaffolding materials may be produced by known methods, or commercially available products may be used. Examples of commercially available products include Cytodex-1 (GE Healthcare) and Corning (registered trademark) low concentration Synthemax (registered trademark) II microcarriers (Corning, Inc.).
- a scaffold material containing atelocollagen can be prepared by covering (coating) all or part of the surface of the scaffold material with atelocollagen to prepare a scaffold material containing the desired atelocollagen.
- the purity of the atelocollagen used for coating is not particularly limited, but it is preferable that it is highly pure (e.g., 90% or more, more preferably 95% or more, and most preferably 100%).
- the scaffold material may be coated with any cell-supporting substrate such as an extracellular matrix (ECM) in addition to atelocollagen.
- ECM extracellular matrix
- the material is substantially free of native collagen (e.g., 10% or less, more preferably 5% or less (e.g., 4%, 3%, 2%, 1%, 0%).
- the cell support substrate can be any material intended for the attachment of stem cells or feeder cells (if used).
- Such cell support substrates include collagen, gelatin, poly-L-lysine, poly-D-lysine, laminin (or a partial structure of laminin), and fibronectin, as well as mixtures thereof, such as Matrigel, and lysed cell membrane preparations (Lancet, 2005.365.9471.1636-1641).
- purity means a mass percent concentration (hereinafter, “mass percent concentration” is simply referred to as “concentration”) as an indicator of high quality (low rate of impurity contamination), but may also refer to the concentration of a specific component (e.g., atelocollagen) in a mixture (e.g., a mixture of atelocollagen and other cell support substrates).
- concentration concentration
- the shape of the scaffold material is not particularly limited, and examples thereof include cylindrical, oval, and spherical shapes, with spherical shapes being preferred. Specific examples of such spherical scaffold materials include microcarriers.
- the inventors have previously found that pluripotent stem cells can be grown using microcarriers with diameters of 105 ⁇ m or less, 105 to 250 ⁇ m, 250 to 425 ⁇ m, and 425 to 600 ⁇ m in culture using a bioreactor.
- the size of the scaffold material is also not particularly limited, but when using a spherical scaffold material such as a microcarrier, the particle size (diameter) of the scaffold material is typically 50 to 1000 ⁇ m, may be 70 to 700 ⁇ m, and is preferably 100 to 400 ⁇ m. In addition, in a preferred embodiment, the particle size is 600 ⁇ m from the viewpoint of cell proliferation rate.
- the particle size can be measured by the Coulter counter method described in the international standard ISO 13319 "Measurement of particle size distribution - Electrical detection zone method".
- Type I collagen molecules are composed of about 95% helical (helix) parts and about 5% non-helical parts (telopeptides). This non-helical part is a region with high antigenicity and is cleaved by proteases (protein-degrading enzymes).
- proteases protein-degrading enzymes.
- the atelocollagen contained in the scaffold material is a natural polymeric material with extremely low antigenicity that is highly purified after digestion and removal of the highly antigenic telopeptide part with proteases such as pepsin (Matrix, 1992, 12. 274-281).
- collagen present in the living body is an insoluble fibrous protein with a "triple helix structure" in which three polypeptide chains are wound in a helix, and is also called native collagen.
- the origin of the atelocollagen used in the present invention is not limited, and examples include those derived from mammals (e.g., humans, mice, rats, monkeys, cows, horses, pigs, dogs, etc.). From the viewpoint of preventing contamination with components derived from different animals, it is preferable to use atelocollagen derived from the same origin as the cells to be cultured.
- atelocollagen may be produced by known methods, or a commercially available product may be used.
- atelocollagen can be purified by treating collagen extracted from cells or tissues containing atelocollagen, or collagen secreted from cultured cells, with a protease.
- the concentration of atelocollagen in the scaffold material ((mass of atelocollagen/mass of scaffold material containing atelocollagen) x 100) is not particularly limited as long as it is a concentration that exhibits a cell death inhibitory effect on cells. Such a concentration can be appropriately set by a person skilled in the art using the methods described in the Examples and conventionally known methods.
- the concentration of atelocollagen in the scaffold material is, for example, 0.1% or more (e.g., 0.1%, 1%, 3%, 5%, 10%, 20%, 25%, 30% or more) and 100% or less.
- the concentration of atelocollagen in the cell support substrate is 90% or more (e.g., 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 97%, 98%, 99% or 100%).
- the scaffold material is substantially composed of atelocollagen, and "substantially composed of atelocollagen" does not mean that the atelocollagen concentration is 100%, but rather that the concentration is close to 100% (e.g., 95% or more, preferably 95.5% or more (e.g., 96%, 97%, 98%, 99% or 100%)).
- the concentration of atelocollagen in the medium is not particularly limited, and by appropriately setting the concentration of atelocollagen, it is possible to control the cell proliferation rate.
- the concentration of atelocollagen in the medium is, for example, 0.01 to 20%, preferably 0.05 to 5%, and more preferably 0.1 to 2%. In addition, it is also preferable that the concentration of atelocollagen in the medium is 0.5% to 20%, 1% to 15%, or 5% to 10%.
- the conditions for the culture (typically, steps s1 to s4) in the production method (I) of the present invention are not particularly limited, but are about 30 to 40° C., preferably about 37° C., and culture is performed in an atmosphere of CO 2 -containing air, with a CO 2 concentration of preferably about 2 to 5%.
- the temperature in each container can be controlled by referring to a conventionally known temperature control method, such as temperature control at room temperature, providing a heater to each container, and temperature control of the material supplied to the container.
- the cell culture density in the culture (typically, steps s1 to s4) in the production method (I) of the present invention is not particularly limited as long as the cells can grow, and is usually 1.0 ⁇ 10 2 to 1.0 ⁇ 10 7 cells/cm 2 , preferably 1.0 ⁇ 10 3 to 1.0 ⁇ 10 6 cells/cm 2 , and more preferably 1.0 ⁇ 10 4 to 1.0 ⁇ 10 5 cells/cm 2 .
- the period for performing step s1 in the production method (I) of the present invention is not particularly limited as long as iPS cells are established in step s3, but typically includes, for example, 30 minutes to 24 hours, 2 hours to 6 hours, etc.
- the method of reducing the concentration of the reprogramming factor in the liquid medium is not particularly limited, but can be achieved by replacing the medium used in step s1 with a medium to which no reprogramming factor is added.
- the medium replacement may be a complete replacement of the medium used in step s1, or a partial replacement, as long as the concentration of the reprogramming factor in the medium is reduced by dilution, thereby reducing or terminating the contact (frequency) between the somatic cells and the reprogramming factor.
- "reducing the concentration of the reprogramming factor” may also include a state in which no reprogramming factor is present in the medium.
- the concentration of the reprogramming factor in the medium in step s2 is, for example, 1/10 or less, preferably 1/100 or less, more preferably 1/300 or less, even more preferably 1/1000 or less, and even more preferably 1/3000 or less, compared to step s1.
- the process may proceed from step s2 to step s3 if the reprogramming factor in the medium is reduced to a desired concentration as described above.
- Step s3 in the production method (I) of the present invention is a step of culturing the somatic cells in a liquid medium to establish iPS cells, and the establishment of induced pluripotent stem cells can be appropriately confirmed by the expression of reprogramming factors introduced by a method known per se (e.g., Oct3/4, SOX2, Nanog, TRA-1-60, TRA-1-81, SSEA3, SSEA4, alkaline phosphatase, etc.).
- the period for carrying out step s3 in the production method (I) of the present invention is not particularly limited as long as iPS cells are established, but is typically, for example, 10 days or more, preferably 14 days or more. There is also no particular upper limit, but it is typically 40 days or less, preferably 30 days or less.
- each material supply source (G1 to Gn) to each container are materials necessary for carrying out each of the above-mentioned steps (e.g., somatic cells, culture medium, reprogramming factors, etc.).
- somatic cells e.g., somatic cells, culture medium, reprogramming factors, etc.
- gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of the gases can be omitted. The same applies to the other containers described below.
- the manufacturing method (I) further includes a step s4 of expanding the iPS cells after the step s3 of establishing the iPS cells.
- the number of times of the expansion is p (p is an integer of 1 or more, i.e., p ⁇ 1).
- the expansion culture step s4 will be described in detail below.
- the configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in the expansion culture step s4 can be referenced from the configurations shown in the explanation of steps s1 to s3 above, and therefore will not be explained here.
- step s4 some or all of the contents (i.e., culture medium containing iPS cells) in the nth sealed container (An) established in step s3 are transferred to container B1 and cultured until the desired cell density is reached.
- the contents transferred at each step are the same as those from step s3 to step s4.
- step s4 of expanding iPS cells in step s4 of expanding iPS cells, p containers (B1 to Bp) corresponding to the number p of times of expansion culture are connected in series via connecting pipelines after the nth container An in which the above-mentioned step s3 is carried out.
- Necessary material supply sources (Gb1 to Gbp) are connected to the input/output ports of each container (B1 to Bp) so that the process corresponding to each container is carried out. Although only one material supply source is shown to be connected to each container in the figure, the number is not limited. Input/output ports that have not reached the liquid level are not shown in Figure 8.
- the iPS cells are moved sequentially in one direction by the feed mechanisms Fn and Fb1 to Fb(p-1), respectively, and one expansion culture is carried out inside each of the containers (B1 to Bp), thereby carrying out a total of p expansion cultures, completing step s4 in container Bp and obtaining the desired number of iPS cells.
- the number of times expansion culture is performed is not particularly limited as long as the desired number of iPS cells can be obtained, but in a normal processing operation, it is preferably about 1 to 5 times, more preferably about 2 to 5 times.
- the period for which step s4 is performed in the production method (I) of the present invention is not particularly limited as long as the desired number of iPS cells can be obtained, but is typically, for example, 1 to 40 days, 3 to 20 days, or 5 to 10 days.
- each material supply source Gb1 to Gbp
- each container the materials supplied from each material supply source (Gb1 to Gbp) to each container are necessary for the expansion culture, and are mainly culture media. Gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of these gases can be omitted.
- the manufacturing method (II) comprises steps s1 to s3 in the manufacturing method (I) described above, and step s5 of inducing differentiation of iPS cells.
- step s4 of expansion culture is added after steps s1 to s3, and thus the manufacturing method (II) comprises the above steps s1 to s4, and step s5 of inducing differentiation of iPS cells.
- the production method (II) may be an independent cell production method that does not have production method (I) as a preceding step.
- this corresponds to the cell production method of the present invention in which [multiple cell production steps are divided into multiple containers, cells are moved from container to container in the order of the steps, and the steps associated with each container are carried out sequentially].
- FIG. 9 is a block diagram for explaining the manufacturing method (II).
- a container C1 for carrying out step s5 is further connected through a connecting pipeline after the last container X1 among the containers used in the manufacturing method (I).
- the container X1 may be the last container An of steps s1 to s3 in FIG. 1, or the last container Bp of step s4 in FIG. 8.
- the container C1 has one or more openable and closable inlet/outlet ports, and materials necessary for differentiation induction in step s5 are supplied to the inside of the container C1 through the inlet/outlet ports (external material supply sources are not shown). In FIG. 9, inlet/outlet ports that have not reached the liquid level are not shown.
- the iPS cells are transferred from the container X1 to the container C1 by the feed mechanism Fx1, and the above-mentioned step s5 is carried out in the container C1.
- the materials supplied to each container are necessary for differentiation induction.
- materials themselves necessary for differentiation induction reference can be made to the prior art, including those described below.
- Gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of the gases can be omitted.
- the desired cells or organoids can be produced by inducing differentiation of artificial pluripotent stem cells.
- the obtained cells may be undifferentiated cells such as stem cells and precursor cells, or may be terminally differentiated cells.
- the undifferentiated cells to be removed in the production method (II) of the present invention refer to cells other than stem cells and precursor cells intended to be produced by the production method (II) of the present invention.
- the term "differentiated cells” may be used as a term that encompasses both undifferentiated cells and terminally differentiated cells that can be produced by the production method (II) of the present invention. They may also be germ cells.
- undifferentiated cells refers to cells that have not reached terminal differentiation in the cell lineage, and examples of undifferentiated cells include stem cells and precursor cells excluding pluripotent stem cells.
- stem or progenitor cells include epiblast-like cells, primordial germ cells (also referred to as “primordial germ cell-like cells”), germline stem cells, ectodermal (row) cells such as neural crest cells, neural stem cells, neural progenitor cells, glial progenitor cells, retinal stem cells, corneal stem cells, keratinocyte epidermal stem cells, melanocyte stem cells, mammary stem cells, mesodermal (row) cells such as hematopoietic progenitor cells, bone marrow stem cells, lymphatic stem cells, B progenitor cells, T progenitor cells, mesenchymal stem cells, cardiac stem cells, cardiac progenitor cells, vascular endothelial progenitor cells, vascular pericytes, platelet progenitor cells
- terminal differentiated cells refers to cells that have reached terminal differentiation in the cell lineage.
- terminally differentiated cells include, but are not limited to, osteoblasts, chondrocytes, adipocytes, hepatic mesothelial cells, bile duct epithelial cells, hepatic stellate cells, hepatic sinusoidal endothelial cells, Kupffer cells, pit cells, vascular endothelial cells, blood cells (e.g., red blood cells, platelets, white blood cells, mast cells, dendritic cells, etc.), pancreatic ductal epithelial cells, pancreatic ductal cells, acinar center cells, acinar cells, laminar cells, and endothelial cells.
- Gerhans include islets of Gerhans, cardiac muscle cells, fibroblasts, smooth muscle cells, type I alveolar epithelial cells, type II alveolar epithelial cells, Clara cells, ciliated epithelial cells, basal cells, goblet cells, neuroendocrine cells, Kruczyk cells, renal tubular epithelial cells, urothelial cells, columnar epithelial cells, glomerular epithelial cells, glomerular endothelial cells, octopus podocytes, mesangial cells, nerve cells, glial cells (e.g., astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells, etc.).
- White blood cells include lymphocytes (e.g., B cells, T cells, NK cells, etc.), granulocytes (e.g., neutrophils, eosinophils, basophils, etc.), monocytes, etc.
- the cells or organoids (target cells or organoids) obtained by the production method (II) of the present invention are neural crest cells, neural precursor cells, nerve cells, cerebral cortical organoids, hematopoietic precursor cells, platelets, T cells, epiblast-like cells, primordial germ cells, or cardiomyocytes. They may also be inner enamel epithelium, ameloblasts, stratum intermedium cells, stellate reticulum cells, outer enamel epithelium, dental papilla cells, or odontoblasts.
- Organoids refers to a structure formed by the accumulation of cells, and typically has a structure and function similar to that of an organ in the body.
- Organoids obtained by the differentiation induction method of the present invention include, for example, neural organoids (e.g., cerebral cortex organoids, cerebellar organoids, spinal cord organoids, midbrain organoids, choroid plexus organoids, hippocampal organoids, hypothalamic organoids, anterior pituitary organoids, and basal ganglia organoids, etc.), lung organoids, liver organoids, airway epithelial organoids, intestinal organoids, pancreatic organoids, kidney organoids, airway organoids, stomach organoids, thyroid organoids, thymus organoids, testicular organoids, esophageal organoids, skin organoids, fallopian tube organoids, ovarian organoids, salivary gland organoids, ocular vesicle organoids, optic cup organoids, bladder organoids, prostate organoids, cartilage organoids
- Whether a certain structure is an organoid can be confirmed, for example, by observing with a microscope to see whether a layer structure is formed or not, or by examining the expression of marker proteins. Specifically, in the case of cerebral cortical organoids, a dome-shaped neuroepithelium with Foxg1 expressed throughout is observed, and this neuroepithelium forms a layer structure. In this structure, a layer of neural precursor cells corresponding to the Pax6- and Sox2-positive ventricular zone is observed inside the epithelium, and on the outside of this, a first layer consisting of Reelin/Carletinin-positive Cajal-Retzius cells and a Ctip2/Tbr1-positive deep layer are observed.
- markers include HHEX, SOX2, HNF4A, AFP, and ALB; in pancreatic organoids, markers include PDX1, SOX17, and SOX9; in organoids that differentiate into the intestine, markers include CDX2 and SOX9; and in kidney organoids, markers include Pax2 and Six2.
- pluripotent stem cells into neural crest cells can be induced by the methods described in Fukuta M. et al., PLoS One, 2014, 9(12): e112291 and Kamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47.
- pluripotent stem cells can be seeded in a culture vessel and cultured as adherent cells (suspension culture using a scaffold material), and then differentiated into neural crest cells by culture as adherent cells (suspension culture using a scaffold material) in a medium containing a TGF ⁇ inhibitor and a GSK3 ⁇ inhibitor.
- neural crest cells From neural crest cells, it is also possible to produce cells such as mesenchymal stem cells, neural progenitor cells, nerve cells, glial cells, bone cells, chondrocytes, corneal cells, and melanocytes. For example, differentiation into these cells can be induced based on the methods described in Fukuta M. et al., PLoS One, 2014, 9(12): e112291, Horikiri T. et al., PLoS One, 2017, 12(1): e0170342, and Kamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47.
- neural crest cells are seeded onto a plate coated with fibronectin, the medium is replaced with DMEM/F12 supplemented with N-2 Supplement, BDNF, GDNF, NT-3, and NGF, and the cells are cultured at 37°C under 5% CO2 for approximately 14 days to obtain neural progenitor cells and neurons.
- neural crest cells can be plated and cultured in CDM medium containing SB431542 and CHIR99021 for one day, after which the medium is replaced with Neurobasal medium supplemented with B-27 Supplement, N-2 Supplement, L-glutamine, Penicillin/Streptomycin, BDNF, GDNF, NT-3, and NGF, and cultured at 37°C in 5% CO2 for approximately 35 days to obtain neural progenitor cells and neurons.
- CDM medium containing SB431542 and CHIR99021 for one day, after which the medium is replaced with Neurobasal medium supplemented with B-27 Supplement, N-2 Supplement, L-glutamine, Penicillin/Streptomycin, BDNF, GDNF, NT-3, and NGF, and cultured at 37°C in 5% CO2 for approximately 35 days to obtain neural progenitor cells and neurons.
- Neural crest cells are seeded in a culture vessel and cultured for one day in CDM medium containing SB431542 and CHIR99021. After one day, the medium is replaced with ⁇ MEM containing FBS.
- Mesenchymal stromal cells can be obtained about 14 days after the start of differentiation induction.
- a method for differentiating pluripotent stem cells into T cells includes, for example, a method comprising (1) a step of differentiating pluripotent stem cells into hematopoietic progenitor cells, and (2) a step of differentiating the hematopoietic progenitor cells into T cells.
- the step (1) may be, for example, a step of culturing pluripotent stem cells in an induction medium for hematopoietic progenitor cells, as described in WO2013/075222, WO2016/076415, Liu S. et al., Cytotherapy, 17 (2015); 344-358, etc.
- the step (2) may be, for example, a step (2-1) of inducing CD4CD8 bi-positive T cells from hematopoietic progenitor cells, and a step (2-2) of inducing CD8 positive T cells from CD4CD8 bi-positive T cells, as described in WO2016/076415, etc.
- the step of inducing differentiation of pluripotent stem cells into platelets can be, for example, (1) a method including a step of differentiating pluripotent stem cells into hematopoietic progenitor cells and a step of differentiating hematopoietic progenitor cells into platelets.
- the step (2) can be, for example, a step of culturing hematopoietic progenitor cells in a medium containing TPO and/or SCF for about 7 to 15 days, as described in WO2012/157586, US2014/127815, Nakamura, Eto, et al. Cell Stem Cell 14, 535-548 (2014), etc. This step can produce a cell population containing megakaryocytes and platelets.
- the process of inducing differentiation of pluripotent stem cells into primordial germ cells includes, for example, a method comprising the steps of: (1) culturing pluripotent stem cells in a culture medium containing activin A and a GSK3 ⁇ inhibitor for approximately 40 to 60 hours to differentiate them into epiblast-like cells; and (2) culturing the epiblast-like cells in a culture medium containing BMP for approximately 4 to 8 days to differentiate them into primordial germ cells (primordial germ cell-like cells), as described in WO2017/002888 and the like.
- Examples of methods for inducing differentiation of pluripotent stem cells into cardiomyocytes include the methods described in WO2015/141827 and Laflamme MA and Murry CE, Nature. 473(7347):326-35 (2011).
- Other examples include a method for producing cardiomyocytes by forming embryoid bodies through suspension culture of induced pluripotent stem cells, a method for producing cardiomyocytes in the presence of a substance that suppresses BMP signaling (WO2005/033298), a method for producing cardiomyocytes by sequentially adding Activin A and BMP (WO2007/002136), and a method for producing cardiomyocytes in the presence of a substance that promotes activation of the canonical (classical) Wnt signaling pathway (WO2007/126077).
- marker proteins for cardiomyocytes include NKX2.5 (a cardiac muscle-specific transcription factor) and TNNT2 (troponin T), while marker proteins for cardiac progenitor cells include KDR (a receptor for vascular endothelial growth factor (VEGF)) and ISL1 (a LIM homeodomain transcription factor).
- NKX2.5 a cardiac muscle-specific transcription factor
- TNNT2 troponin T
- marker proteins for cardiac progenitor cells include KDR (a receptor for vascular endothelial growth factor (VEGF)) and ISL1 (a LIM homeodomain transcription factor).
- liver organoids as described in WO2013/047639, liver progenitor cells (organ cells), mesenchymal stem cells, and vascular endothelial cells can be induced from pluripotent stem cells, and the mixture can be cultured in suspension to produce liver organoids.
- organ cells as described in WO2013/047639, liver progenitor cells (organ cells), mesenchymal stem cells, and vascular endothelial cells can be induced from pluripotent stem cells, and the mixture can be cultured in suspension to produce liver organoids.
- the production method (II) of the present invention may involve culturing under feeder-free conditions and/or xeno-free conditions for all or part of the period. From the viewpoint of clinical use, it is preferable that the differentiation induction method of the present invention is carried out under feeder-free and xeno-free conditions for the entire period.
- a cell or organoid obtained by the production method (II) of the present invention (hereinafter, also referred to as the "differentiated cell or organoid of the present invention”).
- the manufacturing method (II) of the present invention may include a step of recovering the obtained target cells or organoids.
- the recovered cells may be cryopreserved using a cell cryopreservation solution.
- the cells recovered in the container may be subjected to cell counting using a cell counter, or may be labeled with an antibody against a cell surface marker and purified by flow cytometry, mass cytometry, magnetic cell separation, or the like.
- step s5 shown in FIG. 9(a) may be step s5a of inducing differentiation of iPS cells into ectodermal cells, mesodermal cells, or endodermal cells.
- step s5a may be followed by step s5b of further differentiation induction.
- Step s5b is a step of inducing differentiation of the ectodermal cells, etc. obtained in step s5a to obtain further other cells.
- a container C2 for carrying out a further differentiation induction step s5b is further connected via a connecting pipe Jc1 after the container C1 in which the step s5a is carried out.
- the container C2 has one or more openable and closable input/output ports (reference numbers omitted). Materials necessary for the differentiation induction in step s5b are supplied to the inside of the container C2 through the input/output ports.
- the cells (the ectodermal, mesodermal, or endodermal cells) are moved from the container C1 to the container C2 by a feed mechanism Fc1, and the step s5b is carried out in the container C2.
- step s5b a further differentiation induction step may be added as necessary, in which case the containers corresponding to the added step are connected in the same manner as described above.
- a step s6 of removing undifferentiated cells is added after the step s5, and a container D1 associated with the step s6 is connected in the same manner as described above.
- a container D1 for carrying out the step s6 is further connected through a connecting pipe Jx2 after the last container X2 (e.g., container C1 or C2) among the containers used in the differentiation induction step s5 (including the steps s5a and s5b).
- Differentiated cells are moved from the container X2 to the container D1 by the feed mechanism Fx2.
- the container D1 has one or more openable inlet/outlet ports, and materials necessary for the step s6 are supplied into the container D1 through the inlet/outlet ports, and the step s6 is carried out. As a result, differentiated cells from which undifferentiated cells have been removed remain in the container D1.
- step s6 The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in step s6 can be referenced from the configurations described in steps s1 to s5 above.
- the method for removing undifferentiated cells is not particularly limited as long as it can remove cells other than the cells produced by the production method, and can be performed by adding a known undifferentiated cell removing agent or the like to the medium (for example, Di Mao., et al. Angewandte Chemie International Edition; 9 January 2017, Ben-David, U., et al. Cell Stem Cell, 12, 167 (2013), WO2019/187918, JP2016-93178, Yoshiki Nakashima, et. al., Molecular Therapy Vol. 26 No. 7 July 2018, etc.).
- a known undifferentiated cell removing agent or the like for example, Di Mao., et al. Angewandte Chemie International Edition; 9 January 2017, Ben-David, U., et al. Cell Stem Cell, 12, 167 (2013), WO2019/187918, JP2016-93178, Yoshiki Nakashima, et. al., Molecular Therapy Vol. 26 No
- step s7 is added after step s6 to take out a sample for quality testing of differentiated cells, and a container E1 associated with step s7 is connected in the same manner as described above.
- a container E1 for carrying out step s7 is further connected after the container D1 via a connecting pipe Jd1.
- the container E1 has one or more openable and closable input/output ports.
- the feed mechanism Fd1 moves the contents from the container D1 to the container E1, and the sample required for testing is taken out through the input/output port of the container E1.
- the sample for quality testing may be taken out of the container D1 after step s6.
- step s7 The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in step s7 can be referenced from the configurations described in steps s1 to s6 above.
- the purpose of the quality test is to confirm whether the cells, organoids, etc. produced by the production method (II) of the present invention are the desired ones.
- the test items for the quality test are not particularly limited, but include basic tests such as the morphology of the cells or organoids, the presence or absence of expression of cell surface markers, sterility tests, endotoxin tests, and evaluation of cell viability, and testing equipment suitable for each item can be used.
- step s7 is completed and the cells are certified as non-defective
- the connecting lines and piping connected to the container E1 may be removed, and the container E1 and the differentiated cells (in the form of a cell suspension) therein may be provided (shipped, etc.) as is to a user (such as a patient undergoing cell therapy), or may be transferred to a syringe-type container or vial and provided to the user.
- the container E1 may be provided to the user with a syringe connected to the inlet/outlet port for removing the differentiated cells.
- the container E1 may be provided as a medicine, etc., as described below.
- the differentiated cells or organoids of the present invention can be suitably used in immunotherapy and regenerative medicine, so in another aspect, a pharmaceutical comprising the differentiated cells or organoids of the present invention (hereinafter, sometimes referred to as the "pharmaceutical of the present invention") is provided.
- the pharmaceutical of the present invention is provided, for example, in the form of an immunotherapy agent or a cell transplantation agent.
- the present invention also includes a method for treating a disease in which an effective amount of the differentiated cells or organoids of the present invention is administered or transplanted into a primate to be treated. Specific examples of primates are as described above in "1. Method for producing pluripotent stem cells", but are preferably humans.
- the differentiated cells or organoids of the present invention can be administered or transplanted into the body of a subject in need thereof.
- the transplantation is preferably performed in a region of the body where the cells can be fixed at a certain position, for example, subcutaneously, intraperitoneally, peritoneal epithelium, omentum, adipose tissue, muscle tissue, or under the capsule of each organ such as the pancreas or kidney.
- the transplantation is subcutaneous, which is less invasive.
- the cells to be transplanted may be administered in a therapeutically effective amount, which may vary depending on factors such as the age, weight, size of the transplantation site, and severity of the disease of the transplantation subject, and are not particularly limited, but may be, for example, about 10 x 10 4 cells to 10 x 10 11 cells.
- the differentiated cells or organoids of the present invention are used as medicines, it is desirable to use cells or organoids derived from iPS cells established from somatic cells with the same or substantially the same HLA genotype of the recipient individual, from the viewpoint of preventing rejection reactions.
- substantially the same means that the HLA genotype matches the transplanted cells to such an extent that the immune response can be suppressed with an immunosuppressant, for example, somatic cells with an HLA type that matches the three loci of HLA-A, HLA-B, and HLA-DR, or four loci including HLA-C.
- an immunosuppressant for example, somatic cells with an HLA type that matches the three loci of HLA-A, HLA-B, and HLA-DR, or four loci including HLA-C.
- capsules such as polyethylene glycol or silicone, or in porous containers, to avoid rejection reactions, and then transplanted.
- the differentiated cells or organoids of the present invention are prepared as parenteral preparations such as injections, suspensions, or drops by mixing with a medicamentically acceptable carrier according to conventional methods.
- a method for producing an immunotherapy agent or cell transplantation therapy agent which includes a step of formulating the differentiated cells or organoids of the present invention.
- Such a method may include a step of preparing the differentiated cells or organoids of the present invention.
- it may also include a step of preserving the differentiated cells or organoids of the present invention.
- pharma- ceutically acceptable carriers examples include aqueous solutions for injection, such as physiological saline, isotonic solutions containing glucose and other adjuvants (e.g., D-sorbitol, D-mannitol, sodium chloride, etc.).
- the cells of the present invention may be compounded with, for example, buffers (e.g., phosphate buffer, sodium acetate buffer), soothing agents (e.g., benzalkonium chloride, procaine hydrochloride, etc.), stabilizers (e.g., human serum albumin, polyethylene glycol, etc.), preservatives, antioxidants, etc.
- buffers e.g., phosphate buffer, sodium acetate buffer
- soothing agents e.g., benzalkonium chloride, procaine hydrochloride, etc.
- stabilizers e.g., human serum albumin, polyethylene glycol, etc.
- the immunotherapy agent or cell transplantation therapy agent of the present invention is provided in a frozen state under conditions normally used for cryopreservation of cells, and can be thawed when used.
- it may further contain serum or a substitute thereof, an organic solvent (e.g., DMSO), etc.
- an organic solvent e.g., DMSO
- the concentration of serum or a substitute thereof is not particularly limited, but may be about 1 to about 30% (v/v), preferably about 5 to about 20% (v/v).
- the concentration of the organic solvent is not particularly limited, but may be 0 to about 50% (v/v), preferably about 5 to about 20% (v/v).
- the differentiated cells or organoids of the present invention can also be used in methods for screening candidate drugs that are useful for treating or preventing disease.
- the device comprises: The part that produces iPS cells (hereinafter referred to as the iPS cell production department), A portion for expanding iPS cells (hereinafter also referred to as the expansion culture portion), A part for inducing differentiation of iPS cells to form differentiated cells (hereinafter also referred to as a differentiation induction part), A part for removing undifferentiated cells (hereinafter also referred to as an undifferentiated cell removing part), The part where differentiated cells are examined (hereinafter referred to as the examination part) It can be divided into:
- the containers A1 to A3 have connecting pipes J1 and J2, respectively.
- the containers are connected in series in the order of the steps through the connecting pipes J1 and J2, respectively, which are switched to a communicating state.
- the container has a feed mechanism F1 for moving the contents to the next container.
- Each of the material supply sources G1 to G3 is appropriately selected according to the process to be carried out in the respective container.
- the steps s1 to s3 are carried out in order using the iPS cell production section to produce iPS cells from somatic cells, as described in the production method (I) above.
- step s2 of reducing the concentration of the reprogramming factor in the liquid medium may be divided into multiple processing steps (multiple steps), in which case multiple containers A2 to A(n-1) can be used for step s2.
- step s2 is a single step, and the apparatus has a configuration including a single vessel A2 for carrying out step s2.
- step s2 may be divided into a plurality of processing stages (a plurality of steps), in which case a necessary number of containers A2 to A(n-1) can be added and inserted for step s2, similar to the configuration shown in Fig. 1. Even if further containers are inserted for step s2, or if other containers for additional steps are inserted between the series connection of containers A1 to A3, containers A1 to A3 are still considered to be connected in series.
- the expansion culture section in the device is a section for carrying out step s4 in the above-mentioned production method (I).
- the device further has p containers B1 to Bp as the expansion culture section for carrying out step s4 of expanding iPS cells in a liquid medium p times.
- p is an integer of 1 or more, i.e., p ⁇ 1.
- Each of the containers B1 to Bp has one or more inlet/outlet ports that can be opened or closed.
- connection modes of the containers when the number of times of expansion culture (p times) is one and when it is two or more are as follows.
- Container B1 is connected or connectable to container A3 via a connecting pipe Jn.
- the device has a feed mechanism Fn that moves the contents of container A3 to container B1 through the connecting pipe Jn that is switched to a communicating state.
- the number p of expansion cultures is 2 or more, two or more containers B1 to Bp are used.
- Container B1 is connected to container A3 via a connecting pipeline, or is in a state where it can be connected.
- the sealed containers B1 to Bp are connected in series in the order of step s4 via connecting pipelines Jb1 to Jb(p-1), respectively, or are in a state where they can be connected.
- the device has a feed mechanism Fn and Fb1 to Fb(p-1) that move the contents of container A3 to containers B1 to Bp in order through connecting pipelines Jn, which is switched to a communicating state, and Jb1 to Jb(p-1), respectively.
- the procedure for carrying out step s4 in the expansion culture section is as described in the above manufacturing method (I).
- the iPS cells may be those obtained by carrying out steps s1 to s3, or they may be iPS cells that have already been prepared (e.g., commercially available).
- the differentiation induction section of the device is a section for carrying out step s5 in the above manufacturing method (II).
- the above manufacturing method (II) an example using containers C1 and C2 was specifically shown with reference to Fig. 9, but the differentiation induction section of the device further uses q sealed containers C1 to Cq for carrying out step s5, as shown in Fig. 12.
- q is an integer of 1 or more, that is, q ⁇ 1.
- Each of the containers C1 to Cq has one or more openable/closable inlet/outlet ports.
- the connection modes of the respective containers are as follows. 12, when the q containers are one container C1, the container C1 is connected or connectable to the container A3 (or to the last container Bp among the containers B1 to Bp) via a connecting pipe Jn (or Jbp).
- the device has a feed mechanism Fn (or) Fbn that moves the contents of the container A3 (or the container Bp) to the container C1 through the connecting pipe Jn (or Jbp) that is switched to a communicating state.
- the containers C1 to Cq are connected in series or are connectable through the connecting pipeline in the order of step s5.
- the container C1 is connected or is connectable to the container A3 (or the last container Bp among the containers B1 to Bp) through the connecting pipeline Jn (or Jbp).
- the differentiation induction unit of the device has a feed mechanism Fn (or Fbp) and feed mechanisms Jc1 to Jc(q-1) that move the contents of the container A3 (or the contents of the container Bp) to the containers C1 to Cq in order through the connecting pipeline Jn (or Jbp) switched to the communicating state and through Jc1 to Jc(q-1).
- step s5 in the differentiation induction section of the device is as described in the manufacturing method (II) above.
- the differentiation induction section may be an independent cell manufacturing device, and there may be no iPS cell production section or expansion culture section in the preceding stage.
- this corresponds to the cell manufacturing device of the present invention in which [multiple cell manufacturing steps are divided into multiple containers, and cells are configured to move from container to container through connecting pipes in the order of the steps, whereby the steps associated with each container are performed sequentially].
- the undifferentiated cell removal unit in the device is a part that performs step 6 in the above-mentioned manufacturing method (II).
- the device has a configuration for performing step s6 as the undifferentiated cell removal unit.
- the container D1 undifferentiated cells are removed from the contents of the last container Cq (X2 in FIG. 10) among the containers C1 to Cq in the differentiation induction section (step s5).
- the vessel D1 has one or more openable/closable inlet/outlet ports.
- the vessel D1 is connected to the rearmost vessel Cq (in FIG. 10,
- the undifferentiated cell removal unit of the device transfers the contents of the container Cq to the container D1 through the connecting pipe Jx2 that is switched to the communicating state.
- step s6 in the undifferentiated cell removal section is as described in the manufacturing method (II) above.
- the inspection section of the device is a section that carries out step s7 in the manufacturing method (II).
- the device further has a container E1 as an inspection section.
- the container E1 is a container for taking out a sample for inspecting the differentiated cells obtained by the above step s6.
- the container E1 has one or more openable/closable inlet/outlet ports.
- the container E1 is connected to the container D1 via a connecting pipe Jd1 or is in a state where it can be connected.
- the inspection section of the device has a feed mechanism Fd1 that moves the contents of the container D1 to the container E1 through the connecting pipe Jd1 that has been switched to a communicating state.
- step s7 in the inspection section is as described in the manufacturing method (II) above.
- FIG. 13 is a diagram showing an example of a preferred embodiment of the device.
- the device further has a substrate for arranging containers used in the device.
- containers necessary for carrying out the steps are arranged on the substrate Y10, and each container is fixed to the substrate Y10.
- a total of 10 containers (A1 to A3, B1, B2, C1 to C3, D1, E1) are fixed to the substrate surface in the order of the steps.
- a syringe G1 which is a material supply source for supplying somatic cells, is connected to the input/output port 111 of the container A1, and a syringe for extracting differentiated cells, which are the final product, is connected to the input/output port of the container E1.
- Each container has an input/output port (e.g., the portion indicated by the symbols 111, 112, and 113) that can be opened and closed.
- each container is connected or can be connected in the order of the steps via the above-mentioned connection pipe line (e.g., the portion indicated by the symbol J1) that can be switched between a communication state and a non-communication state.
- the cells move in the direction of the arrows in the figure, and in each vessel, they undergo a process corresponding to that vessel.
- Fig. 13 the mechanism for switching the connecting pipe line between a connected state and a non-connected state, and the mechanism for setting the connecting pipe line to a connectable state are not shown.
- the above-mentioned feeding mechanism for moving the contents of the vessel to the vessel in the next stage through the connecting pipe line switched to the connected state is not shown.
- the containers are arranged on a board, they are easy to handle, even when a large number of containers are used. In addition, because the containers are arranged on the board in the order of the process, confusion among the multiple tube piping is suppressed. Arranging them on a board is also preferable because it is suitable for visually checking the manufacturing process and obtaining position information, and serves as a mark for workers to recognize and identify the work process. Furthermore, adding position information makes it easier to process information for manufacturing process management using electronic data.
- the material of the substrate is not particularly limited, and examples include hard materials such as metals and plastics, and flexible plastic materials. It is preferable for the material to be one that does not generate dust, fine particles, or volatile gases, and that can be wiped clean with alcohol.
- the substrate Y10 can be folded in two with the folding center line Y11 as an axis.
- the outer peripheral shape of the substrate Y10 is linearly symmetrical with respect to the folding center line Y11.
- a predetermined number of the above-mentioned containers in the example of FIG. 13, containers (A1, A2, A3, B1, B2) are arranged so as to be lined up in order along the folding center line Y11 in one direction d1.
- the remaining containers in the example of FIG. 13, containers (C1, C2, C3, D1, E1)) are arranged so as to be lined up in order along the folding center line Y11 in a direction d2 opposite to the direction d1.
- the last container among the containers in one area e1 (in the example of Figure 13, container B2) and the first container among the containers in the other area e2 (in the example of Figure 13, container C1) are connected or can be connected by a connecting pipe Jb2.
- FIG. 14 is a diagram illustrating an example of a case where the substrate is folded in two. The folding may be performed in such a manner that a sharp crease having a V-shaped cross section is formed. However, as shown in Fig. 14, a curved crease having a U-shaped cross section is preferable because the containers located in the two regions e1 and e2 are not excessively close to each other.
- the inlet/outlet port of the container may extend away from the fold as shown in FIG. 14(a), or may extend toward the fold as shown in FIG. 14(b). Furthermore, when the substrate is folded in half, the container may be located in a position sandwiched between the folded substrates. In order to facilitate observation of the inside of the container and access to the container with an external tube without the substrate covering the container, it is preferable that the container be located on the outside of the folded substrate as shown in FIG. 14.
- FIG. 15 is a diagram showing another example of a preferred embodiment of the device.
- the device further has two flexible sheets Y31 and T32 stacked on each other.
- the area that will become all the containers of the device, the area that will become one or more input/output ports, and the area that will become the connecting pipeline are formed at predetermined positions between the two flexible sheets Y31 and T32. That is, the two flexible sheets are joined to each other, leaving these areas as non-jointed areas.
- FIG. 15 only the container A1 and the connecting pipeline J1 are shown for the purpose of explanation. As shown in FIG.
- the two flexible sheets Y31 and T32 form the container A1 and the connecting pipeline J1 between them, and the two flexible sheets Y31 and T32 are joined to each other around the outer periphery of these areas to form one sheet Y30.
- the area where the flexible sheets Y31 and T32 are joined to each other may be only a strip-shaped area adjacent to the outline of the area where a container, pipeline, etc. is to be formed, or it may be the entire area other than the area where the container, pipeline, etc. is to be formed.
- FIG. 15(b) is an end view of FIG. 15(a) cut at the cut surface w1-w1.
- FIG. 15(b) shows the outline of the container A1 seen in the background in broken lines.
- An actuator J1a for opening and closing the connection pipeline is provided on the outer surfaces of the two flexible sheets Y31 and Y32.
- the pressing actuator J1a is, for example, a direct acting press device such as a pinch valve, and is provided so as to penetrate the sheet Y30 in the area to the side of the connection pipeline, thereby making it possible to pinch and press the connection pipeline from the front and back.
- the pressing actuator J1a operates to take a pressing position (a position in which the area to become the connection pipeline is pressed from the outside of the flexible sheets Y31 and T32 to make it non-connected) and a non-pressing position (a position in which the area to become the connection pipeline is not pressed and made connected).
- the pressing actuator J1a is in a non-pressing position, and the connection pipe J1 is in a connected state.
- the region that becomes the connection pipe functions as a connection pipe that can be switched between a connected state and a non-connected state. As shown in FIG.
- the region that becomes one or more input/output ports in each container extends from the region that becomes each container (A1, A2, A3, etc.) to the outer edge of the flexible sheet (substrate Y10 in FIG. 13) to form an open end.
- the open end is provided with a structure (not shown) for an openable input/output port.
- the device can be constructed with a large number of containers in a simple manner. From the standpoint of cost, this type of construction can be disposable after one use.
- the outer peripheral shape of the two overlapping and joined flexible sheets Y31 and Y32 in Fig. 15 is a shape that can be folded in two around the folding center line Y11 as an axis, as shown in Fig. 13.
- the outer peripheral shape of the sheets is symmetrical with respect to the folding center line Y11.
- a predetermined number of regions that will become sealed containers in the example of FIG. 13, containers (A1, A2, A3, B1, B2)) are formed in order along the folding center line Y11 in one direction d3.
- the one or more inlet/outlet port regions (111, 112, 113) extend in a direction away from the folding center line Y11.
- the inlet/outlet port regions (111, 112, 113) extend to the outer periphery of the joined flexible sheets Y31 and Y32 to form open ends.
- a region that will become a connecting pipe that connects the regions that will become containers is formed.
- regions which will become the remaining containers are formed in order along the folding center line Y11 in a direction d4 opposite to the one direction d3. Furthermore, from the outer periphery of each of the regions which will become the remaining containers, which is located farther from the folding center line Y11, regions which will become the one or more input/output ports extend in a direction away from the folding center line Y11 and extend to the outer periphery of the two flexible sheets to form an open end.
- regions which will become connecting pipelines connecting the regions which will become the containers are formed.
- the rearmost sealed container among the containers in the one region e3 (in the example of FIG. 13, container B2) and the first container among the containers in the other region e4 (in the example of FIG. 13, container C1) are connected by a region that becomes the connecting pipe Jb2 that crosses the folding center line.
- this type of foldable configuration is preferable because the inlet/outlet ports of each container face in the same direction and are close to each other, making the length of each tube connected to the container the shortest and all of the tubes the same (or similar) length.
- FIG. 16 is a diagram showing an example of a preferred embodiment for arranging and fixing a plurality of containers on a substrate
- FIG. 17 is a diagram showing an embodiment of the substrate shown in FIG. 16 and its use state.
- a pocket Y20 capable of accommodating a container is provided at a position where the container is to be arranged on the main surface of a substrate Y10 formed from a bendable flexible sheet.
- the substrate Y10 is used so that its main surface is a vertical surface, and five pockets are arranged vertically in each of the regions e1 and e2 on both sides of the folding center line Y11.
- Each pocket is configured to a size that can appropriately accommodate a container.
- Containers A1, A2, A3, B1, and B2 are inserted into the five pockets of the region e1 in order from top to bottom, and containers (C1, C2, C3, D1, and E1) are inserted into the five pockets of the region e2 in order from bottom to top.
- This arrangement allows the ten containers to be compactly arranged in two rows (containers A1, A2, A3, B1, B2, and containers C1, C2, C3, D1, E1), with containers B2 and C1 close to each other and easy to connect to each other, just like connecting other containers to each other.
- Providing a pocket on the substrate surface for inserting the container is preferable because it allows the container to be quickly attached to and detached from the substrate. Also, as shown in Figure 17, by arranging the container vertically, a compact device that does not occupy a large space can be obtained.
- FIG. 18(a) is a diagram showing another preferred embodiment for arranging and fixing a plurality of containers on a substrate.
- FIG. 18(b) is a diagram showing an embodiment of the substrate shown in FIG. 18(a) and its use state.
- a pocket Y20 capable of accommodating a container is provided on a substrate Y10 formed of a flexible sheet.
- the openings of the five pockets provided in each of the regions e1 and e2 on both sides of the folding center line Y11 all face the direction of the folding center line Y11.
- the double-headed arrows indicate the direction in which a container is inserted and removed from each pocket.
- Containers A1, A2, A3, B1, and B2 are inserted into the five pockets of the region e1 in order from right to left in the figure, and containers (C1, C2, C3, D1, and E1) are inserted into the five pockets of the region e2 in order from left to top in the figure.
- the inlet/outlet ports of each container face in the same direction (upward in FIG. 14) and approach each other, as shown in FIG. 18(b), which is preferable.
- the opening and closing of the container's inlet and outlet ports, starting and stopping the supply of material from an external material supply source, opening and closing of the connecting pipelines, starting and stopping the feed mechanism, temperature control, time control, etc. in the device can be performed manually, automatically by a control device (a computer that executes a control program, a sequence circuit, etc.), or semi-automatically by combining these.
- a control device a computer that executes a control program, a sequence circuit, etc.
- This cell manufacturing method is a method for producing iPS cells using the cell manufacturing apparatus of the present invention, and as described in the above manufacturing method (I), steps s1 to s3 are carried out in each of containers A1 to A3, and iPS cells are obtained in container A3.
- the manufacturing method includes at least the following steps s1 to s3.
- Step s1 of contacting somatic cells with reprogramming factors in a liquid medium in a container A1 of the cell manufacturing apparatus (i) after completion of step s1, a step s2 is performed in which the contents of container A1 are transferred into container A2 through connecting pipe J1 that has been switched to a communicating state, and the concentration of the reprogramming factor in the liquid medium is reduced in container A2.
- the manufacturing method may include a step of manufacturing differentiated cells from iPS cells, a step of removing undifferentiated cells from the suspension containing the differentiated cells, and a step of inspecting the obtained differentiated cells.
- Example 1 (Production of iPS cells from human whole blood)
- human whole blood was centrifuged to obtain peripheral blood mononuclear cells (PBMCs), and iPS cells were produced from the PBMCs.
- PBMCs peripheral blood mononuclear cells
- the cell production device according to the present invention a self-made sealed container shown in FIG. 2 was used, and the containers were arranged in series in a bottle rack (similar to a test tube stand) as shown in FIG. 19, so that the cell production device was configured so that the containers could be connected in series.
- the volume of each container was 20 ml.
- Each container had a first inlet/outlet port for gas, a general-purpose second inlet/outlet port, and a general-purpose third inlet/outlet port, and the general-purpose inlet/outlet port was used as a material supply line and as a connection line.
- Step s1 A step of contacting PBMCs with reprogramming factors in a liquid medium in container A1.
- Step s2 A step of reducing the concentration of the reprogramming factor in the liquid medium in container A2.
- Step s3 A step of culturing in container A3 for 14 days to establish iPS cells.
- Step s4-1 A step of performing a first expansion culture in container B1.
- Step s4-2 A step of performing a second expansion culture in the container B2. (Step s4-2 is the same as step s4-1 in terms of the operation.)
- a Chemoclave (registered trademark) bag spike which is a connector for injecting and suctioning a drug solution, was connected to the outlet of the blood collection bag.
- a 5 ml syringe was connected to the bag spike, and 4 ml of blood was sucked into the syringe.
- the syringe was removed from the bag spike, and the bag spike was closed with a cap (Combi-Stopper).
- An injection needle (23G) was attached to a syringe containing 4 ml of human whole blood in a safety cabinet.
- the injection needle was pierced into the rubber stopper of a BD Vacutainer CPT, and the human whole blood was drawn and injected by negative pressure.
- the BD Vacutainer CPT was placed in a centrifuge and PBMCs were separated by centrifugation (1500-1800 G, 15 minutes (heparin), 20 minutes (citric acid)).
- Step s1 contacting PBMC with reprogramming factors in container A1
- Step s1 Injection of PBMCs into container A1
- the second inlet/outlet port of container A1 was connected to the third inlet/outlet port of container A2 in a safety cabinet via a lock connector (TS-LC11 manufactured by Terumo Corporation). In actual production, the connection may be made in advance.
- the gas inlet and outlet ports of vessel A1 and vessel A2 were closed.
- a syringe for a suction pump was connected to the second input/output port of container A2
- a BD Vacutainer (registered trademark) Luer lock access device (hereinafter referred to as the Luer lock access device) was connected to the third input/output port of container A1, and the above-mentioned BD Vacutainer CPT containing a liquid containing centrifuged PBMCs was connected to the Luer lock access device.
- Air was sucked out from inside the container A2 using a syringe for a suction pump connected to the second inlet/outlet port of the container A2.
- the PBMCs in the BD Vacutainer CPT were made to flow into the syringe, and then the PBMCs in the syringe were made to flow into the container A1.
- the entire apparatus including container A1, was incubated for 2 hours in a safety cabinet equipped with a hot plate at 37°C.
- Step s2 Reducing the concentration of reprogramming factors in the liquid medium
- a lock connector Teumo Corporation, TS-LC11
- the second input/output port of container A1 and the third input/output port of container A2 were connected with a connecting pipe inside a safety cabinet (in actual production, the connection was already made in advance).
- container A1 and container A2 each contained half the amount of cells (PBMCs) compared to the original vial, and reprogramming vectors diluted 200 times compared to the original vial.
- Step s3 Establishment of iPS cells by culturing for 14 days
- 5 mL of atelocollagen bead solution (KOKEN: MIC-00) was injected from the third inlet/outlet port of container A3 with a syringe in a safety cabinet.
- the syringe was removed, and the second inlet/outlet port of container A2 and the third inlet/outlet port of container A3 were connected in the safety cabinet using a lock connector (in actual production, they were connected in advance).
- the first inlet/outlet port (for gas) of vessel A2 was closed, and the lock connector connected to the third inlet/outlet port of vessel A2 was removed in a safety cabinet.
- the first inlet/outlet port was confirmed to be closed, and the opening of the connected lock connector for the second inlet/outlet port of the container A1 was closed using a fluid dispensing connector.
- the third inlet/outlet port of the container A1 was also closed using the cap of the fluid dispensing connector.
- a syringe containing 10 mL or more of liquid medium (StemFit AK03) is connected to the third inlet/outlet port of container A2, and 10 mL of the liquid medium is injected.
- the amount of liquid medium in container A3 becomes 10 mL.
- container A3 contains 1/4 the amount of PBMC compared to the original vial, and initialization vector diluted 400 times compared to the original vial.
- the lock connector connected to the third inlet/outlet port of container A3 was removed inside the safety cabinet.
- the third inlet/outlet port of container A3 was also closed using the cap of a fluid dispensing connector (BRAUN: 415080).
- BRAUN fluid dispensing connector
- the third inlet/outlet port of container A2 was also closed using the cap of the fluid dispensing connector.
- the entire device was incubated in a safety cabinet equipped with a hot plate at 37°C for 14 days. This resulted in the establishment of iPS cells.
- the third inlet/outlet port of container A3 When liquid medium is to be continuously supplied, the third inlet/outlet port of container A3 is not closed, and a syringe containing liquid medium (StemFit AK03) is connected to the third inlet/outlet port of container A3 via an extension tube (Seaman CTL1001S2) to continuously supply the liquid medium.
- a syringe or a waste liquid bag may be connected via an extension tube without closing the third inlet/outlet port of container A3.
- the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.).
- a hygroscopic material e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.
- an absorbent article containing the material e.g., water-absorbent pad, water-absorbent sheet, etc.
- liquid medium was continuously supplied (0.1 ml/h).
- a Coudec Syringe Pump (Daiken Medical Co., Ltd., CSP-120) was used as the automatic liquid delivery pump, and a Pump Uniter Stand (Daiken Medical Co., Ltd., PUS-200S) and a Pump Uniter (Daiken Medical Co., Ltd., PU3-200S) were used to secure the syringe pump.
- Step s4 Expansion culture of iPS cells
- the syringe (Terumo Corporation, 50 mL) was removed to open the first inlet/outlet port, which was a gas port of the container A3, and then the waste liquid syringe connected to the second inlet/outlet port of the container A3 was pulled to release the positive pressure of the container A3.
- a lock connector (Terumo, TS-LC11), a three-way stopcock (Terumo, Telfusion), and a sampling syringe (Terumo, 2.5 ml) were used.
- the three-way stopcock was connected to the lock connector, and then to the third inlet/outlet port of the first expansion culture container B1.
- the sampling syringe was connected to the side branch of the three-way stopcock.
- the three-way stopcock in the direction of the third inlet/outlet port of the container B1 was closed, and then the clip in the first inlet/outlet port of the container A3 was closed.
- a three-way stopcock (Terumo Corporation, Terufusion) connected to the third inlet/outlet port of container B1 was turned on in all directions, and all of the liquid medium contained in a syringe (Terumo Corporation, 50 mL) connected to the third inlet/outlet port of container A3 was pushed out. As a result, the contents of container A3 were transferred to container B1.
- the sampling syringe connected to the three-way stopcock was pulled to sample the cell suspension.
- 1/10 of the total volume (1.5 ml) of the contents of container A3 containing the established iPS cells was sampled.
- the three-way stopcock was operated to turn the direction of the sampling syringe OFF.
- the extension tube (Seaman, CTL1001S2) with the attached syringe (Terumo, 50 mL) connected to the third input/output port of container A3 was removed inside the safety cabinet.
- the third input/output port of container A3 was closed using the cap of the fluid dispensing connector.
- the extension tube connected to the waste liquid syringe connected to the second input/output port of container A3 was removed inside the safety cabinet.
- the second input/output port of container A3 was closed using the cap of the fluid dispensing connector. It was confirmed that the gas port (first input/output port) of the used container A3 was closed.
- the three-way stopcock (Terumo, Terufusion) connected to the third inlet/outlet port of container B1 was removed inside the safety cabinet.
- a syringe (Terumo, 50 ml) containing liquid medium (StemFit AK03) was connected to the third inlet/outlet port of container B1 via an extension tube, and the liquid medium was continuously supplied.
- a syringe (Terumo, 50 ml) was connected to the third inlet/outlet port of container B1 via an extension tube (a waste liquid bag may be connected, and the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., absorbent pad, absorbent sheet, etc.)).
- a hygroscopic material e.g., absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.
- an absorbent article containing the material e.g., absorbent pad, absorbent sheet, etc.
- the entire device was placed in a safety cabinet equipped with a 37°C hot plate, and expansion culture was carried out for 7 days.
- the number of iPS cell colonies contained in 10 ml of the 15 ml medium volume was 1 to 2. From this result, it is considered that the number of iPS cell colonies established using PBMCs (approximately 1.5 x 107 cells) collected from 4 ml of whole blood is 1 to 2.
- Example 2 (Production of iPS cells from commercially available PBMCs)
- iPS cells were obtained by carrying out steps s1 to s4 in the same manner as in Example 1 above, except that a commercially available PBMC was used (i.e., the centrifugation step of whole blood was omitted).
- Step s1 Contacting somatic cells (PBMCs) with reprogramming factors
- PBMCs peripheral blood mononuclear cells
- vectors which were also filled in vials, were also collected with a syringe. In both cases, in the production of clinical cells, the vectors are provided sealed in containers equipped with connectors that can be connected aseptically.
- SRV iPS-2 Vector 4.6 ⁇ 10 7 CIU/mL
- PBMC Human PBMC 10M (PRECISION 93210-10M)
- the entire apparatus including container A1, was incubated for 2 hours in a safety cabinet equipped with a 37°C hot plate to complete step s1.
- Step s2 (reducing the concentration of reprogramming factors in vessel A2), step s3 (establishment of iPS cells in vessel A3), and step s4 (expansion culture in vessel B1) were carried out under the same conditions and with the same procedures as in Example 1 above.
- step s3 (Evaluation of iPS cells at the completion of step s3 (establishment)) After 14 days of culture in step s3 (after establishment of iPS cells), a 1.5 ml sample of cell suspension was obtained from container A3. The sample was subjected to live staining using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope together with GFP, a fluorescent protein from the SRV TM iPSC-2 Vector. Figure 27 shows the number of TRA-1-60 positive + GFP positive colonies (iPS cell colonies).
- Example 3 (Production of iPS cells from human whole blood (1))
- human whole blood was centrifuged to obtain PBMCs, and iPS cells were produced from the PBMCs in the same manner as in Example 1, except that a GREX 10M-CS (manufactured by Wilson Wolf Corporation) was used as the sealed container constituting the cell production apparatus according to the present invention.
- GREX 10M-CS manufactured by Wilson Wolf Corporation
- the GREX 10M-CS is a generally cylindrical sealed container having a container body and a lid (multi-port cap).
- the bottom surface of the container body is made of a gas permeable membrane.
- the volume of the container is 100 ml.
- the containers were connected in series and steps s1 to s4 were carried out.
- the GREX 10M-CS multi-port cap has the following input and output ports: Sample Line Tubing (a MicroClave (registered trademark) Connector is provided at the tip of the Sample Line Tubing). Hereinafter, this will also be referred to as the sample port.
- Reduction Line Tubing The Weldable Reduction Line branches off from the Reduction Line Tubing.
- Harvest Line Tubing Hereinafter referred to as the harvest port.
- the weldable harvest line branches off from the harvest port.
- Gas inlet/outlet port to which a Pall Versapor Vent Filter is connected hereinafter also referred to as the gas port.
- Step s1 contacting PBMC with reprogramming factors in container A1
- the acquisition of PBMCs from human whole blood was carried out in the same manner as in Example 1 above.
- a BD Vacutainer CPT for storing PBMCs as a fraction after centrifugation a container GREX 10M-CS, a Luer lock access device, a syringe (Terumo Corporation, 50 ml), an injection needle (Terumo Corporation, 23G x 1), and a three-way stopcock type R (Terumo Corporation, Terufusion) were prepared.
- a three-way stopcock type R and a Luer lock access device were connected to the MicroClave Connector attached to the sample port of container A1.
- the vectors were extracted from the vials using a syringe.
- the initialization vector is provided sealed in a container that can be connected via a sterile connector. Approximately 25 ml of the syringe is drawn up in advance. An injection needle was attached to the syringe, and 0.1 ml (total amount) of SRV iPS-2 Vector (4.6 x 10 7 CIU/ml 0.1 ml) was extracted.
- the injection needle was removed from the syringe and the syringe was connected to the side flow path of the R-type three-way stopcock.
- the three-way stopcock was turned off in the flow path toward the MicroClave Connector of container A1.
- the BD Vacutainer CPT which had been centrifuged to remove PBMCs, was set in the Luer lock access device.
- the BD Vacutainer CPT was pushed into the Luer lock access device.
- the stopcock of the three-way stopcock was turned off in the flow path toward the Luer lock access device.
- the syringe containing the serum and PBMCs was rotated 90 degrees to a vertical position, and the syringe was pushed in while the content liquid was held at the bottom of the syringe, causing the cell suspension to flow into container A1.
- the BD Vacutainer CPT was removed from the Luer lock access device. Incubated in a CO2 incubator for 2 hours.
- Step s2 Reducing the concentration of reprogramming factors in the liquid medium in container A2
- Only the Reduction Line Tubing of the vessel A1 was opened, and the gas port and the harvest port were closed.
- a syringe containing 50 ml of StemFit AK03 liquid medium was connected to the Reduction Line Tubing of the container A1, and 50 ml of the liquid medium was injected.
- the Reduction Line Tubing of vessel A1 was closed.
- the cells (PBMC) were allowed to stand in a CO 2 incubator for 30 minutes until they naturally settled in container A2.
- container A2 contained PBMCs in an amount equal to 1/1 of the amount in the original vial, and the initialization vector diluted 500-fold compared to the amount in the original vial.
- a waste liquid bag may be used, and the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.).
- a waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.).
- a hygroscopic material e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, poly
- a syringe (50 ml) with a retracted plunger was connected via the fluid dispensing connector to inject 50 ml of air into the opening of the gas port of container A2.
- the tube of the gas port of vessel A2 was opened. Air was forced into container A2 from a syringe connected to the gas port of container A2.
- the supernatant of the liquid medium in container A2 (the liquid present in the region from the top end of the container body to 8 cm) was collected as waste liquid into a waste liquid collecting syringe (50 ml) connected to the weldable reduction line of container A2.
- container A2 the liquid medium containing PBMCs remained at a height of about 2 cm from the bottom.
- the gas port tube of vessel A2 and the weldable reduction line tube were closed.
- container A2 contained PBMCs in an amount equal to 1/1 of the amount in the original vial, and the initialization vector (relative to the original vial) diluted 500-fold compared to the original vial.
- Step s3 Establishment of iPS cells by culturing for 14 days
- the Weldable Harvest Line of the container A2 is connected to the MicroClave Connector of the container A3.
- the MicroClave Connector can be connected only once in a general environment (it cannot be reconnected).
- 10 ml of an atelocollagen bead solution (KOKEN: MIC-00) contained in a syringe was injected from the harvesting port of container A2 in a safety cabinet.
- a syringe containing 50 ml of the liquid medium StemFit AK03 was connected to the Reduction Line Tubing of the container A2, and 50 ml of the liquid medium was injected.
- the liquid content of the container A2 flowed into the container A3 together with the injected 50 ml liquid medium. After that, the Reduction Line Tubing of the container A2 was closed. The device was placed in a CO2 incubator without disconnecting the piping, and a 14-day culture was started.
- container A3 contained 1/1 the amount of PBMCs compared to the original vial and initialization vector diluted 3000 times compared to the original vial.
- the liquid medium was injected from a syringe via an extension tube (Seaman CTL1001S2).
- a Coudec Syringe Pump CSP-120 (Daiken Medical Co., Ltd. CSP-120) was used as the syringe pump, and a Pump Uniter Stand (Daiken Medical Co., Ltd.: PUS-200S) and a Pump Uniter (Daiken Medical Co., Ltd.: PU3-200S) were used to secure the syringe pump equipment.
- Step s4 Expansion culture of iPS cells
- a syringe JMS, 100 ml
- the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.).
- a hygroscopic material e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.
- an absorbent article containing the material e.g., water-absorbent pad, water-absorbent sheet, etc.
- a fluid dispensing connector was connected to the gas port of the container A3, and a syringe filled with air (Terumo, 50 ml) was connected.
- the clip of the Weldable Harvest Line of the container A2 connected to the MicroClave Connector of the container A3 was closed. Air was sent from the syringe, and the supernatant medium in the container A3 was discharged into the syringe for waste liquid. This operation was repeated twice, and the syringe connected to the fluid dispensing connector was removed and filled with air again. All of the supernatant in the container A3 was discharged into the syringe for waste liquid. After that, the clip of the gas port of the container A3 was closed to prevent backflow. In addition, the Weldable Reduction Line of the container A3 was closed with a clip to prevent backflow of the waste liquid.
- the Weldable Harvest Line of container A3 was connected to the MicroClave Connector of container B1 via a three-way stopcock (Terufusion TERUMO).
- a syringe (Terumo, 2.5 ml) for obtaining samples was connected to the three-way stopcock.
- the MicroClave Connector can be connected once even in a general environment (it cannot be reconnected).
- a cell suspension prepared by first adding 50 ml of liquid medium (StemFit AK03) is transferred from container A3 to container B1.
- the clip on the gas port of container B1 was released.
- 50 ml of liquid medium was added to container A3 through the harvesting port.
- the clip on the harvesting port of container A3 was closed.
- the MicroClave Connector of container B1 was turned ON in all directions. A fluid dispensing connector was connected to the gas port of container A3, and an air-filled syringe (Terumo, 50 ml) was connected. Air was introduced into container A3 through the gas port using the syringe.
- the device was stored in a CO 2 incubator and cultured for 7 days.
- Example 4 (Production of iPS cells from commercially available PBMCs)
- iPS cells were obtained by carrying out steps s1 to s4 in the same manner as in Example 3 above, except that a commercially available PBMC was used (i.e., the centrifugation step of whole blood was omitted).
- Example 5 Provide and evaluation of cardiomyocytes from iPS cells (step s5 of inducing differentiation of iPS cells)
- the cell production device according to the present invention was used to carry out the method for producing differentiated cells according to the present invention, and the differentiation of iPS cells was actually induced to produce cardiomyocytes.
- iPS cells obtained by the production method of the present invention are used.
- the method for producing differentiated cells according to the present invention was carried out using iPS cells provided by the iPS Cell Research Foundation, Kyoto University (a research strain of human clinical iPS cells (15M66)).
- step s5 of inducing differentiation of iPS cells is further divided into three steps (s5-1, s5-2, s5-3), and each of these steps is assigned one-to-one to three sealed containers (C1, C2, C3) as described below. Each step is carried out sequentially in each container while the contents are transferred from container to container.
- Step s5-1 A step of inducing differentiation of iPS cells in the container C1 to obtain cardiac mesoderm via mesoderm.
- Step s5-2 A step of transferring the cardiac mesoderm to a container C2, and inducing differentiation of the cardiac mesoderm in the container C2 to obtain cardiac progenitor cells.
- Step s5-3 a step of transferring myocardial precursor cells to a container C3, and inducing differentiation of the myocardial precursor cells in the container C3 to obtain cardiomyocytes.
- the three sealed containers were made of GREX 10M-CS, which was the same as the containers used in Examples 3 and 4 above.
- the external appearance of the sealed container GREX 10M-CS is as shown in Fig. 29.
- Step s5-1 induction of differentiation of iPS cells into cardiac mesoderm
- an iPS cell line 15M66, 1 x 10 6 cells
- an atelocollagen bead solution (KOKEN: MIC-00)
- 10 mL of liquid medium StemFit AK03 (Ajinomoto) (containing 10 ⁇ M concentration of Y-27632 (Fujifilm Wako))
- 20 ml of medium A from a PSC Cardiomyocyte Differentiation Kit (Thermo Fisher Scientific) was poured into the container C1.
- the medium A was continuously supplied at a flow rate of 0.5 ml/h and cultured for 2 days while discharging the medium from the vessel, thereby inducing differentiation of the iPS cells into mesoderm and then cardiac mesoderm.
- Step s5-2 induction of differentiation from cardiac mesoderm to cardiac progenitor cells
- 20 ml of medium B from the PSC Cardiomyocyte Differentiation Kit was poured into the container C2.
- the B medium was continuously supplied at a flow rate of 0.5 ml/h and cultured for 2 days while discharging the medium from the vessel, thereby inducing differentiation of cardiac mesoderm to obtain cardiac progenitor cells.
- Step s5-3 Induction of Differentiation from Cardiac Muscle Progenitor Cells to Cardiac Muscles
- 20 ml of C medium from the PSC Cardiomyocyte Differentiation Kit (Thermo Fisher Scientific) was poured into the container C3.
- the C medium was continuously supplied at a flow rate of 0.5 ml/h, and the medium was discharged from the vessel while the vessel was cultured for 10 days, thereby inducing differentiation of the cardiac muscle precursor cells to obtain cardiac muscle cells.
- step s5-3 the contents of the container (step s5-3) were sampled and treated with collagenase to dissolve the atelocollagen beads. From the obtained cell pellet, mRNA was extracted using SuperPrep II Cell Lysis & RT Kit for qPCR (Toyobo Co., Ltd.: SCQ-401), and cDNA was synthesized.
- Example 6 Provide and evaluation of pancreatic progenitor cells from iPS cells (step s5 of inducing differentiation of iPS cells))
- the cell manufacturing device according to the present invention was used to carry out the method for manufacturing differentiated cells according to the present invention, and pancreatic progenitor cells were actually produced by inducing differentiation of iPS cells.
- the method for manufacturing differentiated cells according to the present invention as a preferred embodiment, iPS cells obtained by the manufacturing method of the present invention are used, but in this example, the method for manufacturing differentiated cells according to the present invention (step s5 above) was carried out using iPS cells provided by the Kyoto University iPS Cell Research Foundation (a research strain of human clinical iPS cells (15M66)).
- step s5 of inducing differentiation of iPS cells was further divided into three steps (s5-1, s5-2, s5-3), and each of these steps was assigned one-to-one to three sealed containers (C1, C2, C3) as described below, and each step was carried out sequentially in each container while the contents were transferred from container to container.
- Step s5-1 A step of inducing differentiation of iPS cells in the container C1 to obtain a primitive gut tube through definitive endoderm.
- Step s5-2 A step of transferring the primitive gut to a container C2, and inducing differentiation of the primitive gut in the container C2 to obtain posterior foregut endoderm.
- Step s5-3 a step of transferring the posterior foregut endoderm to a container C3, and inducing differentiation of the posterior foregut endoderm in the container C3 to obtain pancreatic progenitor cells.
- the three sealed containers were made of GREX 10M-CS, which was the same as the containers used in Examples 3 and 4 above.
- the external appearance of the sealed container GREX 10M-CS is as shown in Fig. 29.
- Step s5-1 Induction of differentiation of iPS cells into definitive endoderm
- an iPS cell line 15M66, 1 ⁇ 10 6 cells
- an atelocollagen bead solution (KOKEN: MIC-00)
- 10 mL of liquid medium StemFit AK03 (Ajinomoto) (containing 10 ⁇ M concentration of Y-27632 (Fujifilm Wako)) were poured into container C1.
- the procedure thereafter followed the recommended protocol for the TEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) (https://cdn.stemcell.com/media/files/pis/DX20464-PIS_1_4_0.pdf).
- Step s5-2 Induction of Differentiation from Definitive Endoderm to Primitive Gut
- 10 ml of Medium 2A from the STEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) was poured into the container C2.
- Medium 2B was continuously supplied at a flow rate of 0.5 ml/h, and the cells were cultured until Day 9. This induced differentiation of the definitive endoderm to obtain a primitive gut.
- Step s5-3 Differentiation induction from primitive gut to posterior foregut endoderm
- 10 ml of Medium 3 from STEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) was poured into the container C3. From Day 10, Medium 3 was continuously supplied at a flow rate of 0.5 ml/h, and the cells were cultured until Day 13. This resulted in differentiation induction of the primitive gut to posterior foregut endoderm.
- Step s5-4 Differentiation induction from posterior foregut endoderm to pancreatic progenitor cells
- 10 ml of Medium 4 from STEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) was injected into the container C3. From Day 10, Medium 4 was continuously supplied at a flow rate of 0.5 ml/h, and the cells were cultured until Day 19. This resulted in differentiation induction of the posterior foregut endoderm, and pancreatic progenitor cells were obtained.
- pancreatic progenitor cell marker positive cells On Day 19, the contents of the container (step s5-4) were sampled, and immunostaining was performed for pancreatic progenitor cell marker proteins PDX-1 (pancreatic progenitor cell marker) and NKX6.1 (pancreatic progenitor cell marker) according to a standard method.
- the results of double staining with anti-PDX-1 antibody Human/Mouse PDX-1/IPF1 Alexa Fluor 647 MAb (Clone 267712) (R&D Systems, Inc. IC2419R-100UG)
- Hoechst staining Hoechst staining
- step s5-4 the contents of the container (step s5-4) were sampled and treated with collagenase to dissolve the atelocollagen beads. From the obtained cell pellet, mRNA was extracted using SuperPrep II Cell Lysis & RT Kit for qPCR (Toyobo Co., Ltd.: SCQ-401), and cDNA was synthesized.
- the method for producing iPS cells, the method for producing differentiated cells, and the cell production device of the present invention make it possible to produce iPS cells and differentiated cells more cheaply and easily than before, and also enable automated production.
- the present invention is preferably applicable to applications such as establishing iPS cells from the somatic cells of a patient requiring transplantation therapy, producing various differentiated cells from the iPS cells, and transplanting them into the patient (autotransplantation).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Cell Biology (AREA)
- Virology (AREA)
- Sustainable Development (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
本発明は、人工多能性幹細胞の製造方法、および分化細胞の製造方法に関する。また、本発明は、前記製造方法を実施するための細胞製造装置、および該細胞製造装置を用いた細胞製造方法に関する。 The present invention relates to a method for producing induced pluripotent stem cells and a method for producing differentiated cells. The present invention also relates to a cell production device for carrying out the production method, and a cell production method using the cell production device.
近年、人工多能性幹細胞(induced pluripotent stem cell:iPS細胞)由来の分化細胞を用いた再生医療の研究がさかんに行われている。とりわけ、患者の体細胞(例えば、末梢血単核球など)からiPS細胞を樹立し、さらに、該iPS細胞から分化誘導される種々の分化細胞やオルガノイドを該患者に移植(自家移植)するような治療法は、拒絶反応のリスクを低減し得る治療法として注目されている(特許文献1、非特許文献1、2)。
In recent years, research into regenerative medicine using differentiated cells derived from induced pluripotent stem cells (iPS cells) has been actively conducted. In particular, a treatment in which iPS cells are established from a patient's somatic cells (e.g., peripheral blood mononuclear cells) and various differentiated cells or organoids induced to differentiate from the iPS cells are then transplanted (autotransplant) into the patient has attracted attention as a treatment that can reduce the risk of rejection (
自家移植を企図した場合、生体(例えば、ヒト)から得た細胞(体細胞)からiPS細胞を得るには、液体培地中で体細胞に初期化因子を接触させ、該液体培地中の初期化因子の濃度を低減させ、さらに培養を行ってiPS細胞を樹立するといった、長期間にわたる多段の工程が必要である。また、樹立したiPS細胞から種々の分化細胞を得るためにも、所望する細胞への分化誘導や細胞の品質検査といった、長期間にわたる多段の工程が必要である。 When autologous transplantation is intended, obtaining iPS cells from cells (somatic cells) obtained from a living organism (e.g., a human) requires a long-term, multi-step process, such as contacting the somatic cells with reprogramming factors in a liquid medium, reducing the concentration of the reprogramming factors in the liquid medium, and then culturing the cells to establish iPS cells. Furthermore, obtaining various differentiated cells from the established iPS cells also requires a long-term, multi-step process, such as inducing differentiation into the desired cells and testing the quality of the cells.
前記のような多段かつ長時間を要する工程および製造条件の調節を行なうために、従来では、全ての工程を自動的に順番に実施し得るように構成された細胞製造装置がメーカーより提案される様になり、一部の臨床用細胞においても製造の報告例がある。そのような細胞製造装置の共通の特徴は、図42(a)に一例を示すように、単一の培養容器X10内へと、多数の材料供給用バッグX20から、工程に必要な材料が順番に供給されて、該単一の培養容器X10内において全ての工程が順番に実施されるという点にある。多数の材料供給用バッグX20からは、それぞれに軟質チューブ(以下、「チューブ」ともいう)X30が延びており、各チューブX30は、開閉制御可能なピンチバルブX40を通過したのち、合流を繰り返して1つのチューブとなり、それがペリスタポンプX50を通過して、培養容器X10に接続されている。ピンチバルブX40は、制御部(図示せず)の命令に応じて動作する電磁バルブである。該ピンチバルブがチューブを外側から押し潰すように押圧すると、該チューブ内の流路が閉鎖される。ピンチバルブとペリスタポンプがコンピュータープログラムによって制御されて、実施すべき各工程に必要な材料が培養容器X10内に順次送り込まれ、各工程が順次実施される。また、廃液のための流路についても、ペリスタポンプを流れた廃液(古い培養液など)が、チューブを通り、ピンチバルブを通過したのち、廃液バッグへと排出されるように構成されている。廃液は液体として廃棄されても良いが、廃液バッグ内に吸湿性の素材(例えば、吸水性樹脂等、より具体的には、ポリアクリル酸、ポリアクリル酸ナトリウム、ポリアクリル酸共重合体等)や、該素材を含む吸収性物品(例えば、吸水パッド、吸水シート等)を封入した吸湿性の廃液バッグを使うことで液体成分を分別することなく焼却処分が可能となり廃棄処理が容易となる。これらの構成により、該単一の培養容器X10内において、全ての工程が実施される。 In order to adjust the above-mentioned multi-stage and time-consuming processes and manufacturing conditions, manufacturers have proposed cell manufacturing devices that are configured to automatically perform all processes in sequence, and there have been reports of the production of some clinical cells. A common feature of such cell manufacturing devices is that, as shown in FIG. 42(a), materials required for the processes are supplied in sequence from a number of material supply bags X20 into a single culture vessel X10, and all processes are performed in sequence within the single culture vessel X10. A soft tube (hereinafter also referred to as "tube") X30 extends from each of the many material supply bags X20, and each tube X30 passes through a pinch valve X40 that can be opened and closed, and then repeatedly merges to become one tube, which passes through a peristaltic pump X50 and is connected to the culture vessel X10. The pinch valve X40 is an electromagnetic valve that operates according to commands from a control unit (not shown). When the pinch valve presses the tube from the outside so as to crush it, the flow path in the tube is closed. The pinch valve and the peristaltic pump are controlled by a computer program, and the materials required for each process to be performed are sequentially sent into the culture vessel X10, and each process is performed sequentially. The flow path for the waste liquid is also configured so that the waste liquid (such as old culture liquid) that flows through the peristaltic pump passes through the tube and the pinch valve, and then is discharged into a waste liquid bag. The waste liquid may be disposed of as a liquid, but by using a hygroscopic waste liquid bag in which a hygroscopic material (e.g., water-absorbing resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbing pad, water-absorbing sheet, etc.) is enclosed in the waste liquid bag, the liquid components can be incinerated without separation, making disposal easier. With these configurations, all processes are performed in the single culture vessel X10.
前記のような従来の細胞製造装置において、多数のチューブが1つのチューブへと合流し、単一の密閉容器X10に接続される様子は、図42(b)に模式的に例示するとおりである。図42(b)のとおり、密閉容器X10に近い流路ほど、多数の材料供給用バッグにとって共用の流路となっている。図42(b)では、ピンチバルブ、蠕動ポンプ、および廃液用の流路の図示を省略している。 In the conventional cell manufacturing device described above, the manner in which multiple tubes merge into one tube and are connected to a single sealed container X10 is as shown diagrammatically in FIG. 42(b). As shown in FIG. 42(b), the closer the flow path is to the sealed container X10, the more the flow path is shared by multiple material supply bags. In FIG. 42(b), pinch valves, peristaltic pumps, and waste liquid flow paths are omitted from the illustration.
前記のような従来の細胞製造装置は、流路を切り替えるためのバルブと制御回路が複雑であるため、そもそも非常に高価である。また、本発明者らの詳細な検討によれば、前記のような従来の細胞製造装置には、次の問題が含まれていることがわかった。
従来の細胞製造装置では、多段に合流するチューブが必要であり、その合流するチューブの各部をそれぞれピンチバルブにセットする作業が複雑であり、手間を要する。
多数の流路が通過する共用部分が多いため、モーター(動力)による流速の管理は元より、流路内の気泡を感知するセンサーの取り付けや、チューブ内の指定の部位への送液の到達を確認するセンサーの取り付けなどが必要である、加えて、給液バッグから培養容器X10までの流路長は必然的にチューブX30部分を確保する必要があるため長くなるため、100ml以下など低容量の液体の輸送は困難であった。
単一の密閉容器内で全工程を実施するため、最後の工程が完了した後でなければ、その細胞製造装置で他の細胞の製造を開始することができない。
The conventional cell manufacturing devices described above are very expensive because of the complex valves and control circuits for switching the flow paths. Furthermore, the inventors' detailed studies have revealed that the conventional cell manufacturing devices described above have the following problems.
Conventional cell manufacturing devices require multiple junction tubes, and the task of setting each section of the junction tubes in a pinch valve is complicated and time-consuming.
Since there are many shared areas through which many flow paths pass, it is necessary to not only manage the flow rate using a motor (power), but also to install sensors to detect air bubbles in the flow paths and sensors to confirm that the liquid has reached a specified location in the tube.In addition, the length of the flow path from the supply bag to the culture vessel X10 is necessarily long because it is necessary to secure the tube X30 section, making it difficult to transport small volumes of liquid, such as 100 ml or less.
Because all steps are carried out within a single sealed container, production of other cells cannot begin in the cell production device until the last step is completed.
本発明の目的は、従来の細胞製造装置における上記問題を抑制または解消し得る、新たな製造方法(iPS細胞の製造方法、分化細胞の製造方法)、および新たな細胞製造装置とそれを用いた細胞製造方法等を提供することにある。 The object of the present invention is to provide a new manufacturing method (a method for manufacturing iPS cells and a method for manufacturing differentiated cells) that can suppress or eliminate the above problems with conventional cell manufacturing devices, as well as a new cell manufacturing device and a cell manufacturing method using the same.
本発明の主たる構成は、以下のとおりである。
[1]人工多能性幹細胞の製造方法であって、
接続管路を介して直列に接続されたn(n≧3)個の密閉容器間を、第1番目の密閉容器(A1)から第n番目の密閉容器(An)まで、それぞれ送り機構によって、細胞を一方向に順次移動させ、各密閉容器内において人工多能性幹細胞の製造工程を順次実施することを有し、
前記密閉容器(A1~An)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、各密閉容器からその次の密閉容器へと内容物を移動させる機構であり、
前記人工多能性幹細胞の製造工程が、
第1番目の密閉容器(A1)内において、液体培地中で体細胞に初期化因子を接触させる工程(s1)と、
第2番目の密閉容器(A2)から第(n-1)番目の密閉容器(A(n-1))内において、液体培地中の初期化因子の濃度を低減させる工程(s2)と、
第n番目の密閉容器(An)内において、液体培地中で前記体細胞を培養して、人工多能性幹細胞を樹立する工程(s3)とを有する、
前記人工多能性幹細胞の製造方法。
[2]上記工程(s3)の後に、人工多能性幹細胞をp回(p≧1)拡大培養する工程(s4)をさらに有し、
上記第n番目の密閉容器(An)の後には、前記拡大培養の回数に対応するp個の密閉容器(B1~Bp)が、接続管路を介して直列に接続され、
密閉容器(An)から密閉容器(Bp)まで、それぞれ送り機構によって、人工多能性幹細胞が一方向に順次移動し、密閉容器(B1~Bp)のそれぞれの内部で1回の拡大培養が実施され、それにより、合計p回の拡大培養が実施され、
密閉容器(B1~Bp)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、各密閉容器からその次の密閉容器へと内容物を移動させる機構である、
[1]に記載の人工多能性幹細胞の製造方法。
[3]上記密閉容器が、
可撓性材料で構成された容器本体を有する密閉容器、
硬質材料で構成された容器本体を有する密閉容器、および、
可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器
からなる群から選択される密閉容器である、
[1]または[2]に記載の人工多能性幹細胞の製造方法。
[4]上記送り機構が、
移動元の密閉容器内に流体を追加することによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
移動元の密閉容器の容積を減少させることによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
上記接続管路に設けたポンプ装置によって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
移動元の密閉容器に対して、その次の密閉容器から吸引力を作用させることによって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
重力を利用して、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、および、
磁性が付与されたマイクロキャリアに細胞を接着させ、該マイクロキャリアに外部から磁力を作用させて、移動元の密閉容器内のマイクロキャリアとそれに接着した該細胞を、その次の密閉容器へと移動させる機構
からなる群から選ばれる機構、または、該群から選ばれる2以上の機構を組み合わせた機構である、
[1]~[3]のいずれか1つに記載の人工多能性幹細胞の製造方法。
[5]分化細胞の製造方法であって、
[1]~[4]のいずれか1つに記載の製造方法における人工多能性幹細胞を製造する工程と、
製造された人工多能性幹細胞を分化誘導する工程(s5)とを有し、
前記人工多能性幹細胞の製造方法で用いられた密閉容器のうち最後尾の密閉容器(X1)の後には、前記工程(s5)を実施するための密閉容器(C1)が、接続管路を介してさらに接続され、
密閉容器(C1)は、1以上の開閉可能な入出用ポートを有し、該入出用ポートを通じて、工程(s5)の分化誘導に必要な材料が密閉容器(C1)の内部に供給され、
送り機構によって、密閉容器(X1)から密閉容器(C1)へと、人工多能性幹細胞が移動し、密閉容器(C1)において前記工程(s5)が実施され、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(X1)から密閉容器(C1)へと内容物を移動させる機構である、
前記分化細胞の製造方法。
[6]上記分化誘導する工程の後に、未分化細胞を除去する工程(s6)をさらに有し、
上記分化誘導する工程で用いられた密閉容器のうち最後尾の密閉容器(X2)の後には、前記工程(s6)を実施するための密閉容器(D1)が、接続管路を介してさらに接続され、
密閉容器(D1)は、1以上の開閉可能な入出用ポートを有し、
送り機構によって、密閉容器(X2)から密閉容器(D1)へと、分化細胞が移動し、密閉容器(D1)において前記工程(s6)が実施され、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(X2)から密閉容器(D1)へと内容物を移動させる機構である、
[5]に記載の分化細胞の製造方法。
[7]細胞製造装置であって、
液体培地中で体細胞に初期化因子を接触させる工程(s1)を実施するための密閉容器(A1)と、
前記液体培地中の初期化因子の濃度を低減させる工程(s2)を実施するための密閉容器(A2)と、
前記液体培地中で前記体細胞を培養して、人工多能性幹細胞を樹立する工程(s3)を実施するための密閉容器(A3)とを有し、
密閉容器(A1)~(A3)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
密閉容器(A1)~(A3)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程の順に直列に接続されているか、または前記工程の順に直列に接続可能な状態となっており、かつ、
当該細胞製造装置は、
連通状態に切り替えられた前記接続管路を通じて、密閉容器(A1)の内容物を密閉容器(A2)へと移動させる送り機構を有し、かつ、
連通状態に切り替えられた前記接続管路を通じて、密閉容器(A2)の内容物を密閉容器(A3)へと移動させる送り機構を有する、
前記細胞製造装置。
[8]上記人工多能性幹細胞を、液体培地中でp回(p≧1)拡大培養する工程(s4)を実施するためのp個の密閉容器(B1~Bp)をさらに有し、
密閉容器(B1~Bp)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
(i)前記拡大培養の回数であるp回が1回である場合には、
前記p個の密閉容器は、1個の密閉容器(B1)であって、
密閉容器(B1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、上記密閉容器(A3)に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、上記密閉容器(A3)の内容物を密閉容器(B1)へと移動させる送り機構を有し、
(ii)前記拡大培養の回数であるp回が2回以上である場合には、
前記p個の密閉容器は、2個以上の密閉容器(B1~Bp)であって、
密閉容器(B1)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、上記密閉容器(A3)に接続されているか、または接続可能な状態となっており、
密閉容器(B1)~(Bp)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程(s4)の順に直列に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、上記密閉容器(A3)の内容物を密閉容器(B1)~(Bp)へと順に移動させる送り機構を有する、
[7]に記載の細胞製造装置。
[9]上記人工多能性幹細胞を分化誘導する工程(s5)を実施するための、q個(q≧1)の密閉容器(C1)~(Cq)をさらに有し、
密閉容器(C1)~(Cq)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
(i)前記q個の密閉容器が1個の密閉容器(C1)の場合には、
密閉容器(C1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、上記密閉容器(A3)に、もしくは密閉容器(B1)~(Bp)のうちの最後尾の密閉容器(Bp)に、接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、前記密閉容器(A3)または密閉容器(Bp)の内容物を密閉容器(C1)へと移動させる送り機構を有し、
(ii)前記q個の密閉容器が2個以上の密閉容器(C1)~(Cq)である場合には、
密閉容器(C1)~(Cq)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程(s5)の順に直列に接続されているか、または接続可能な状態となっており、かつ、密閉容器(C1)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、上記密閉容器(A3)に、もしくは密閉容器(B1)~(Bp)のうちの最後尾の密閉容器(Bp)に、接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、前記密閉容器(A3)または最後尾の密閉容器(Bp)の内容物を、密閉容器(C1)~(Cq)へと順に移動させる送り機構を有する、
[7]または[8]に記載の細胞製造装置。
[10]密閉容器(C1)~(Cq)のうちの最後尾の密閉容器(Cq)の内容物から、未分化細胞を除去する工程(s6)を実施するための密閉容器(D1)をさらに有し、
密閉容器(D1)は、1以上の開閉可能な入出用ポートを有し、
密閉容器(D1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、前記最後尾の密閉容器(Cq)に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、
連通状態に切り替えられた前記接続管路を通じて、上記最後尾の密閉容器(Cq)の内容物を密閉容器(D1)へと移動させる送り機構を有する、
[9]に記載の細胞製造装置。
[11]上記密閉容器が、
可撓性材料で構成された容器本体を有する密閉容器、
硬質材料で構成された容器本体を有する密閉容器、および、
可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器
からなる群から選択される密閉容器である、
[7]~[10]のいずれか1つに記載の細胞製造装置。
[12]上記送り機構が、
移動元の密閉容器内に流体を追加することによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
移動元の密閉容器の容積を減少させることによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
上記接続管路に設けたポンプ装置によって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
移動元の密閉容器に対して、その次の密閉容器から吸引力を作用させることによって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
重力を利用して、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、および、
磁性が付与されたマイクロキャリアに細胞を接着させ、該マイクロキャリアに外部から磁力を作用させて、移動元の密閉容器内のマイクロキャリアとそれに接着した該細胞を、その次の密閉容器へと移動させる機構
からなる群から選ばれる機構、または、該群から選ばれる2以上の機構を組み合わせた機構である、
[7]~[11]のいずれか1つに記載の細胞製造装置。
[13]上記全ての密閉容器を配置するための基板をさらに有し、
該基板上には上記密閉容器が配置され、各密閉容器は該基板に固定されており、
各密閉容器は、連通状態と非連通状態に切り替え可能な上記接続管路を介して、上記工程の順に接続されているか、または接続可能な状態となっている、
[7]~[12]のいずれか1つに記載の細胞製造装置。
[14]上記基板が、折り曲げ中心線を軸として2つ折り可能であり、
(i)前記折り曲げ中心線によって分けられた基板面の2つの領域(e1)、(e2)のうちの一方の領域(e1)には、上記密閉容器のうちの所定数の密閉容器が、該折り曲げ中心線に沿って、一つの向き(d1)に、順に並ぶように配置され、
(ii)前記折り曲げ中心線によって分けられた基板面の2つの領域(e1)、(e2)のうちの他方の領域(e2)には、上記密閉容器のうちの残りの密閉容器が、該折り曲げ中心線に沿って、前記向き(d1)とは逆の向き(d2)に、順に並ぶように配置され、
(iii)前記一方の領域(e1)の密閉容器のうちの最後尾の密閉容器と、前記他方の領域(e2)の密閉容器のうちの先頭の密閉容器とが、連通状態と非連通状態に切り替え可能な上記接続管路によって接続されているか、または接続可能な状態となっている、
[13]に記載の細胞製造装置。
[15]2枚の互いに重ね合わせられた可撓性シートをさらに有し、
上記の全ての密閉容器となる領域と、1以上の入出用ポートとなる領域と、接続管路となる領域とが、前記2枚の可撓性シートの間の所定の位置に形成されるように、これらの領域を非接合領域として残して、前記2枚の可撓性シートは互いに接合されており、
該2枚の可撓性シートの外面には、該接続管路を開閉するための押圧用アクチュエーターが設けられており、
該押圧用アクチュエーターは、前記接続管路となる領域を該可撓性シートの外側から押圧して非連通状態とする押圧ポジションと、前記接続管路となる領域を押圧せず連通状態とする非押圧ポジションとを取るように作動し、該押圧用アクチュエーターの作動により、前記接続管路となる領域は、連通状態と非連通状態とに切り替え可能な接続管路として機能し、
1以上の入出用ポートとなる領域は、各密閉容器となる領域から前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、該開口端部には、開閉可能な入出用ポートのための構成が付与されている、
[7]~[14]いずれか1つに記載の細胞製造装置。
[16]上記互いに重ね合わせられ互いに接合された2枚の可撓性シートの外周形状が、折り曲げ中心線を軸として2つ折り可能な形状であり、
(i)前記折り曲げ中心線によって分けられた2つの領域(e3)、(e4)のうちの一方の領域(e3)には、
上記密閉容器のうちの所定数の密閉容器となる領域が、該折り曲げ中心線に沿って、一つの向き(d3)に、順に並ぶように形成され、
前記所定数の密閉容器となる領域の各外周のうちの該折り曲げ中心線から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域が、該折り曲げ中心線から離れる方向に延び、かつ、前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、
前記所定数の密閉容器となる領域同士の間には、該密閉容器となる領域同士を接続する上記接続管路となる領域が形成されており、
(ii)前記折り曲げ中心線によって分けられた2つの領域(e3)、(e4)のうちの他方の領域(e4)には、
上記密閉容器のうちの残りの密閉容器となる領域が、該折り曲げ中心線に沿って、前記一つの向き(d3)とは逆の向き(d4)に、順に並ぶように形成され、
前記残りの密閉容器となる領域の各外周のうちの該折り曲げ中心線から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域が、該折り曲げ中心線から離れる方向に延び、かつ、前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、
前記残りの密閉容器となる領域同士の間には、該密閉容器となる領域同士を接続する接続管路となる領域が形成されており、
(iii)前記一方の領域(e3)の密閉容器のうちの最後尾の密閉容器と、前記他方の領域(e4)の密閉容器のうちの先頭の密閉容器とが、前記折り曲げ中心線を横切る接続管路となる領域によって接続されている、
[15]に記載の細胞製造装置。
[17][7]~[16]のいずれか1つに記載の細胞製造装置を用いた細胞製造方法であって、
上記密閉容器(A1)内において、液体培地中で体細胞に初期化因子を接触させる工程(s1)と
前記工程(s1)の完了後に、連通状態に切り替えられた前記接続管路を通じて、該密閉容器(A1)の内容物を上記密閉容器(A2)内に移動させ、該密閉容器(A2)内において、前記液体培地中の初期化因子の濃度を低減させる工程(s2)と、
前記工程(s2)の完了後に、連通状態に切り替えられた前記接続管路を通じて、該密閉容器(A2)の内容物を上記密閉容器(A3)内に移動させ、該密閉容器(A3)内において、前記液体培地中で人工多能性幹細胞を樹立する工程(s3)と
を少なくとも有する、前記細胞製造方法。
[18]上記分化誘導する工程(s5)が、人工多能性幹細胞を、外胚葉系細胞、中胚葉系細胞、または内胚葉系細胞へと分化誘導する工程(s5a)である、
[6]に記載の分化細胞の製造方法。
[19]上記工程(s5a)の後に、上記の外胚葉系細胞、中胚葉系細胞、または内胚葉系細胞を分化誘導する工程(s5b)をさらに有し、
上記密閉容器(C1)の後には、前記工程(s5b)を実施するための密閉容器(C2)が、接続管路を介してさらに接続され、
密閉容器(C2)は、1以上の開閉可能な入出用ポートを有し、該入出用ポートを通じて、工程(s5b)の分化誘導に必要な材料が密閉容器(C2)の内部に供給され、
送り機構によって、密閉容器(C1)から密閉容器(C2)へと、前記の外胚葉系細胞、中胚葉系細胞、または内胚葉系細胞が移動し、密閉容器(C2)において前記工程(s5b)が実施され、
前記接続管路は、連通状態と非連通状態とに切替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(C1)から密閉容器(C2)へと内容物を移動させる機構である、
[18]に記載の分化細胞の製造方法。
[20]上記未分化細胞を除去する工程(s6)の後に、分化細胞を検査するためのサンプルを取り出す工程(s7)をさらに有し、
上記密閉容器(D1)の後には、前記工程(s7)を実施するための密閉容器(E1)が、接続管路を介してさらに接続され、
密閉容器(E1)は、1以上の開閉可能な入出用ポートを有し、該1以上の入出用ポートを通じて、前記サンプルが外部に取り出され、
送り機構によって、密閉容器(D1)から密閉容器(E1)へと内容物が移動し、
前記接続管路は、連通状態と非連通状態とに切替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(D1)から密閉容器(E1)へと内容物を移動させる機構である、
[6]に記載の分化細胞の製造方法。
The main configuration of the present invention is as follows.
[1] A method for producing induced pluripotent stem cells, comprising:
The method comprises: sequentially moving cells in one direction from a first sealed container (A1) to an nth sealed container (An) by a feed mechanism among n (n≧3) sealed containers connected in series via a connecting pipeline; and sequentially carrying out a production process of induced pluripotent stem cells in each sealed container;
Each of the sealed containers (A1 to An) has one or more openable/closable inlet/outlet ports,
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
the feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to a communicating state,
The process for producing the induced pluripotent stem cells comprises:
A step (s1) of contacting a reprogramming factor with a somatic cell in a liquid medium in a first sealed container (A1);
A step (s2) of reducing the concentration of the reprogramming factor in the liquid medium in the second sealed container (A2) to the (n-1)th sealed container (A(n-1));
and (s3) culturing the somatic cells in a liquid medium in the n-th closed container (An) to establish induced pluripotent stem cells.
A method for producing the induced pluripotent stem cells.
[2] After the step (s3), the method further comprises a step (s4) of expanding the induced pluripotent stem cells p times (p≧1);
p sealed containers (B1 to Bp) corresponding to the number of times of expansion culture are connected in series to the n-th sealed container (An) via a connecting pipeline;
The artificial pluripotent stem cells are sequentially moved in one direction from the sealed container (An) to the sealed container (Bp) by the respective feeding mechanisms, and one expansion culture is carried out in each of the sealed containers (B1 to Bp), thereby carrying out a total of p expansion cultures;
Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports,
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to the communicating state.
A method for producing the induced pluripotent stem cell described in [1].
[3] The sealed container is
A sealed container having a container body made of a flexible material;
A sealed container having a container body made of a hard material;
A sealed container having a container body made of a composite material of a flexible material and a hard material.
A method for producing an artificial pluripotent stem cell according to [1] or [2].
[4] The feed mechanism is
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container;
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container;
a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline;
a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container;
A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and
a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
A method for producing induced pluripotent stem cells according to any one of [1] to [3].
[5] A method for producing differentiated cells, comprising the steps of:
A step of producing induced pluripotent stem cells by the production method according to any one of [1] to [4];
and a step (s5) of inducing differentiation of the produced induced pluripotent stem cells,
a sealed container (C1) for carrying out the step (s5) is further connected to the rear end of the sealed container (X1) among the sealed containers used in the method for producing induced pluripotent stem cells via a connecting pipeline;
The sealed container (C1) has one or more openable/closable inlet/outlet ports, and a material necessary for differentiation induction in step (s5) is supplied to the inside of the sealed container (C1) through the inlet/outlet ports;
The artificial pluripotent stem cells are transferred from the sealed container (X1) to the sealed container (C1) by a transfer mechanism, and the step (s5) is carried out in the sealed container (C1);
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (X1) to the sealed container (C1) through the connecting pipe line switched to a communicating state.
The method for producing the differentiated cells.
[6] The method further comprises a step (s6) of removing undifferentiated cells after the differentiation induction step;
a sealed container (D1) for carrying out the step (s6) is further connected to the rear end of the sealed container (X2) among the sealed containers used in the differentiation induction step via a connecting pipeline;
The sealed container (D1) has one or more openable/closable inlet/outlet ports,
The differentiated cells are transferred from the sealed container (X2) to the sealed container (D1) by a transfer mechanism, and the step (s6) is carried out in the sealed container (D1);
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (X2) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
The method for producing differentiated cells described in [5].
[7] A cell manufacturing device, comprising:
A sealed container (A1) for carrying out a step (s1) of contacting a somatic cell with a reprogramming factor in a liquid medium;
A sealed container (A2) for carrying out a step (s2) of reducing the concentration of the reprogramming factor in the liquid medium;
and a sealed container (A3) for carrying out a step (s3) of culturing the somatic cells in the liquid medium to establish induced pluripotent stem cells;
Each of the sealed containers (A1) to (A3) has one or more openable/closable inlet/outlet ports,
The sealed containers (A1) to (A3) are connected in series in the order of the steps via a connecting pipe line that can be switched between a communicating state and a non-communicating state, or are capable of being connected in series in the order of the steps, and
The cell manufacturing device comprises:
a feed mechanism for moving the content of the sealed container (A1) to the sealed container (A2) through the connecting pipe line switched to a communicating state,
a feed mechanism for moving the content of the sealed container (A2) to the sealed container (A3) through the connecting pipe line switched to a communicating state;
The cell manufacturing device.
[8] The method further comprises: a step (s4) of expanding the induced pluripotent stem cells in a liquid medium p times (p≧1) in a number of sealed containers (B1 to Bp);
Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports,
(i) When the number of times of expansion culture, p, is 1,
The p number of sealed containers is one sealed container (B1),
the sealed container (B1) is connected to or can be connected to the sealed container (A3) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the cell manufacturing apparatus has a transfer mechanism that transfers the content of the sealed container (A3) to the sealed container (B1) through the connecting pipeline that has been switched to a communicating state;
(ii) When the number of times of the expansion culture, p, is 2 or more,
The p number of sealed containers is two or more sealed containers (B1 to Bp),
the sealed container (B1) is connected to or can be connected to the sealed container (A3) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the sealed containers (B1) to (Bp) are connected in series or are connectable in the order of the step (s4) via connecting pipes that can be switched between a communicating state and a non-communicating state;
The cell manufacturing apparatus has a feed mechanism that moves the contents of the sealed container (A3) to the sealed containers (B1) to (Bp) in order through the connecting pipeline that is switched to a communicating state.
The cell manufacturing apparatus described in [7].
[9] Further comprising q (q≧1) sealed containers (C1) to (Cq) for carrying out the step (s5) of inducing differentiation of the induced pluripotent stem cells;
Each of the sealed containers (C1) to (Cq) has one or more openable/closable inlet/outlet ports,
(i) When the q number of sealed containers is one sealed container (C1),
the sealed container (C1) is connected or connectable to the sealed container (A3) or to the last sealed container (Bp) among the sealed containers (B1) to (Bp) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the cell manufacturing apparatus has a transfer mechanism that transfers the contents of the sealed container (A3) or the sealed container (Bp) to the sealed container (C1) through the connecting pipeline that has been switched to a communicating state;
(ii) When the q number of sealed containers is two or more sealed containers (C1) to (Cq),
the sealed containers (C1) to (Cq) are connected in series or are connectable in the order of the step (s5) via a connecting pipe that can be switched between a communicating state and a non-communicating state, and the sealed container (C1) is connected or is connectable to the sealed container (A3) or to the last sealed container (Bp) among the sealed containers (B1) to (Bp) via a connecting pipe that can be switched between a communicating state and a non-communicating state,
The cell manufacturing apparatus has a feed mechanism that moves the contents of the sealed container (A3) or the last sealed container (Bp) to the sealed containers (C1) to (Cq) in sequence through the connecting pipeline that has been switched to a communicating state.
The cell manufacturing apparatus according to [7] or [8].
[10] Further comprising a sealed container (D1) for carrying out a step (s6) of removing undifferentiated cells from the content of the last sealed container (Cq) among the sealed containers (C1) to (Cq),
The sealed container (D1) has one or more openable/closable inlet/outlet ports,
the sealed container (D1) is connected to or can be connected to the rearmost sealed container (Cq) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
The cell manufacturing device comprises:
A feed mechanism is provided for moving the contents of the rearmost sealed container (Cq) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
The cell manufacturing apparatus described in [9].
[11] The sealed container is
A sealed container having a container body made of a flexible material;
A sealed container having a container body made of a hard material;
A sealed container having a container body made of a composite material of a flexible material and a hard material.
The cell manufacturing apparatus according to any one of [7] to [10].
[12] The feed mechanism includes:
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container;
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container;
a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline;
a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container;
A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and
a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
The cell manufacturing apparatus according to any one of [7] to [11].
[13] Further comprising a substrate for arranging all of the sealed containers;
The sealed containers are disposed on the substrate, and each sealed container is fixed to the substrate;
The sealed containers are connected or connectable in the order of the steps via the connecting pipes that can be switched between a communicating state and a non-communicating state.
A cell manufacturing apparatus according to any one of [7] to [12].
[14] The substrate can be folded in two around a folding center line,
(i) in one region (e1) of two regions (e1) and (e2) on the substrate surface separated by the folding center line, a predetermined number of the sealed containers are arranged in order in one direction (d1) along the folding center line;
(ii) in the other region (e2) of the two regions (e1) and (e2) of the substrate surface separated by the folding center line, the remaining sealed containers among the sealed containers are arranged in order along the folding center line in a direction (d2) opposite to the direction (d1);
(iii) the rearmost sealed container among the sealed containers in the one region (e1) and the frontmost sealed container among the sealed containers in the other region (e2) are connected or in a connectable state by the connecting pipe line that can be switched between a communicating state and a non-communicating state;
The cell manufacturing apparatus described in [13].
[15] A method for manufacturing a flexible sheet-type display device, comprising:
the two flexible sheets are bonded to each other while leaving the regions that become all of the above-mentioned sealed containers, the region that becomes one or more inlet/outlet ports, and the region that becomes the connecting pipeline at predetermined positions between the two flexible sheets as non-bonded regions;
a pressing actuator for opening and closing the connection pipe line is provided on the outer surfaces of the two flexible sheets;
the pressing actuator operates to take a pressing position in which the region that will become the connecting pipeline is pressed from the outside of the flexible sheet to bring it into a non-communicating state, and a non-pressing position in which the region that will become the connecting pipeline is not pressed to bring it into a communicating state, and by operation of the pressing actuator, the region that will become the connecting pipeline functions as a connecting pipeline that can be switched between a communicating state and a non-communicating state;
The region that will become one or more inlet/outlet ports extends from the region that will become each sealed container to the outer periphery of the two flexible sheets to form an open end, and the open end is provided with a structure for an openable/closable inlet/outlet port.
[7] to [14] A cell manufacturing apparatus according to any one of the above.
[16] The outer peripheral shape of the two flexible sheets that are overlapped and joined to each other is a shape that can be folded in two around a folding center line,
(i) In one of the two regions (e3) and (e4) separated by the folding center line,
A predetermined number of regions that become sealed containers among the sealed containers are formed so as to be arranged in sequence in one direction (d3) along the folding center line,
from an outer periphery of each of the regions that will become the predetermined number of sealed containers that is located farther from the folding center line, a region that will become the one or more inlet/outlet ports extends in a direction away from the folding center line and extends to an outer periphery of the two flexible sheets to form an open end,
Between the regions that will become the predetermined number of sealed containers, a region that will become the connecting pipeline that connects the regions that will become the sealed containers is formed,
(ii) In the other region (e4) of the two regions (e3) and (e4) separated by the folding center line,
The remaining sealed containers are arranged in order along the folding center line in a direction (d4) opposite to the one direction (d3),
a region that becomes the one or more inlet/outlet ports extends from an outer periphery of each of the outer peripheries of the remaining region that becomes the sealed container, the outer periphery being located farther from the folding center line, in a direction away from the folding center line, and extends to an outer periphery of the two flexible sheets to form an open end;
Between the remaining regions that will become the sealed containers, a region that will become a connecting pipe that connects the regions that will become the sealed containers is formed,
(iii) the rearmost sealed container among the sealed containers in the one region (e3) and the frontmost sealed container among the sealed containers in the other region (e4) are connected by a region that becomes a connecting pipeline that crosses the folding center line;
The cell manufacturing apparatus described in [15].
[17] A cell manufacturing method using the cell manufacturing device according to any one of [7] to [16],
A step (s1) of contacting a reprogramming factor with a somatic cell in a liquid medium in the sealed container (A1); and a step (s2) of transferring the content of the sealed container (A1) into the sealed container (A2) through the connecting pipe switched to a communicating state after the completion of the step (s1), and reducing the concentration of the reprogramming factor in the liquid medium in the sealed container (A2).
The cell production method comprises at least a step (s3) of transferring the contents of the sealed container (A2) into the sealed container (A3) through the connecting pipeline that has been switched to a communicating state after completion of the step (s2), and establishing artificial pluripotent stem cells in the liquid medium within the sealed container (A3).
[18] The differentiation-inducing step (s5) is a step (s5a) of inducing differentiation of induced pluripotent stem cells into ectodermal cells, mesodermal cells, or endodermal cells.
The method for producing differentiated cells described in [6].
[19] The method further comprises, after the step (s5a), a step (s5b) of inducing differentiation of the ectodermal cell, mesodermal cell, or endodermal cell,
A sealed container (C2) for carrying out the step (s5b) is further connected to the rear of the sealed container (C1) via a connecting pipeline,
The sealed container (C2) has one or more openable/closable inlet/outlet ports, and a material necessary for differentiation induction in step (s5b) is supplied to the inside of the sealed container (C2) through the inlet/outlet ports;
the ectodermal cells, mesodermal cells, or endodermal cells are transferred from the sealed container (C1) to the sealed container (C2) by a transfer mechanism, and the step (s5b) is carried out in the sealed container (C2);
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (C1) to the sealed container (C2) through the connecting pipe line switched to a communicating state.
The method for producing differentiated cells according to [18].
[20] The method further comprises, after the step (s6) of removing undifferentiated cells, a step (s7) of removing a sample for testing differentiated cells;
A sealed container (E1) for carrying out the step (s7) is further connected to the sealed container (D1) via a connecting pipeline,
The sealed container (E1) has one or more openable/closable inlet/outlet ports, and the sample is taken out through the one or more inlet/outlet ports;
The contents are transferred from the sealed container (D1) to the sealed container (E1) by the feeding mechanism,
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (D1) to the sealed container (E1) through the connecting pipe line switched to a communicating state.
The method for producing differentiated cells described in [6].
以下、密閉容器を単に「容器」ともいう。本発明の製造方法および製造装置では、細胞を処理するために、単一の容器ではなく、複数の容器が用いられる。そして、iPS細胞や分化細胞を製造するために必要な複数の工程のそれぞれが、複数の容器のいずれかに対応付けられ、各工程がそれぞれに対応付けられた容器内で実施される。各容器内でその工程が完了すると、そこで処理された細胞は、送り機構によって、接続管路を通じて、無菌的に次段の容器に送られ、細胞は次の工程の処理を受ける。例えば、全工程が、工程s1~s3を有する場合、好ましい態様例では、1つの工程が1つの容器に対応付けられて、工程s1が第1番目の容器A1で実施され、工程s2が第2番目の容器A2で実施され、工程s3が第3番目の容器A3で実施され、そこで目的の細胞が得られる。 Hereinafter, the sealed container will be simply referred to as a "container." In the manufacturing method and manufacturing apparatus of the present invention, multiple containers are used to process cells, rather than a single container. Each of the multiple processes required to produce iPS cells or differentiated cells is associated with one of the multiple containers, and each process is carried out in the corresponding container. When the process in each container is completed, the cells processed there are aseptically sent by a sending mechanism through a connecting pipeline to the next container, where the cells are processed in the next process. For example, when all the processes include steps s1 to s3, in a preferred embodiment, one process is associated with one container, step s1 is carried out in the first container A1, step s2 is carried out in the second container A2, and step s3 is carried out in the third container A3, and the target cells are obtained there.
前記の構成により、次の効果が得られる。
上記した従来の製造装置における複雑な配管や制御機構が単純化され得、より安価に製造装置を構成することができるようになる。また、製造の各工程は、それぞれに対応付けられた各容器で完結しており、容器の連結の順番が製造工程の順番を決定するので、それらの容器の連結の順番を変更することによって、一連の製造工程を自在に構築することが可能になる。
The above-described configuration provides the following effects.
The complicated piping and control mechanism in the above-mentioned conventional manufacturing apparatus can be simplified, and the manufacturing apparatus can be constructed more inexpensively. Also, since each manufacturing process is completed by each corresponding container, and the order of connecting the containers determines the order of the manufacturing processes, it is possible to freely configure a series of manufacturing processes by changing the order of connecting the containers.
さらに、次の効果が得られる。
多段に合流するチューブが不要となり、それを装置にセットする複雑な作業も不要になる。
多数の流路が合流する構造が減少するので、共用部分を洗浄する手間も減少する。
所定の工程ごとに専用の容器を用いることにより、各工程における細胞や環境などの視認やモニタリングが容易になる。
内部の様子を外部から目視等で観察できる容器を用いれば、工程にトラブルが生じた際に、そのトラブルがどの工程で(即ち、どの容器で)生じたものかを把握することが容易である。従来のような単一の容器を用いた装置の場合、トラブルが生じた工程を知るためには、そのトラブルが生じた時期を特定する必要があり、トラブルが生じた工程を知るのに手間がかかる。
本発明によれば、例えば、第1の容器で第1の工程が完了し、細胞が第2の工程のために第2の容器へと送られるので、無菌試験等の工程内試験を行う管理区分が明確となり、第1の容器で新たな第1の工程を開始することが可能である。よって、製造工程の並列度が高められ、1つの製造装置あたりのスループットが向上する。
反応容器と工程とが一対一で対応し得るので、製造すべき細胞の違い(例えば、分化細胞の違い)に応じて、容器を簡単に追加挿入して工程を追加したり、容器を簡単に抜き取って工程を省略することができ、各工程をブロックのように増減することができる。よって、製造工程の立案段階において、製造計画書の内容に応じた容器接続のアッセンブリーが可能となる。
また、細胞が工程ごとに容器から容器へと移動する構成であるから、各工程における細胞を容器と共に装置から切り離して移動させることができる。この結果、細胞を収容した移動可能な容器は、製造ラインの流れ作業に適した可動性を獲得する、という効果も得られる。
In addition, the following effects can be obtained.
This eliminates the need for tubes that merge in multiple stages, and also eliminates the complicated task of setting them up in the device.
Since the number of structures in which multiple flow paths join together is reduced, the effort required to clean common areas is also reduced.
By using a dedicated container for each specific process, it becomes easier to visually check and monitor the cells and the environment at each process.
If a container is used whose internal state can be visually observed from the outside, when a trouble occurs in a process, it is easy to determine which process (i.e., which container) the trouble occurred in. In the case of a conventional device using a single container, in order to know the process in which the trouble occurred, it is necessary to specify the time when the trouble occurred, and it is time-consuming to know the process in which the trouble occurred.
According to the present invention, for example, the first step is completed in a first container, and the cells are sent to a second container for the second step, so that the management division for performing in-process tests such as sterility tests is clear, and it is possible to start a new first step in the first container. Therefore, the parallelism of the manufacturing process is increased, and the throughput per manufacturing device is improved.
Since there is a one-to-one correspondence between reaction vessels and processes, vessels can be easily added to add processes or removed to omit processes according to differences in the cells to be produced (for example, differences in differentiated cells), and each process can be increased or decreased like a block. Therefore, at the planning stage of the production process, it is possible to assemble the vessel connections according to the contents of the production plan.
In addition, since the cells are moved from container to container for each process, the cells can be moved together with the container separately from the device at each process, which provides the effect that the movable containers containing the cells have the mobility suitable for assembly line work on a manufacturing line.
図1は、本発明によるiPS細胞の製造方法を説明するためのブロック図であり、かつ、本発明による細胞製造装置の構成の一例を模式的に示すブロック図でもある。各密閉容器の入出用ポート、密閉容器同士の間の接続管路は、開閉可能に構成され、各密閉容器内を密閉状態とすることができる。よって、本発明の製造方法および製造装置では、閉鎖系での細胞加工や細胞培養が可能になっている。
以下、本発明の製造方法および製造装置に用いられる密閉容器を、単に「容器」という場合もあり、必要に応じて「密閉容器」という場合もある。
図1における黒い太線(h1、h2、h3など)は、軟質チューブなどの配管ラインを示している。図1では、発明の構成の主要部分を簡単に示すために、配管ライン上に設けられるコネクターや開閉バルブなどの図示は省略している。他の図の例も同様である。
図1では、各容器に接続される材料供給源(図1中の符号G1~Gn)の数は、それぞれ1つだけが示されているが、その数は限定されない。材料供給源から入出用ポートに材料を送るための送り機構は、限定されず、供給すべき材料に応じたものであってよい。例えば、ガスであれば、材料供給源であるボンベの内圧やあらゆるポンプが利用可能であり、液体や細胞懸濁液であれば、シリンジポンプやペリスタポンプなどの各種ポンプが利用可能である。他の図の例も同様である。本明細書において、「細胞懸濁液」と「細胞を含む培地」は互換的に用いられる。
各容器には、工程の操作内容に応じて材料廃棄用の外部容器(密閉容器または開放容器)が接続されてもよいが、図1では図示を省略している。他の図の例も同様である。
本発明では、容器から次段の容器へと、接続管路を通じて細胞を移動させるための送り機構が設けられる。図1では、説明のため、接続管路上にポンプとして設けられた送り機構(図1中の符号F1~Fn)を示しているが、後述のとおり、送り機構の態様は様々である。他の図の例も同様である。
1 is a block diagram for explaining the method for producing iPS cells according to the present invention, and is also a block diagram showing a schematic example of the configuration of a cell production device according to the present invention. The inlet/outlet ports of each sealed container and the connecting pipes between the sealed containers are configured to be openable and closable, so that the inside of each sealed container can be sealed. Therefore, the production method and production device of the present invention enable cell processing and cell culture in a closed system.
Hereinafter, the sealed container used in the manufacturing method and manufacturing apparatus of the present invention may be referred to simply as a "container" or, if necessary, as a "sealed container."
The thick black lines (h1, h2, h3, etc.) in Fig. 1 indicate piping lines such as soft tubes. In Fig. 1, in order to simply show the main parts of the configuration of the invention, connectors, on-off valves, etc. provided on the piping lines are omitted. The same is true for the examples in the other figures.
In FIG. 1, the number of material supply sources (reference numerals G1 to Gn in FIG. 1) connected to each container is shown as only one, but the number is not limited. The mechanism for sending the material from the material supply source to the input/output port is not limited and may be one according to the material to be supplied. For example, for gas, the internal pressure of the cylinder, which is the material supply source, or any pump can be used, and for liquid or cell suspension, various pumps such as syringe pumps and peristaltic pumps can be used. The examples in the other figures are similar. In this specification, the terms "cell suspension" and "culture medium containing cells" are used interchangeably.
Each container may be connected to an external container (sealed container or open container) for disposing of materials depending on the operation content of the process, but this is omitted in Fig. 1. The examples in the other figures are similar.
In the present invention, a feeding mechanism is provided for moving cells from a container to a container at the next stage through a connecting pipeline. For the sake of explanation, Fig. 1 shows a feeding mechanism (indicated by symbols F1 to Fn in Fig. 1) provided as a pump on the connecting pipeline, but as described below, there are various modes of the feeding mechanism. The examples in the other figures are similar.
1.人工多能性幹細胞の製造方法
先ず、本発明によるiPS細胞の製造方法(以下、「製造方法(I)」ともいう)を、本発明による細胞製造装置の構成例を参照しながら詳細に説明する。密閉容器等、装置各部の説明は、後述の細胞製造装置の各部の説明でもある。
1. Method for producing induced pluripotent stem cells First, the method for producing iPS cells according to the present invention (hereinafter also referred to as "production method (I)") will be described in detail with reference to an example of the configuration of a cell production device according to the present invention. The explanation of each part of the device, such as the sealed container, is also the explanation of each part of the cell production device described below.
図1に一例を示すように、当該製造方法(I)では、接続管路J1~J(n-1)を介して直列に接続されたn個の容器(A1~An)が用いられる。ここで、nは3以上の整数(即ち、n≧3)である。各容器には、開閉可能な入出用ポート(例えば、容器A1では符号h1など)が設けられ、各入出用ポートには、各容器に対応付けられた工程が実施されるように、必要な材料供給源(G1~Gn)が接続されている。容器に収容されている内容物K1は、細胞懸濁液であって、次段の容器へと移動するごとに細胞が変化するか、または、細胞懸濁液の構成が変化する。接続管路(より詳細な構成は後述する)は、連通状態と非連通状態とに切り替え可能である。当該製造方法(I)の重要な点は、送り機構F1~F(n-1)によって、接続管路J1~J(n-1)を通じて、第1番目の容器A1から第n番目の容器Anまで、細胞を一方向に順次移動させ、各容器内において、下記の工程s1~s3を順次実施していくという点にある。
工程s1:第1番目の容器A1内において、液体培地中で体細胞に初期化因子を接触させる工程。
工程s2:第2番目の容器A2から第(n-1)番目の容器A(n-1)内において、液体培地中の初期化因子の濃度を低減させる工程。
工程s3:第n番目の容器An内において、液体培地中で前記体細胞を培養して、iPS細胞を樹立する工程。
As shown in FIG. 1, the manufacturing method (I) uses n containers (A1 to An) connected in series via connecting pipelines J1 to J(n-1). Here, n is an integer equal to or greater than 3 (i.e., n≧3). Each container is provided with an inlet/outlet port (e.g., reference symbol h1 for container A1) that can be opened and closed, and each inlet/outlet port is connected to a necessary material supply source (G1 to Gn) so that a process associated with each container is performed. The content K1 contained in the container is a cell suspension, and the cells change or the composition of the cell suspension changes each time the content moves to the next container. The connecting pipeline (more detailed configuration will be described later) can be switched between a communicating state and a non-communicating state. The important point of this manufacturing method (I) is that the cells are sequentially moved in one direction from the first container A1 to the nth container An through the connecting pipelines J1 to J(n-1) by the feed mechanisms F1 to F(n-1), and the following steps s1 to s3 are sequentially carried out in each container.
Step s1: A step of contacting somatic cells with reprogramming factors in a liquid medium in a first container A1.
Step s2: A step of reducing the concentration of the reprogramming factor in the liquid medium in the second container A2 to the (n-1)th container A(n-1).
Step s3: A step of culturing the somatic cells in a liquid medium in the n-th container An to establish iPS cells.
容器から容器へと細胞を送りながら、各容器においてそれに対応付けられた工程を実施することで、最後の容器Anにおいて最後の工程が完了し、そこでiPS細胞が樹立する。このような複数の容器での工程の順次の実施により、上記した効果が得られ、従来の問題が抑制、軽減される。 By transferring the cells from container to container and carrying out the corresponding process in each container, the final process is completed in the last container An, where iPS cells are established. By carrying out the processes in multiple containers in sequence in this way, the above-mentioned effects are achieved, and conventional problems are prevented or alleviated.
図1の例では、工程s2が複数の処理段階(複数の工程)に分かれており、該複数の工程が容器A2~A(n-1)で順次実施されて工程s2が完了する。工程s2が完了すると、細胞は容器Anへと送られ、該容器Anで工程s3が実施される。このように、各容器内において、それぞれの容器に対応付けられた工程が順次実施されて行き、最後の容器An内において最終工程s3が完了し、該容器An内でiPS細胞が樹立する。 In the example of FIG. 1, step s2 is divided into multiple processing stages (multiple steps), and the multiple steps are carried out sequentially in containers A2 to A(n-1) to complete step s2. When step s2 is completed, the cells are sent to container An, where step s3 is carried out. In this way, the steps associated with each container are carried out sequentially in each container, and the final step s3 is completed in the last container An, and iPS cells are established in that container An.
(n個の容器(A1~An))
工程s2をどの程度の数の段階に分けるかは、特に限定はされないが、通常の処理操作では、1~3段階程度が好ましく、1段階の処理操作であっても、液体培地中の初期化因子の濃度を好ましく低減させることが可能である。よって、当該製造方法(I)における容器の数nは、特に限定はされないが、3~5程度が好ましい数である。工程s2が1段階の処理操作の場合には、工程s2に用いられるのは容器A2のみであり、上記nは3であって、当該製造方法(I)では、接続管路J1、J2を介して直列に接続された3個の容器(A1~A3)が用いられる。
(n containers (A1 to An))
There is no particular limitation on the number of stages into which step s2 is divided, but in a normal processing operation, about 1 to 3 stages are preferable, and even a one-stage processing operation can preferably reduce the concentration of the reprogramming factor in the liquid medium. Therefore, the number n of containers in the production method (I) is not particularly limited, but is preferably about 3 to 5. When step s2 is a one-stage processing operation, only container A2 is used in step s2, and the above n is 3, and in the production method (I), three containers (A1 to A3) connected in series via connecting pipes J1 and J2 are used.
(容器の直列的な接続)
本発明では、複数の容器が接続管路を介して直列に接続されるが、次のような並列接続を任意に含んだ接続であっても、各工程が、複数の容器に対応付けられて、容器A1~Anへと順次実施されるものであり、本発明でいう直列的な接続に含まれる。
(i)複数の容器(A'11、A'12、A'13、...)から1つの容器(A2)へと合流するといったように、最初に並列接続が含まれた接続。
(ii)1つの容器A1から複数の容器(A2a、A2b、A2c、...)へと並列に分岐し、それら複数の容器から1つの容器A3へと合流するといったように、途中に並列接続が含まれた接続。
(iii)1つの容器A(n-1)から、複数の容器(A'n1、A'n2、A'n3、...)へと並列に分岐して終わるといったように、最後に並列接続が含まれた接続。
(Series connection of containers)
In the present invention, multiple containers are connected in series via connecting pipelines, but even if the connection optionally includes a parallel connection as described below, each process is associated with multiple containers and performed sequentially on containers A1 to An, and this is included in the serial connection referred to in the present invention.
(i) A connection that initially includes a parallel connection, such as the merging of multiple vessels (A'11, A'12, A'13, . . . ) into one vessel (A2).
(ii) A connection that includes a parallel connection along the way, such as branching in parallel from one container A1 to multiple containers (A2a, A2b, A2c, ...) and then merging from those multiple containers into one container A3.
(iii) A connection that includes a parallel connection at the end, such as branching from one container A(n-1) to multiple containers (A'n1, A'n2, A'n3, ...) in parallel.
細胞を製造するための全工程が2つの工程である場合には、各工程は2つの容器に分けて実施される。しかし、工程の数が3以上である場合には、用いられる容器の数は、必ずしもその工程の数と同じである必要はなく(即ち、1つの工程が1つの容器に対応付けられる必要はなく)、容器の数は工程の数よりも少なくてもよい。例えば、上記した3つの工程s1~s3の場合、2つの容器を用いてもよく、容器A1で工程s1と工程s2を順次実施し、容器A2で工程s3を実施してもよいし、容器A1で工程s1を実施し、容器A2で工程s2と工程s3を実施してもよい。3以上の工程をいくつの容器に分けるかは、適宜に選択することができる。上記した本発明の効果が顕著に示される点からは、工程の数と同じ数の容器を用い、1つの容器で1つの工程を実施することが好ましい。図1の例では、1つの工程s2をさらに複数の工程に分け、分けられた各工程に1つの容器を対応付けている。 If the total number of steps for cell production is two, each step is divided into two containers and carried out. However, if the number of steps is three or more, the number of containers used does not necessarily have to be the same as the number of steps (i.e., one step does not have to correspond to one container), and the number of containers may be less than the number of steps. For example, in the case of the three steps s1 to s3 described above, two containers may be used, and steps s1 and s2 may be carried out sequentially in container A1 and step s3 may be carried out in container A2, or step s1 may be carried out in container A1 and steps s2 and s3 may be carried out in container A2. The number of containers into which three or more steps are divided can be appropriately selected. From the viewpoint of clearly demonstrating the effects of the present invention described above, it is preferable to use the same number of containers as the number of steps and carry out one step in one container. In the example of FIG. 1, one step s2 is further divided into multiple steps, and one container is associated with each divided step.
(密閉容器)
容器としては、細胞を液体培地などと共に収容し、該細胞を、無菌の環境下で加工し培養し得るものが利用可能である。該容器には、従来の細胞培養や細胞製造において用いられる種々の培養用の密閉容器を利用することができる。本発明で用いられる複数の容器の態様は、互いに異なっていてもよいが、低コスト化を図ることや、モジュール化、組織培養工学的な評価のための規格化・統一化された容器の使用、培養容器の温度管理安定性、供給培地液量の規定、培養細胞数の管理、培地成分の評価、ロット間差の是正、培地の酸素・二酸化炭素供給値の設定、容器の受け入れ試験の高速化、容器を連結したもののガス滅菌・ガンマ線滅菌・紫外線滅菌・包装形体・の統一化、容器の梱包・輸送・保管方法の統一化などの点からは、全ての容器が互いに同じ形態、同じ仕様であることが好ましい。各容器の入出用ポートは、十分な数だけ設けておき、不要の入出用ポートを閉鎖して用いれば、容器の統一化が容易である。
(sealed container)
The container can be one that can accommodate cells together with a liquid medium and process and culture the cells in a sterile environment. The container can be any of various sealed containers for culture used in conventional cell culture and cell production. The containers used in the present invention may be different from each other in terms of reducing costs, modularization, use of standardized and unified containers for tissue culture engineering evaluation, temperature control stability of the culture container, regulation of the amount of medium liquid supplied, management of the number of cultured cells, evaluation of medium components, correction of lot-to-lot differences, setting of oxygen and carbon dioxide supply values for the medium, speeding up the acceptance test of the container, unification of gas sterilization, gamma ray sterilization, ultraviolet sterilization, and packaging form of connected containers, and unification of packaging, transportation, and storage methods of the containers. It is preferable that all the containers have the same shape and specifications. If a sufficient number of inlet and outlet ports are provided for each container and unnecessary inlet and outlet ports are closed, the containers can be easily unified.
(容器の好ましい態様)
容器の好ましい態様としては、
(i)可撓性材料で構成された容器本体を有する密閉容器、
(ii)硬質材料で構成された容器本体を有する密閉容器、
(iii)可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器、などが例示される。
容器本体は、開口部(口部)を有し、該開口部が蓋や栓体などによって閉鎖されて、密閉容器となる。また、開口部に筒状のコネクターなど装着され、該コネクターの管路が活栓などによって閉鎖されて、該容器本体が密閉されてもよい。容器本体の開口部を閉鎖する蓋、栓体、入出用ポートの材料は、従来公知の細胞培養用の密閉容器などを参照して、適宜に選択してよい。
(Preferred embodiment of container)
A preferred embodiment of the container is as follows:
(i) a sealed container having a container body made of a flexible material;
(ii) A sealed container having a container body made of a hard material;
(iii) A sealed container having a container body made of a composite material of a flexible material and a hard material.
The container body has an opening (mouth), and the opening is closed with a lid, a stopper, or the like to form a sealed container. A cylindrical connector or the like may be attached to the opening, and the conduit of the connector may be closed with a stopcock or the like to seal the container body. The materials of the lid, stopper, and inlet/outlet port that close the opening of the container body may be appropriately selected with reference to conventionally known sealed containers for cell culture.
上記(i)の可撓性材料で構成された容器本体を有する密閉容器の構成は、特に限定はされないが、典型的なものとしては、従来公知の細胞培養用バッグや輸液バッグなど、可撓性フィルムで構成された柔軟なバッグが例示される(MACS(登録商標)GMP Cell Culture Bags(Miltenyi Biotec社)、Flexboy(登録商標)bags(SARTORIUS社))。可撓性フィルムは、該バッグ内の収容物の量に応じて変形できる程度の柔軟性を有するフィルムである。該バッグの構造は、限定はされないが、例えば、2枚の長方形や正方形の可撓性フィルムを重ね合わせ、開口部や入出用ポートとなる部分以外の外周縁部を互いに融着(熱融着や高周波融着等)または接着した袋の構造が例示される。該バッグを構成する可撓性フィルムは、細胞培養に必要なO2やCO2が透過可能であるガス透過性であることが好ましい。可撓性フィルムがガスを透過させないものである場合には、後述の入出用ポートを通じて、細胞培養に必要なO2やCO2を適宜に供給すればよい。さらに、可撓性フィルムは、工業的に成形加工性に優れた材料からなるもの、ガンマ線滅菌に耐えうるもの、および、内部の培地の様子を観察することができる透明性であるものが好ましい。バッグを吊り下げるための穴などの付帯的な構造は、従来公知の細胞培養バッグ等を参照して、適宜に設けることができる。本発明の製造方法で用いる上述の密閉容器は、後述する本発明の製造方法での浮遊培養を可能とするために、細胞非接着性であることが好ましい。該密閉容器は、例えば、細胞との接着性を向上させる目的で人工的に処理(例:細胞外マトリクス等によるコーティング処理)されていないものなどを使用できる。 The structure of the sealed container having a container body made of the flexible material (i) above is not particularly limited, but typical examples include flexible bags made of flexible films, such as conventionally known cell culture bags and infusion bags (MACS (registered trademark) GMP Cell Culture Bags (Miltenyi Biotec), Flexboy (registered trademark) bags (SARTORIUS)). The flexible film is a film that has flexibility to the extent that it can be deformed according to the amount of contents contained in the bag. The structure of the bag is not limited, but examples include a bag structure in which two rectangular or square flexible films are overlapped and the outer periphery parts other than the opening and the inlet/outlet port are fused (heat fusion, high-frequency fusion, etc.) or bonded to each other. The flexible film constituting the bag is preferably gas permeable, allowing O 2 and CO 2 necessary for cell culture to pass through. When the flexible film is gas impermeable, O 2 and CO 2 necessary for cell culture may be appropriately supplied through the inlet/outlet port described below. Furthermore, the flexible film is preferably made of a material that is industrially excellent in moldability, can withstand gamma ray sterilization, and is transparent so that the state of the culture medium inside can be observed. Additional structures such as holes for suspending the bag can be provided as appropriate with reference to conventionally known cell culture bags, etc. The above-mentioned sealed container used in the production method of the present invention is preferably non-cell-adhesive in order to enable suspension culture in the production method of the present invention described below. For example, the sealed container may be one that has not been artificially treated (e.g., coated with an extracellular matrix, etc.) for the purpose of improving adhesion to cells.
バッグを構成する可撓性フィルムの材料としては、ポリエチレン、ポリエチレンテレフタレート、ポリプロピレン、エチレン酢酸ビニルコポリマー(EVA)、超低密度ポリエチレン(ULDPE)/エチルビニルアルコール(EVOH)、超低密度ポリエチレン(ULDPE)、ポリオレフィン(PO)、タンクライナー、ポリフッ化ビニリデン、ポリエーテルサルフォン等、細胞培養用バッグ等に用いられる公知の材料が利用可能である。可撓性フィルムは、これらの材料製の単層フィルムであってもよいし、多層のフィルムであってもよい。 The flexible film that constitutes the bag can be made of known materials used for cell culture bags, such as polyethylene, polyethylene terephthalate, polypropylene, ethylene vinyl acetate copolymer (EVA), ultra-low density polyethylene (ULDPE)/ethyl vinyl alcohol (EVOH), ultra-low density polyethylene (ULDPE), polyolefin (PO), tank liner, polyvinylidene fluoride, polyethersulfone, etc. The flexible film can be a single layer film or a multilayer film made of these materials.
上記(ii)の硬質材料で構成された容器本体を有する密閉容器は、特に限定はされないが、フラスコ、組織培養用フラスコ、バイアル、チューブ(試験管)、バイオリアクター、ジャーファーメンター、中空糸膜バイオリアクターなど、細胞懸濁液を収容してもその重量では変形し難いものが例示される。硬質材料としては、ガラス、プラスチック材料(例えば、ポリカーボネート、ポリエステル、ポリアミド、ポリスチレン、アクリロニトリル・ブタジエン・スチレン共重合体(ABS樹脂)、ポリエチレン、ポリプロピレン、ポリ塩化ビニル、生物分解性樹脂)など、従来より細胞培養用容器に用いられている公知の材料が利用可能である。該容器本体の開口部は、蓋または栓体によって密閉され、それにより密閉容器が構成される。該入出用ポートとして、必要数の管が該蓋または該栓体を外内に貫通したものが好ましい。硬質材料で構成された容器本体を有する密閉容器は、本発明のために設計・製造されたものであってもよいが、入出用ポートを備えた市販の細胞培養容器を用いてもよい。例えば、Wilson Wolf社製のG-Rex(登録商標)10N-CSやG-Rex100N-CSなどは、蓋に十分な数の入出用ポートを備え、容器本体の底面に、細胞培養に必要なO2やCO2が透過可能なガス透過性膜を備えた細胞培養デバイスであり、本発明における密閉容器として好ましく利用可能である。 The sealed container having a container body made of the above-mentioned (ii) hard material is not particularly limited, but examples thereof include flasks, flasks for tissue culture, vials, tubes (test tubes), bioreactors, jar fermenters, hollow fiber membrane bioreactors, and the like, which are difficult to deform due to the weight of the cell suspension even when the cell suspension is contained therein. As the hard material, known materials that have been used in cell culture containers in the past, such as glass and plastic materials (e.g., polycarbonate, polyester, polyamide, polystyrene, acrylonitrile-butadiene-styrene copolymer (ABS resin), polyethylene, polypropylene, polyvinyl chloride, biodegradable resins), can be used. The opening of the container body is sealed with a lid or a stopper, thereby forming a sealed container. As the inlet/outlet port, a required number of tubes are preferably inserted through the lid or the stopper from the outside to the inside. The sealed container having a container body made of a hard material may be one designed and manufactured for the present invention, but a commercially available cell culture container equipped with an inlet/outlet port may also be used. For example, G-Rex (registered trademark) 10N-CS and G-Rex 100N-CS manufactured by Wilson Wolf are cell culture devices that have a sufficient number of inlet/outlet ports on the lid and a gas-permeable membrane on the bottom of the container body that allows the permeation of O2 and CO2 necessary for cell culture, and can be preferably used as the sealed container in the present invention.
上記(iii)の可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器としては、例えば、骨子部分が硬質材料で構成され、壁部が可撓性フィルムで構成されたものが例示される。このような容器は、硬質容器の立体的な特徴を持ちながら、安価であり、使い捨てが可能であるという特徴を有する。 An example of an airtight container having a container body made of a composite material of flexible and hard materials as described above in (iii) is one in which the framework is made of a hard material and the walls are made of a flexible film. Such containers have the three-dimensional characteristics of hard containers, but are also inexpensive and disposable.
(密閉容器内の密閉性)
本発明で利用可能な密閉容器は、接続管路のための開口部を含んだ入出用ポートを閉鎖した場合に、閉鎖系での細胞培養が可能であるような、密閉された内部空間が得られる容器である。
前記の「閉鎖系での培養が可能であるような、密閉された内部空間」は、外部から微生物やウイルスが入り込まない程度に、即ち、容器内の無菌性が保たれる程度に、壁部、開口部、入出用ポートが閉鎖された空間である。これに加えて、容器内にガスを送り込んで、内容物を所定の入出用ポートを通じて外部に押し出すなどの際に、意図せぬ他の部位からガスや内容物がリークしないように、気密に密閉された空間であることが好ましい。
例えば、細菌やウイルスは通過し得ないが、流体(とりわけガス)が通過し得る程度の多孔質フィルターが入出用ポートに設けられた容器は、外気が該多孔質フィルターを通過して容器内に流入し得るが、容器内の無菌性は保たれる。よって、その容器内は、前記の「閉鎖系での培養が可能であるような、密閉された内部空間」である。なお、そのような多孔質のフィルターを備えた入出用ポートは、バルブや活栓などによって気密に閉鎖し得るように構成されてもよい。
また、上記したWilson Wolf社製のG-Rex(登録商標)10N-CSやG-Rex100N-CSなどの容器は、容器本体の底の壁部がガス透過性膜となっており、細菌やウイルスなどは通過し得ず、細胞培養に必要なO2ガス分子やCO2ガス分子が透過可能であり、容器内は気密に密閉された空間であると言える。よって、容器内の無菌性を保つことができ、また、入出用ポートを通じて容器内にガスを送り込んで、内容物を所定の入出用ポートを通じて外部に押し出すことも可能である。よって、その容器内は、前記の「閉鎖系での培養が可能であるような密閉された内部空間」である。
(Sealability inside sealed containers)
The sealed container that can be used in the present invention is a container that, when the input/output port including the opening for the connecting pipeline is closed, provides a sealed internal space that allows cell culture in a closed system.
The above-mentioned "sealed internal space that allows for culture in a closed system" is a space whose walls, openings, and inlet/outlet ports are closed to the extent that microorganisms or viruses do not enter from the outside, that is, to the extent that sterility within the container is maintained. In addition, it is preferable that the space is airtightly sealed so that gas or contents do not leak from other unintended locations when, for example, gas is fed into the container and the contents are pushed out through a specified inlet/outlet port.
For example, a container having an inlet/outlet port provided with a porous filter that cannot pass bacteria or viruses but can pass fluids (especially gases) can allow outside air to pass through the porous filter and flow into the container, but the sterility of the container is maintained. Therefore, the inside of the container is the above-mentioned "sealed internal space that allows culture in a closed system." In addition, the inlet/outlet port equipped with such a porous filter may be configured to be airtightly closed by a valve, stopcock, etc.
In addition, the containers such as the above-mentioned G-Rex (registered trademark) 10N-CS and G-Rex 100N-CS manufactured by Wilson Wolf have a gas-permeable membrane at the bottom wall of the container body, which cannot be penetrated by bacteria or viruses, but is permeable by O2 gas molecules and CO2 gas molecules necessary for cell culture, and the inside of the container can be said to be an airtight sealed space. Therefore, it is possible to maintain the sterility of the inside of the container, and it is also possible to send gas into the container through the inlet/outlet port and push the contents out through a specified inlet/outlet port. Therefore, the inside of the container is the above-mentioned "sealed internal space that allows culture in a closed system."
(密閉容器の容積)
容器の容積は、特に限定はされず、工業的により大量のiPS細胞を一度に製造し得るような大容積であってもよいが、自家iPS細胞由来の分化細胞を用いた移植治療などの用途には、1~1000ml程度が好ましく、10~100ml程度がより好ましい。
(Volume of sealed container)
The volume of the container is not particularly limited, and may be a large volume that allows industrially large amounts of iPS cells to be produced at once; however, for applications such as transplantation therapy using differentiated cells derived from autologous iPS cells, the volume is preferably about 1 to 1,000 ml, and more preferably about 10 to 100 ml.
(入出用ポート)
入出用ポートは、容器に物質を入れ、また、容器から物質を出すために設けられる。入出用ポートは、次のような用途のために各容器に必要な数だけ設けられてよい。
(i)各容器内で実施される工程に必要な材料(体細胞、液体培地、各種試薬など)を、外部から容器内に送り込むための、供給用ポートとしての用途。
(ii)各容器内に材料を外部から容器内に送り込む際や、材料を容器内から取り出す際の圧力変化を無くす圧力調整孔(ガス抜き孔、空気供給孔、液体流出孔)としての用途。
(iii)各容器内の工程で不要になった材料(後述の初期化因子や未分化細胞を含んだ液体培地や、古い液体培地など)を外部に排出するための排出用ポートとしての用途。
(iv)検査用のサンプルや収穫物の取出口としての用途。
(v)接続管路を容器に接続するための開口としての用途。
(vi)後述の送り機構を構成するための、流体注入用ポートや吸引用ポートとしての用途。
入出用ポートは、前記の用途以外にも、必要に応じた種々の入出口として設けてよい。
(Entry/Exit Port)
Input and output ports are provided for introducing materials into and removing materials from the container. As many input and output ports as necessary may be provided on each container for such purposes as:
(i) Use as a supply port for delivering materials (such as somatic cells, liquid culture medium, and various reagents) required for the process carried out in each vessel from the outside into the vessel.
(ii) Use as a pressure adjustment hole (gas vent hole, air supply hole, liquid outflow hole) to eliminate pressure changes when materials are sent into each container from the outside or when materials are removed from the container.
(iii) Use as a discharge port for discharging materials (such as liquid medium containing reprogramming factors or undifferentiated cells described below, or old liquid medium) that are no longer needed in the process within each container.
(iv) As an outlet for sampling or harvesting products for testing.
(v) Use as an opening for connecting a connecting line to a vessel.
(vi) Use as a fluid injection port or suction port to construct the feeding mechanism described below.
The inlet/outlet port may be provided as various inlets/outlets as required in addition to the above uses.
(入出用ポートの数)
1つの容器に設けられる入出用ポートの数は、必ずしも上記の用途の数である必要はない。流路を切り替えるバルブや分岐管などを用いて、または、外部管路の着脱などによって、1つの入出用ポートが複数の用途のために用いられてもよい。例えば、上記(i)の細胞培養に必要なガスを供給するための入出用ポートは、上記(vi)の内容物を容器内から押し出す際にガスを供給するための入出用ポートとしても利用可能である。また、上記(ii)の圧力調整孔としての入出用ポートは、上記(iii)の排出用としての入出用ポートとしても利用可能である。液体培地の交換において、新しい液体培地を容器内に供給して、該容器内から古い液体培地を押し出すような場合には、2つの入出用ポートが用いられる。1つの入出用ポートを多数の用途に用いると、1つの入出用ポートに対する複数の外部管路の着脱や切り替えなどが手間になるか、配管が複雑になる場合がある。よって、用途の数だけ入出用ポートを設けてもよいし、1つの入出用ポートを適当な数の用途に用いてもよい。このような点から、1つの容器に設けられる入出用ポートの数は、概ね3~10程度である。
(Number of entry/exit ports)
The number of inlet/outlet ports provided in one container does not necessarily have to be the number of the above applications. One inlet/outlet port may be used for multiple applications by using a valve or branch pipe for switching the flow path, or by attaching and detaching an external pipeline. For example, the inlet/outlet port for supplying gas necessary for cell culture in (i) above can also be used as an inlet/outlet port for supplying gas when pushing out the contents from the container in (vi) above. In addition, the inlet/outlet port as a pressure adjustment hole in (ii) above can also be used as an inlet/outlet port for discharge in (iii) above. In the case of replacing a liquid culture medium, when a new liquid culture medium is supplied into a container and an old liquid culture medium is pushed out from the container, two inlet/outlet ports are used. If one inlet/outlet port is used for multiple applications, it may be troublesome to attach and detach or switch multiple external pipelines to one inlet/outlet port, or the piping may become complicated. Therefore, the number of inlet/outlet ports may be provided for the number of applications, or one inlet/outlet port may be used for an appropriate number of applications. For these reasons, the number of inlet/outlet ports provided in one container is generally about 3 to 10.
入出用ポートの数は、容器ごとに異なっていてもよいが、容器の仕様を統一して互換性や低コスト化を図る点からは、どの密閉容器にも同数の入出用ポートを設けておき、使用しない入出用ポートを閉鎖して容器を用いてもよい。 The number of inlet/outlet ports may vary from container to container, but in order to standardize container specifications to ensure compatibility and reduce costs, it is possible to provide the same number of inlet/outlet ports on all sealed containers and close unused inlet/outlet ports when using the container.
(入出用ポートのコネクター)
入出用ポートは、容器の壁部や蓋などに設けられた単純な貫通孔であってよく、また、該貫通孔に気密に挿通された短い継手用のパイプ材などであってもよい。外部の配管や外部のシリンジの先端部などを、堅固にかつリーク無きように入出用ポートに接続でき、しかも、容易に着脱し得るという点からは、該入出用ポートが適当なコネクターを備えていることが好ましい。しかし、容器の栓体などに多数のコネクターを直接固定することは、コネクター同士が干渉して困難な場合がある。そこで、図2の態様では、この問題を解消する入出用ポートの構成が付与されている。同図の例では、各容器10に3つの入出用ポート11が設けられている。各容器10は、容器本体10aの開口部を栓体10bで閉鎖した構成を有する。同図における各入出用ポート11は、栓体10bを内外方向に貫通する管部材11aを有する。同図の例では、各管部材は軟質チューブであるが、貫通部分の管部材を硬質材料からなるものとしてもよい。該管部材11aの外界側の遠位端部(栓体から最も離れた端部)にルアーコネクター11bが設けられている。これにより、3つの入出用ポートのコネクターは互いに干渉せず、各コネクターへの着脱の作業も容易であり、着脱の際に加える力が栓体に大きく影響することもない。3つの管部材のうちの中央の管部材には、管路を閉鎖するためのクリップが装着されている。
(Input/Output port connector)
The inlet/outlet port may be a simple through hole provided in the wall or lid of the container, or may be a short pipe material for a joint airtightly inserted into the through hole. It is preferable that the inlet/outlet port has an appropriate connector so that an external pipe or the tip of an external syringe can be firmly and leak-free connected to the inlet/outlet port and can be easily attached and detached. However, it may be difficult to directly fix a large number of connectors to the stopper of the container because the connectors may interfere with each other. Therefore, in the embodiment of FIG. 2, an inlet/outlet port configuration that solves this problem is provided. In the example of the figure, three inlet/
入出用ポートのコネクターとしては、あらゆる管継手(コネクターやカップリングなどとも呼ばれる)が利用可能であり、例えば、ルアーコネクター(旧ISO594-1、旧ISO594-2など)、誤接続防止コネクター(ISO80369)、無菌接続継手、ワンタッチ式無菌切断継手、チューブ用プッシュイン継手、ワンタッチ継手などが例示される。また、バイアル瓶のゴム栓のように注射針を穿刺するように構成された栓体と、それを穿刺する注射針やケモクレーブバッグスパイクとの組合せも、本発明においては、開閉可能なコネクターに含まれる。 Any type of pipe joint (also called a connector or coupling) can be used as the connector for the inlet/outlet port, such as a luer connector (old ISO 594-1, old ISO 594-2, etc.), a connector to prevent misconnection (ISO 80369), a sterile connection joint, a one-touch type sterile disconnect joint, a push-in joint for tubing, a one-touch joint, etc. Also, in the present invention, the combination of a stopper configured to be pierced by a syringe needle, such as a rubber stopper for a vial, and a syringe needle or a chemoclav bag spike that pierces it, is also included in the openable/closable connector.
(入出用ポートを開閉する構成)
入出用ポートを開閉する構成は、特に限定はされず、係合を解除したコネクターの接続端部を閉鎖するキャップ、入出用ポートとして設けられたチューブの管路を閉じるピンチバルブ、クリップ、活栓などが例示される。また、三方活栓を用いた流路の切り替えも入出用ポートの開閉する機構として用いてもよい。また、入出用ポートに接続されたままの外部管路が開閉可能に構成されている場合も、入出用ポートを開閉する構成に含まれる。また、上記した栓体とそれを穿刺する注射針との組合せも、入出用ポートを開閉する構成に含まれる。
(Configuration for opening and closing input/output ports)
The configuration for opening and closing the inlet/outlet port is not particularly limited, and examples include a cap for closing the connection end of a disengaged connector, a pinch valve for closing the conduit of a tube provided as an inlet/outlet port, a clip, a stopcock, and the like. Switching of flow paths using a three-way stopcock may also be used as a mechanism for opening and closing the inlet/outlet port. In addition, a configuration for opening and closing the inlet/outlet port includes a case where an external conduit that remains connected to the inlet/outlet port is configured to be openable and closable. In addition, a combination of the above-mentioned stopper and an injection needle that pierces it is also included in the configuration for opening and closing the inlet/outlet port.
(入出用ポートの容器内側の構成)
図3は、容器の内側における入出用ポートの構成を例示するブロック図であって、説明のために容器は断面を示している。図3の例では、図2と同様に、容器10は、容器本体10aの開口部を栓体10bで閉鎖した構成を有し、該容器10には、3つの入出用ポート111、112、113が設けられている。各入出用ポートは、管部材(軟質チューブ)が栓体10bを内外方向に貫通する構成を有し、該管部材は容器の内側にも延びている。該管部材の外界側の遠位端部には管継手11b1が設けられている。また、該管部材の外界側には、管路を開閉するためのデバイス12が設けられている。他の入出用ポート112、113の外界側も同様に構成されているが図示を省略している。
(Configuration of the container inside the inlet/outlet port)
3 is a block diagram illustrating the configuration of an inlet/outlet port inside a container, and the container is shown in cross section for the purpose of explanation. In the example of FIG. 3, similar to FIG. 2, the
図3の例では、入出用ポート111、112、113の各管部材は、容器内に延びているが、その長さに特徴がある。
図3(a)、(b)の例では、入出用ポート111の管部材の容器内部分が、内容物(細胞懸濁液など)の液面に達していない。このような容器内部分は、内容物を撹拌することなしに、容器内にガスを導入できるので、容器内部の液量に影響を受けることなくガスを用いた容圧の調節が可能であり、好ましい。該ガスは、細胞培養に必要なO2やCO2のほか、容器内の内容物を押し出すための空気などであってもよい。
図3(a)、(b)の例では、入出用ポート112の管部材の容器内部分が、液面から適当な深さの表層まで内容物内に入り込んでいる。このような容器内部分は、液体培地などに強い対流を生じさせることなく、よって、沈降した細胞を舞い上がらせることなく、容器内に液体培地を供給し得る点で好ましい。よって、このような容器内部分は、細胞培養における液体培地の交換、初期化因子の濃度を低下させるための希釈、細胞の洗浄などの操作に適している。
図3(a)の例では、入出用ポート113の管部材の容器内部分が、入出用ポート112と同様に、液面から適当な深さの表層まで内容物である液体内に入り込んでいる。このような容器内部分は、液体培地の液面付近の液体(上清)を静かに容器外に取り出す場合に好ましく用いることができる。また、図3(a)の例のような2つの入出用ポート112、113を用いることで、これらの入出用ポートの向きによっては、液体培地に渦を生じさせることもでき、旋回培養を行うこともできる。
図3(b)の例では、入出用ポート113の管部材の容器内部分は、容器本体の底面付近に達している。このような容器内部分は、液体培地やガスの供給口として用いれば、強い縦の対流を生じさせ、沈降した細胞に新しい液体培地やガスを送り込むことができる。また、容器内の内容物の排出口や後述の接続管路用のポートとして用いれば、該内容物の液面が容器の底面付近に達するまで、該内容物を排出することができる。
In the example of FIG. 3, the pipe members of the inlet/
In the example of Figures 3(a) and (b), the inside of the container of the tube member of the inlet/
In the example of Figures 3(a) and (b), the inside of the container of the tube member of the inlet/
In the example of Fig. 3(a), the inside of the container of the tube member of the inlet/
In the example of Fig. 3(b), the inside of the vessel of the tube member of the inlet/
(接続管路)
本発明において、接続管路は、容器同士を接続するためのものであって、連通状態と非連通状態とに切り替え可能な構成を有する。連通状態とは、接続管路内を通って容器から次段の容器へと流体が移動できる状態をいい、非連通状態とは、容器から容器へ流体が移動できない状態をいう。該接続管路の態様は、特に限定はされないが、次のような態様が挙げられる。
(態様i)図4(a)に例示するように、容器A1と容器A2とを接続するように接続管路J1が設けられている。該接続管路J1のライン上には、種々の開閉バルブ(管路を開閉するように設けられたシャッターをも含む)、活栓、クリップなどの開閉装置J1aが設けられており、該開閉装置J1aの開閉動作によって、該接続管路J1は、連通状態と非連通状態とに切り替わることができる。
(態様ii)図4(b)に例示するように、容器A1と容器A2とを接続するように接続管路J1が設けられている。該接続管路J1のライン上または両端部には、コネクターJ1bが設けられる。該コネクターJ1bはいわゆる開閉装置ではないが、該コネクターJ1bを結合すると、該接続管路J1は連通状態に切り替わり、該コネクターJ1bを分離すると、該接続管路J1は、図4(b)に示すとおり、非連通状態に切り替わる。このようなコネクターは、入出用ポートに接続するためのコネクターであってもよい。
(態様iii)図4(c)に例示するように、容器A1には、容器A1と容器A2とを接続するための接続管路J1が設けられている。該接続管路J1の先端部には注射針J1cが装着されている。該注射針J1cは、容器A2の栓体に穿刺されていない状態にあり、よって、容器A1と容器A2とは該接続管路J1によって接続されてはないが、接続可能な状態(即ち、穿刺可能な状態)となっている。容器A1から容器A2へと内容物を移動させる際に、該注射針J1cを容器A2の栓体に穿刺することによって、該接続管路J1は非連通状態から連通状態に切り替わる。図4(c)の例では、容器本体の開口部を単一の栓体で密封しているが、開口部を密封する蓋の所定位置に、注射針J1cを穿刺するための栓体を局所的に設けてもよい。注射針J1cの内径は、細胞やマイクロキャリアが通過可能なものであればよく、23ゲージ(G)程度またはそれよりも太い内径の注射針が好ましく利用でき、より好ましくは18G程度が例示される。
(Connecting pipeline)
In the present invention, the connecting pipeline is for connecting containers to each other and has a configuration that can be switched between a communicating state and a non-communicating state. The communicating state refers to a state in which a fluid can move from a container to a container in the next stage through the connecting pipeline, and the non-communicating state refers to a state in which a fluid cannot move from one container to another. The form of the connecting pipeline is not particularly limited, but examples include the following forms.
4(a), a connecting pipe J1 is provided to connect a container A1 and a container A2. On the line of the connecting pipe J1, various opening and closing devices J1a such as open/close valves (including shutters provided to open and close the pipe), stopcocks, clips, etc. are provided, and the connecting pipe J1 can be switched between a connected state and a non-connected state by opening and closing the opening and closing devices J1a.
(Aspect ii) As shown in Fig. 4(b), a connecting pipe J1 is provided to connect container A1 and container A2. A connector J1b is provided on the line or at both ends of the connecting pipe J1. Although the connector J1b is not a so-called opening and closing device, when the connector J1b is connected, the connecting pipe J1 switches to a connected state, and when the connector J1b is separated, the connecting pipe J1 switches to a non-connected state as shown in Fig. 4(b). Such a connector may be a connector for connecting to an input/output port.
(Aspect iii) As shown in FIG. 4(c), the container A1 is provided with a connection pipe J1 for connecting the container A1 and the container A2. An injection needle J1c is attached to the tip of the connection pipe J1. The injection needle J1c is not pierced into the stopper of the container A2, and therefore the container A1 and the container A2 are not connected by the connection pipe J1, but are in a connectable state (i.e., a pierceable state). When the contents are transferred from the container A1 to the container A2, the injection needle J1c pierces the stopper of the container A2, and the connection pipe J1 is switched from a non-communicating state to a communicating state. In the example of FIG. 4(c), the opening of the container body is sealed with a single stopper, but a stopper for piercing the injection needle J1c may be locally provided at a predetermined position of the lid that seals the opening. The inner diameter of the injection needle J1c may be any diameter that allows cells or microcarriers to pass through. An injection needle with an inner diameter of about 23 gauge (G) or larger is preferably used, and more preferably, about 18 G is exemplified.
本発明において、容器が、他の容器に対して、接続管路を介して「接続可能な状態となっている」とは、前記の注射針と栓体とを介した容器同士の関係(栓体に注射針が穿刺されていないので接続はされていないが、栓体に注射針が穿刺されると接続される関係)や、係合が解除されたコネクターを介した容器同士の関係(コネクターが係合していないので接続はされていないが、コネクターが係合すると接続される関係)のように、分断された接続管路が結合可能な状態にあり、該接続管路が結合することで容器同士が接続されるといったような、接続前の状態であることを意味する。 In the present invention, when a container is "connectable" to another container via a connecting pipeline, it means that the containers are in a pre-connection state, such as the relationship between the containers via the injection needle and the stopper (there is no connection because the injection needle has not been pierced into the stopper, but will be connected when the injection needle is pierced into the stopper) or the relationship between containers via disengaged connectors (there is no connection because the connectors are not engaged, but will be connected when the connectors are engaged), in which case the disconnected connecting pipelines are in a connectable state and the containers are connected when the connecting pipelines are joined.
図4(a)~(c)における接続管路が接続された入出用ポートは説明のためのものであって、接続管路を、どの位置のどのような構成の入出用ポートに接続するかは適宜に設計することができる。また、たとえ接続管路それ自体が開閉可能な構成を有していなくとも、該接続管路に接続される入出用ポートなどが開閉可能であって、それにより接続管路が連通状態と非連通状態とに切り替え可能になっていればよい。 The inlet/outlet ports to which the connecting pipelines are connected in Figures 4(a) to (c) are for illustrative purposes only, and the position and configuration of the inlet/outlet ports to which the connecting pipelines are connected can be designed as appropriate. Furthermore, even if the connecting pipeline itself does not have a configuration that allows it to be opened and closed, it is sufficient that the inlet/outlet ports connected to the connecting pipeline can be opened and closed, thereby allowing the connecting pipeline to be switched between a connected state and a non-connected state.
(送り機構)
本発明における送り機構は、連通状態に切り替えられた上記の接続管路を通じて、各容器からその次の容器へと内容物を移動させる機構である。ここでいう「各容器」とは、移動元の容器であって、自体の次段に内容物を移動させるべき容器が存在するような容器(即ち、最後尾の容器以外の容器)である。
(Feed mechanism)
The feed mechanism in the present invention is a mechanism for transferring the contents from each container to the next container through the above-mentioned connecting pipes switched to the communicating state. The "each container" here refers to a source container that has a container to which the contents should be transferred in the next stage (i.e., a container other than the last container).
内容物の移動における次段の容器内の圧力調整(空気抜きなどの圧力開放)は適当な入出用ポートを利用することができる。次段の容器が、細胞培養用バッグなどの柔軟な空のバッグの場合、内容物の移動に伴って次段の容器が膨れるので、圧力調整が必要ない場合もある。移動元の容器内の内容物を外部から吸引して取り出す場合における、該容器内の圧力調整(空気の流入などの圧力開放)も同様であって、適当な入出用ポートを利用することができる。該移動元の容器が、細胞培養用バッグなどの柔軟なバッグの場合、内容物の取り出しに伴って該バッグが収縮するので、圧力調整が必要ない場合もある。 Appropriate inlet/outlet ports can be used to adjust the pressure inside the next container when the contents are being transferred (pressure relief such as by venting air). If the next container is an empty flexible bag such as a cell culture bag, the next container will expand as the contents are transferred, so pressure adjustment may not be necessary. The same applies to pressure adjustment inside the container when the contents of the source container are removed by suction from the outside (pressure relief such as by letting air in), and appropriate inlet/outlet ports can be used. If the source container is a flexible bag such as a cell culture bag, the bag will shrink as the contents are removed, so pressure adjustment may not be necessary.
送り機構の具体的な態様としては、次に示す(i)~(vi)の機構からなる群から選ばれる機構が挙げられ、また、該群から選ばれる2以上の機構を組み合わせた機構であってもよい。 Specific embodiments of the feed mechanism include a mechanism selected from the group consisting of the mechanisms (i) to (vi) shown below, and may also be a mechanism that combines two or more mechanisms selected from the group.
(i)内容物を容器から押し出す機構(1)
この機構は、例えば、図4(a)の構成などにおいて、移動元の容器A1内に流体(ガスや液体培地など)を追加して(入出用ポートを通じて供給して)、移動元の密閉A1の内容物を次段の容器A2へと押し出して移動させる機構である。移動元の容器に送り用のガスを注入する態様では、該ガスを注入するための入出用ポートは、送り機構専用であってもよいし、細胞培養のためのガスを注入するための入出用ポートを兼用してもよい。送り用のガスは、空気(好ましくはフィルターを通した清浄な空気(酸素濃度20%程度、二酸化炭素濃度5%程度))や、細胞培養に必要なO2やCO2など、細胞に悪影響を与えない任意のガスが利用可能である。また、移動元の容器A1内に液体培地を供給して、内容物を次段の容器A2へと押し出して移動させることも可能である。この場合、容器A1内の内容物と供給された液体培地とが混合されて押し出されるので、容器A1内の内容物の希釈と移動とを同時に実施していると解することもできる。移動元の容器A1内への流体の供給には、任意の外部ポンプ装置が利用可能であり、材料供給源の送り出し機構を利用してもよい。前記の説明では、容器A1からA2への移動を例として示しているが、以降の容器A3~Anに内容物を移動させる機構にも適用してよい。
(i) A mechanism for pushing the contents out of the container (1)
This mechanism is a mechanism for adding a fluid (gas, liquid medium, etc.) to the source container A1 (supplied through an inlet/outlet port) in the configuration of FIG. 4(a) and pushing out and moving the contents of the source sealed container A1 to the next container A2. In the embodiment in which a feed gas is injected into the source container, the inlet/outlet port for injecting the gas may be dedicated to the feed mechanism, or may also be used as an inlet/outlet port for injecting gas for cell culture. The feed gas may be any gas that does not adversely affect cells, such as air (preferably clean air passed through a filter (oxygen concentration of about 20%, carbon dioxide concentration of about 5%)) or O 2 or CO 2 necessary for cell culture. It is also possible to supply a liquid medium into the source container A1 and push out and move the contents to the next container A2. In this case, the contents in the container A1 and the supplied liquid medium are mixed and pushed out, so it can be understood that dilution and movement of the contents in the container A1 are performed simultaneously. Any external pump device can be used to supply fluid to the source container A1, or a mechanism for sending out the material from the material source can be used. In the above explanation, the movement from container A1 to A2 is shown as an example, but the same can also be applied to a mechanism for moving the contents to the subsequent containers A3 to An.
(ii)内容物を容器から押し出す機構(2)
この機構は、図5(a)に一例を示すように、移動元の容器A1の容積を減少させることによって、その内容物K1をその次の容器(図示せず)へと押し出して移動させる機構である。図5(a)の例では、容器A1は柔軟な細胞培養用バッグであって、設置面上に横たわっている。エアシリンダーなど押圧装置の先端に取付けられた押圧ヘッド部材1によって、該容器A1が外側から押圧され圧縮されてその容積が減少し、入出用ポート11と接続管路J1を通じて、内容物K1が次段の容器(図示せず)へと押し出される。図の例では、容器A1から次の容器への移動を例として示しているが、以降の容器A3~Anに内容物を移動させる機構にも適用してよい。図中の破線の矢印は、押し出される内容物を示唆している。容器に対する押圧は、押圧装置などを用いて自動で行ってもよいし、人力で行ってもよい。図5(a)の例では、容器として柔軟なバッグが用いられているが、該容器全体がシリンジの構造を有するものなど、容積が変動し内容物を押し出すことができる容器を用いてもよい。
(ii) A mechanism for pushing the contents out of the container (2)
As shown in FIG. 5(a), this mechanism reduces the volume of the source container A1, and pushes out and moves the contents K1 to the next container (not shown). In the example of FIG. 5(a), the container A1 is a flexible cell culture bag lying on the installation surface. The container A1 is pressed from the outside by a
(iii)ポンプ装置を介在させて内容物を移動させる機構
この機構は、図1に例示するように、2つの容器A1、A2の間の接続管路J1に設けたポンプ装置F1によって、移動元の容器A1の内容物K1をその次の容器A2へと移動させる機構である。この機構に利用可能なポンプ装置は、特に限定はされないが、細胞にダメージを与えることなく内容物(懸濁液)を送ることができるポンプ装置が好ましく、例えば、ペリスタポンプ、シリンジポンプなどが好ましいものとして挙げられる。ポンプ装置は、手動で作動するものであってもよいし、駆動源や制御部を備えた自動装置であってもよい。図5(b)の例では、接続管路J1に挿入されたT字管F1cに、ポンプ装置F1としてシリンジポンプが接続されている。該シリンジポンプの吸引と吐出の作動によって、内容物が容器A1から容器A2へと一方向に移動するよう、T字管F1cの前後には、逆流防止弁F1a、F2bが接続されている。図の例では、容器A1からA2への移動を例として示しているが、以降の容器A3、A4...に内容物を移動させる機構にも適用してよい。逆流防止弁の代わりに活栓を用いることもできる。逆流防止弁F1a、F2bに描かれた矢印は、流れが一方向に制限されることを示している。図5(b)におけるT字管F1cの代わりに、三方活栓や三方向電磁弁を用いれば、逆流防止弁F1a、F2bを省略することができ、該シリンジポンプの吸引時には内容物が容器A1から該シリンジポンプへと流れ、該シリンジポンプの吐出時には内容物が該シリンジポンプから容器A2へと流れる。
(iii) A mechanism for moving contents through a pump device As illustrated in FIG. 1, this mechanism moves the contents K1 of the source container A1 to the next container A2 by a pump device F1 provided in a connecting pipe J1 between two containers A1 and A2. The pump device that can be used for this mechanism is not particularly limited, but is preferably a pump device that can send the contents (suspension) without damaging the cells, and examples of such pump devices include a peristaltic pump and a syringe pump. The pump device may be operated manually or may be an automatic device equipped with a drive source and a control unit. In the example of FIG. 5(b), a syringe pump is connected as the pump device F1 to a T-shaped pipe F1c inserted into the connecting pipe J1. Check valves F1a and F2b are connected before and after the T-shaped pipe F1c so that the contents are moved in one direction from the container A1 to the container A2 by the suction and discharge operation of the syringe pump. In the example shown in the figure, the movement from container A1 to A2 is shown as an example, but the mechanism may also be applied to a mechanism for moving the contents to subsequent containers A3, A4, etc. A stopcock may be used instead of the check valve. The arrows drawn on the check valves F1a and F2b indicate that the flow is restricted to one direction. If a three-way stopcock or a three-way solenoid valve is used instead of the T-shaped pipe F1c in FIG. 5(b), the check valves F1a and F2b can be omitted, and the contents flow from container A1 to the syringe pump when the syringe pump is sucking, and the contents flow from the syringe pump to container A2 when the syringe pump is discharging.
(iv)次段の容器から内容物を吸引する機構
この機構は、例えば、図4(a)の構成などにおいて、移動元の容器A1に対して、次段の容器A2から吸引力を作用させて、移動元の容器A1の内容物K1を次段の容器A2へと吸い込んで移動させる機構である。前記吸引力を作用させるためには、例えば、容器A2の入出用ポートを通じて、該容器A2内の空気を容器外へと吸い出し、それにより、接続管路J1を通じて容器A1の内容物K1に吸引力を作用させる。容器A2内の空気を容器外へと吸い出す場合、該空気を容器外へと吸い出すための入出用ポートは、送り機構専用であってもよいし、細胞培養のためのガスを注入するための入出用ポートなどを兼用してもよい。また、次段容器A2全体がシリンジの構造を有するものなど、接続管路J1を通じて容器A1の内容物K1を吸引することができる容器を用いてもよい。この説明では、容器A1からA2への移動を例として示しているが、以降の容器A3~Anに内容物を移動させる機構にも適用してよい。
(iv) Mechanism for sucking contents from the next container This mechanism is a mechanism for sucking and moving the contents K1 of the source container A1 to the next container A2 by applying suction force from the next container A2 to the source container A1 in the configuration of FIG. 4(a), for example. In order to apply the suction force, for example, the air in the container A2 is sucked out of the container through the inlet/outlet port of the container A2, and thereby the suction force is applied to the contents K1 of the container A1 through the connecting pipe J1. When sucking out the air in the container A2 to the outside of the container, the inlet/outlet port for sucking out the air to the outside of the container may be dedicated to the feed mechanism, or may also be used as an inlet/outlet port for injecting gas for cell culture. In addition, a container capable of sucking the contents K1 of the container A1 through the connecting pipe J1, such as one in which the entire next container A2 has a syringe structure, may be used. In this explanation, the movement from container A1 to container A2 is shown as an example, but the present invention may also be applied to a mechanism for moving the contents to subsequent containers A3 to An.
(v)重力を利用して内容物を移動させる機構
この機構は、重力が利用できるように、2つの容器の位置関係と、接続管路の接続とによって構成された機構である。この機構は、例えば、図6(a)に一例を示すように、移動元の容器A1よりも下方に次段の容器A2を配置し、重力によって、移動元の容器の内容物を次段の容器A2へと降下させて移動させる機構である。図6(a)の例では、容器A1の真下に容器A2が配置されており、容器A1の底面に接続管路J1が接続されており、該接続管路J1上に開閉装置J1aが設けられている。容器A1内において工程s1が完了すると、開閉装置J1aが開いて、接続管路J1が連通状態に切り替わり、容器A1内の内容物が、重力によって、接続管路J1を通じて次段の容器A2内へと落下する。
図6(a)の例の他に、サイフォンの原理によって容器A1から次段の容器A2内へと内容物が流出する構成も、重力を利用して内容物を移動させる機構である。
(v) Mechanism for moving contents by utilizing gravity This mechanism is configured by the positional relationship of two containers and the connection of connecting pipes so that gravity can be utilized. For example, as shown in FIG. 6(a), this mechanism is a mechanism in which a next-stage container A2 is placed below a source container A1, and the contents of the source container are moved by gravity by descending to the next-stage container A2. In the example of FIG. 6(a), the container A2 is placed directly below the container A1, a connecting pipe J1 is connected to the bottom surface of the container A1, and an opening/closing device J1a is provided on the connecting pipe J1. When the process s1 is completed in the container A1, the opening/closing device J1a opens, the connecting pipe J1 is switched to a communicating state, and the contents in the container A1 fall into the next-stage container A2 through the connecting pipe J1 by gravity.
In addition to the example of FIG. 6(a), a configuration in which the contents flow from container A1 into container A2 in the next stage by the principle of a siphon is also a mechanism for moving the contents by utilizing gravity.
図6(b)の例では、容器A1と容器A2とが隣接しており、容器A1の内部空間と容器A2の内部空間とは、シャッター装置J1aの開閉可能な仕切り板J1a1によって互いに隔離されている。該仕切り板J1a1によって開閉される領域J1が、本発明でいう連通状態と非連通状態とに切り替え可能な接続管路に該当する。図では、該仕切り板J1a1は閉鎖位置にある。該仕切り板J1a1がシャッター装置J1aの本体内に完全に引き込まれて開放位置に移動すると、容器A1と容器A2とは1つの内部空間として一体になる。図6(b)に示すように、容器A2は容器A1よりも下方に位置しており、容器A1から容器A2へと細胞Mが滑り降りるように、容器A1と容器A2の底面は、1つの傾斜面を構成している。 In the example of FIG. 6(b), container A1 and container A2 are adjacent to each other, and the internal space of container A1 and the internal space of container A2 are isolated from each other by an openable and closable partition plate J1a1 of a shutter device J1a. The area J1 opened and closed by the partition plate J1a1 corresponds to the connection pipeline that can be switched between a connected state and a non-connected state in the present invention. In the figure, the partition plate J1a1 is in a closed position. When the partition plate J1a1 is completely retracted into the main body of the shutter device J1a and moves to an open position, container A1 and container A2 become one internal space. As shown in FIG. 6(b), container A2 is located below container A1, and the bottom surfaces of container A1 and container A2 form a single inclined surface so that cells M can slide down from container A1 to container A2.
仕切り板J1a1は、シャッター装置J1aの本体内に任意の量だけ引き込まれた状態で停止することも可能である。仕切り板J1a1が少しの量だけ引き込まれた状態になると、接続管路が少し開いた状態となる。容器A1内において工程s1が実施されるとき、仕切り板J1a1は閉鎖位置にある。工程s1が完了すると、仕切り板J1a1が開き(即ち、接続管路J1が連通状態に切り替わり)、容器A1内の内容物が、重力によって次段の容器A2内へと移動する。また、図6(b)に示すように、容器A2内にも液体培地を収容しておくと、容器A1内の細胞Mが、重力によって緩やかに液体培地中を降下し、次段の容器A2内に入る。このとき、仕切り板J1a1は全開してもよいが、図6(b)のように、沈降した細胞を主として移動させる点では、全開の半分以下の開放、または、沈降した細胞が移動できる程度の開放が好ましい。これにより、容器A1内の液体培地と容器A2内の液体培地との混合を抑制することができる。このような細胞が主として移動する態様も、本発明では、内容物の移動に該当する。図6では、容器A1からA2への移動を例として示しているが、以降の容器に内容物を移動させる機構にも適用してよい。 The partition plate J1a1 can be stopped in a state where it is retracted into the main body of the shutter device J1a by any amount. When the partition plate J1a1 is retracted by a small amount, the connecting pipe is slightly open. When the process s1 is performed in the container A1, the partition plate J1a1 is in the closed position. When the process s1 is completed, the partition plate J1a1 opens (i.e., the connecting pipe J1 switches to a connected state), and the contents in the container A1 move by gravity into the next container A2. Also, as shown in FIG. 6(b), if a liquid culture medium is also contained in the container A2, the cells M in the container A1 will slowly descend in the liquid culture medium by gravity and enter the next container A2. At this time, the partition plate J1a1 may be fully opened, but in terms of mainly moving the settled cells, as shown in FIG. 6(b), it is preferable to open it less than halfway to the full opening, or to the extent that the settled cells can move. This makes it possible to prevent the liquid medium in container A1 from mixing with the liquid medium in container A2. In the present invention, this mode of mainly cell movement also corresponds to the movement of contents. In FIG. 6, the movement from container A1 to A2 is shown as an example, but the mechanism may also be applied to move contents to subsequent containers.
(vi)磁力を利用して細胞を移動させる機構
この機構は、磁性が付与されたマイクロキャリアと磁石とを用いて、細胞を移動させる機構である。磁性が付与されたマイクロキャリアに細胞を接着させ、該マイクロキャリアを、磁石を用いて磁力で牽引する。これにより、該マイクロキャリアとそれに接着した細胞を、次段の容器A2へと移動させることができる。図7の例では、図6(b)の例と同様に、容器A1と容器A2とが隣接しており、シャッター装置J1aの開閉可能な仕切り板J1a1が、容器A1と容器A2とを分断しまた連通させる。容器A2の底面は、容器A1の底面とよりも下方に位置する必要はない。図7の例では、容器A1、A2の各底面は互いに同じレベルにある1つの水平な面である。なお、細胞の移動に重力をも利用するならば、図6(b)の例と同様に、容器A1と容器A2の底面が1つの傾斜面を構成することが好ましい。
(vi) Mechanism for moving cells using magnetic force This mechanism uses a magnetic microcarrier and a magnet to move cells. Cells are attached to the magnetic microcarrier, and the microcarrier is pulled by magnetic force using a magnet. This allows the microcarrier and the cells attached thereto to be moved to the next vessel A2. In the example of FIG. 7, similar to the example of FIG. 6(b), the vessel A1 and the vessel A2 are adjacent to each other, and the openable and closable partition plate J1a1 of the shutter device J1a separates and communicates the vessel A1 and the vessel A2. The bottom surface of the vessel A2 does not need to be located lower than the bottom surface of the vessel A1. In the example of FIG. 7, the bottom surfaces of the vessels A1 and A2 are one horizontal surface at the same level. If gravity is also used to move the cells, it is preferable that the bottom surfaces of the vessel A1 and the vessel A2 form one inclined surface, similar to the example of FIG. 6(b).
容器A1内において工程s1が実施されるとき、仕切り板J1a1は閉鎖位置にある。工程s1が完了すると、図7のように、該仕切り板J1a1が適切な量だけ開き、容器A1と容器A2は互いに連通する。図7は、外部の磁石F1が移動しながら、細胞が付着したマイクロキャリアを容器A2へと牽引している状態を示している。図7に示すように、容器A2内にも液体培地を収容しておくと、仕切り板J1a1が開いても液体培地は激しく移動せず、よって、細胞が付着したマイクロキャリアM1は、磁力によって緩やかに液体培地中を移動し、次段の容器A2内に入る。このとき、容器A1内の液体培地の一部は、容器A2内の液体培地と混ざるが、容器A1内の液体培地全体が容器A2に移動することはない。図7では、容器A1からA2への移動を例として示しているが、以降の容器に内容物を移動させる機構にも適用してよい。 When step s1 is performed in container A1, the partition plate J1a1 is in the closed position. When step s1 is completed, as shown in FIG. 7, the partition plate J1a1 opens by an appropriate amount, and container A1 and container A2 communicate with each other. FIG. 7 shows a state in which the external magnet F1 moves and pulls the microcarrier with cells attached to it to container A2. As shown in FIG. 7, if liquid culture medium is also contained in container A2, the liquid culture medium does not move violently even when the partition plate J1a1 opens, and therefore the microcarrier M1 with cells attached moves gently through the liquid culture medium by magnetic force and enters the next container A2. At this time, part of the liquid culture medium in container A1 mixes with the liquid culture medium in container A2, but the entire liquid culture medium in container A1 does not move to container A2. FIG. 7 shows an example of movement from container A1 to A2, but it may also be applied to a mechanism for moving contents to a subsequent container.
(磁性が付与されたマイクロキャリア)
本発明に利用可能な、磁性が付与されたマイクロキャリアは、特に限定はされないが、例えば、自体公知のγFe2O3やFe3O4などの可磁化物質の表面をカルボキシデキストランや架橋アガロース等で被覆した磁性粒子に、アミノ基やカルボキシル基、ヒドロキシル基、エポキシ基などの官能基を導入し、これら官能基を介して細胞を認識する抗体などの生理活性物質を結合することで、標的細胞の表面抗原との吸着性を発現させているようなものでもよい。磁性を付与し得るマイクロキャリアの基材として、市販品としては、Corning Synthemaxビトロネクチン基質(Corning社)、Atelocollagen Microspheres(KOKEN社)等が例示される。市販品としては、Global Eukaryotic MicrocarriersTM(グローバル・セルソリューションズ社)などが例示される。
(Magnetic Microcarriers)
The magnetic microcarrier that can be used in the present invention is not particularly limited, but may be, for example, a magnetic particle in which the surface of a magnetizable material such as γFe 2 O 3 or Fe 3 O 4 known per se is coated with carboxydextran or crosslinked agarose, etc., and functional groups such as amino groups, carboxyl groups, hydroxyl groups, and epoxy groups are introduced, and a physiologically active substance such as an antibody that recognizes cells is bound via these functional groups, thereby expressing adsorptivity to surface antigens of target cells. Examples of commercially available substrates for microcarriers that can be magnetic include Corning Synthemax vitronectin substrate (Corning) and Atelocollagen Microspheres (KOKEN). Examples of commercially available products include Global Eukaryotic Microcarriers TM (Global Cell Solutions).
(磁石)
磁石は、永久磁石であっても電磁石であってもよい。電磁石は、必要な時だけ(とりわけ送り方向に移動するときだけ)磁界を発生させ得るので、磁性が付与されたマイクロキャリアの移動には好ましい。磁石の形状(特に容器に対面する面の外形や面積)、磁界の強度、移動速度などは、マイクロキャリアを牽引できるように、適宜に決定することができる。
磁石は、マイクロキャリアが容器から容器へと移動するように、容器から容器へと所定のコースを移動する。また、磁石を静止させ、容器A1、A2を移動させて、相対的に磁石を容器に対して移動させてもよい。磁石の移動は、手動であってもよいし、自動(駆動源による移動)であってもよい。
単一の磁石を移動させてもよいが、マイクロキャリアを移動させる方向に沿って複数の電磁石を配置し、電磁石を移動方向に順番に作動させることで、あたかも磁石が移動しているかのように磁界を移動(シフト)させ、それによりマイクロキャリアを牽引してもよい。
(magnet)
The magnet may be a permanent magnet or an electromagnet. An electromagnet is preferable for moving magnetic microcarriers because it can generate a magnetic field only when necessary (particularly when moving in the feed direction). The shape of the magnet (particularly the outer shape and area of the surface facing the container), the strength of the magnetic field, the moving speed, etc. can be appropriately determined so as to tow the microcarriers.
The magnet moves from vessel to vessel in a predetermined course so that the microcarriers move from vessel to vessel. Alternatively, the magnet may be stationary and the vessels A1 and A2 may be moved to move the magnet relatively to the vessels. The magnet may be moved manually or automatically (moved by a driving source).
Although a single magnet may be moved, multiple electromagnets may be arranged along the direction in which the microcarriers are moved, and the electromagnets may be activated in sequence in the direction of movement, shifting the magnetic field as if the magnet were moving, thereby attracting the microcarriers.
(本発明における細胞の移動)
細胞は液体培地やマイクロキャリアなどと共に細胞懸濁液として移動してもよいし、液体培地中を細胞が主として移動してもよいし、液体培地中をマイクロキャリアとそれに接着した細胞が主として移動してもよい。
(Cell migration in the present invention)
The cells may move as a cell suspension together with a liquid medium or microcarriers, or the cells may move primarily in the liquid medium, or the microcarriers and the cells attached thereto may move primarily in the liquid medium.
以下、当該製造方法(I)において、iPS細胞を製造するための処理や材料について詳細に説明する。 The processes and materials used to produce iPS cells in the production method (I) are described in detail below.
(本発明の製造方法(I))
本発明の人工多能性幹細胞の製造方法は、上述のように、以下の工程を有する。
工程s1:第1番目の容器A1内において、液体培地中で体細胞に初期化因子を接触させる工程。
工程s2:第2番目の容器A2から第(n-1)番目の容器A(n-1)内において、液体培地中の初期化因子の濃度を低減させる工程。
工程s3:第n番目の容器An内において、液体培地中で前記体細胞を培養して、iPS細胞を樹立する工程。
(Production method (I) of the present invention)
As described above, the method for producing induced pluripotent stem cells of the present invention comprises the following steps.
Step s1: A step of contacting somatic cells with reprogramming factors in a liquid medium in a first container A1.
Step s2: A step of reducing the concentration of the reprogramming factor in the liquid medium in the second container A2 to the (n-1)th container A(n-1).
Step s3: A step of culturing the somatic cells in a liquid medium in the n-th container An to establish iPS cells.
本発明の製造方法(I)における培養(典型的には、工程s1~工程s4)は、浮遊培養である。本明細書において、「浮遊培養」とは、細胞または細胞の凝集体が培養液に浮遊して存在する状態を維持する条件で行われる培養、即ち、細胞または細胞の凝集体と培養容器との間に強固な細胞-基質間結合(cell-substratum junction)を作らせない条件での培養を意味する。 The culture in the production method (I) of the present invention (typically, steps s1 to s4) is a suspension culture. In this specification, "suspension culture" means culture carried out under conditions that maintain the state in which cells or cell aggregates are suspended in the culture medium, that is, culture under conditions that do not allow the formation of strong cell-substratum junctions between the cells or cell aggregates and the culture vessel.
本明細書において、「人工多能性幹細胞(induced pluripotent stem cell:iPS細胞)」とは、哺乳動物体細胞または未分化幹細胞に、特定の因子(初期化因子)を導入してリプログラミングすることにより得られる細胞である。人工多能性幹細胞は、生体の種々の異なった形態や機能を持つ組織や細胞に分化でき、三胚葉(内胚葉、中胚葉、外胚葉)のどの系統の細胞にも分化し得る能力を有する。 In this specification, "induced pluripotent stem cells (iPS cells)" are cells obtained by reprogramming mammalian somatic cells or undifferentiated stem cells through the introduction of specific factors (reprogramming factors). Induced pluripotent stem cells can differentiate into tissues and cells with various different forms and functions in the body, and have the ability to differentiate into cells of any lineage of the three germ layers (endoderm, mesoderm, and ectoderm).
現在、人工多能性幹細胞にはさまざまなものがあり、山中らにより、ヒト線維芽細胞にOct3/4、Sox2、Klf4、及びc-Mycの4因子を導入することにより、樹立されたヒトiPSC(Takahashi K, Yamanaka S., et al. Cell, (2007) 131: 861-872.)、上記4因子導入後、Nanogの発現を指標として選別し、樹立したNanog-iPSC(Okita, K., Ichisaka, T., and Yamanaka, S. (2007). Nature 448, 313-317.)、c-Mycを含まない方法で作製されたiPS細胞(Nakagawa M, Yamanaka S., et al. Nature Biotechnology, (2008) 26, 101 - 106)、ウイルスフリー法で6因子を導入して樹立されたiPS細胞(Okita K et al. Nat. Methods 2011 May;8(5):409-12, Okita K et al. Stem Cells. 31(3):458-66.)等も用いることができる。また、Thomsonらにより作製されたOCT3/4、SOX2、NANOG、及びLIN28の4因子を導入して樹立されたiPS細胞(Yu J., Thomson JA. et al., Science (2007) 318: 1917-1920.)、Daleyらにより作製されたiPS細胞(Park IH, Daley GQ. et al., Nature (2007) 451: 141-146)、桜田らにより作製されたiPS細胞(特開2008-307007号)等も用いることができる。
このほか、公開されているすべての論文(例えば、Shi Y., Ding S., et al., Cell Stem Cell, (2008) Vol3, Issue 5,568-574;、Kim JB., Scholer HR., et al., Nature, (2008) 454, 646-650;Huangfu D., Melton, DA., et al., Nature Biotechnology, (2008) 26, No 7, 795-797)、あるいは特許(例えば、特開2008-307007号、特開2008-283972号、US2008/2336610、US2009/047263、WO2007/069666、WO2008/118220、WO2008/124133、WO2008/151058、WO2009/006930、WO2009/006997、WO2009/007852)に記載されている当該分野で公知の人工多能性幹細胞のいずれも用いることができる。
Currently, there are various types of induced pluripotent stem cells, including human iPSCs established by Yamanaka et al. by introducing four factors, Oct3/4, Sox2, Klf4, and c-Myc, into human fibroblasts (Takahashi K, Yamanaka S., et al. Cell, (2007) 131: 861-872.), Nanog-iPSCs established by selecting using the expression of Nanog as an indicator after the introduction of the above four factors (Okita, K., Ichisaka, T., and Yamanaka, S. (2007). Nature 448, 313-317.), iPS cells produced by a method that does not contain c-Myc (Nakagawa M, Yamanaka S., et al. Nature Biotechnology, (2008) 26, 101 - 106), and iPS cells established by introducing six factors using a virus-free method (Okita K et al. Nat. Methods 2011 May;8(5):409-12, Okita K et al. Stem Cells. 31(3):458-66.) and the like can also be used. In addition, iPS cells established by introducing four factors, OCT3/4, SOX2, NANOG, and LIN28, produced by Thomson et al. (Yu J., Thomson JA. et al., Science (2007) 318: 1917-1920.), iPS cells produced by Daley et al. (Park IH, Daley GQ. et al., Nature (2007) 451: 141-146), iPS cells produced by Sakurada et al. (JP Patent Publication No. 2008-307007), and the like can also be used.
In addition, all published papers (e.g., Shi Y., Ding S., et al., Cell Stem Cell, (2008) Vol3, Issue 5,568-574; Kim JB., Scholer HR., et al., Nature, (2008) 454, 646-650; Huangfu D., Melton, DA., et al., Nature Biotechnology, (2008) 26,
人工多能性幹細胞株としては、NIH、理研、京都大学等が樹立した各種iPSC株が利用可能である。例えば、ヒトiPSC株であれば、理研のHiPS-RIKEN-1A株、HiPS-RIKEN-2A株、HiPS-RIKEN-12A株、Nips-B2株等、京都大学のAdiPS細胞、253G1株、253G4株、1201C1株、1205D1株、1210B2株、1383D2株、1383D6株、201B7株、409B2株、454E2株、585A1株、585B2株、606A1株、610B1株、648A1株、1231A3株、FfI-01s04株等が挙げられる。 As induced pluripotent stem cell lines, various iPSC lines established by NIH, RIKEN, Kyoto University, etc. can be used. For example, human iPSC lines include RIKEN's HiPS-RIKEN-1A line, HiPS-RIKEN-2A line, HiPS-RIKEN-12A line, Nips-B2 line, etc., and Kyoto University's AdiPS cells, 253G1 line, 253G4 line, 1201C1 line, 1205D1 line, 1210B2 line, 1383D2 line, 1383D6 line, 201B7 line, 409B2 line, 454E2 line, 585A1 line, 585B2 line, 606A1 line, 610B1 line, 648A1 line, 1231A3 line, FfI-01s04 line, etc.
本明細書において、人工多能性幹細胞は、遺伝性疾患の患者由来の細胞であってもよい。遺伝性疾患の患者由来の多能性幹細胞から分化誘導させた細胞は、該疾患の病態を反映する疾患モデルとなり得るため、該疾患の治療または予防薬のスクリーニングなどに適している。あるいは、遺伝性疾患の患者由来の多能性幹細胞に対して、CRISPR-Casシステムなどを用いたゲノム編集により遺伝子を修復した上で目的の細胞へと分化させることで、該細胞を該疾患の治療薬として用いることも可能となる。 In this specification, the induced pluripotent stem cells may be cells derived from a patient with a genetic disease. Cells induced to differentiate from pluripotent stem cells derived from a patient with a genetic disease can serve as a disease model that reflects the pathology of the disease, and are therefore suitable for screening therapeutic or preventive drugs for the disease. Alternatively, pluripotent stem cells derived from a patient with a genetic disease can be genetically repaired by genome editing using the CRISPR-Cas system or the like, and then differentiated into the desired cells, making it possible to use the cells as a therapeutic drug for the disease.
本明細書において、「体細胞」とは、動物を構成する細胞のうち、生殖細胞以外の細胞を意味する。体細胞としては、特に限定されないが、胎児(仔)の体細胞、新生児(仔)の体細胞、及び成熟した健全な若しくは疾患性の体細胞のいずれも包含されるし、また、初代培養細胞、継代細胞、および株化細胞のいずれも包含される。具体的には、体細胞は、例えば、浮遊性細胞(例:血球系細胞等)であっても接着性細胞であってもよいが、好ましくは浮遊性細胞である。本発明の製造方法で用いる体細胞としては、例えば、皮膚等の線維芽細胞、皮膚細胞、視覚細胞、脳細胞、有毛細胞、口腔粘膜、肺細胞、肝細胞、胃粘膜細胞、腸細胞、脾細胞、膵細胞、腎細胞、神経幹細胞、智歯などに由来する間葉系幹細胞、組織幹細胞、組織前駆細胞、血球系細胞(例:造血幹細胞、末梢血単核球(PBMC)(T細胞および非T細胞を含む)、臍帯血細胞等)、上皮細胞、内皮細胞(例:血管内皮細胞)、筋肉細胞などが挙げられるが、これらに限定されない。 As used herein, "somatic cells" refers to cells other than reproductive cells that make up an animal. Somatic cells are not particularly limited, but include fetal (offspring) somatic cells, neonatal (offspring) somatic cells, and mature healthy or diseased somatic cells, as well as primary culture cells, passaged cells, and established cell lines. Specifically, somatic cells may be, for example, floating cells (e.g., blood cells, etc.) or adhesive cells, with floating cells being preferred. Somatic cells used in the manufacturing method of the present invention include, but are not limited to, for example, fibroblasts such as skin cells, skin cells, visual cells, brain cells, hair cells, oral mucosa, lung cells, liver cells, gastric mucosa cells, intestinal cells, spleen cells, pancreatic cells, kidney cells, neural stem cells, mesenchymal stem cells derived from wisdom teeth, etc., tissue stem cells, tissue progenitor cells, blood cells (e.g., hematopoietic stem cells, peripheral blood mononuclear cells (PBMCs) (including T cells and non-T cells), umbilical cord blood cells, etc.), epithelial cells, endothelial cells (e.g., vascular endothelial cells), muscle cells, etc.
一態様では、体細胞として、血球系細胞(例えば、末梢血単核球)を用いる場合、該細胞は、全血を遠心分離(密度勾配遠心分離)することによって得られる。このような場合、遠心分離機内にセットされる遠沈管として、シリンジ機構を有するものを用いることによって、また、遠心分離で得られる所定の分画を、閉鎖系を維持した状態で取り出すことができる遠心分離機を用いることによって、遠心分離で得られる分画層である末梢血単核球を含む層を、外気に接触させることなしに、第1番目の容器A1に直接的に移動させることができる。 In one embodiment, when blood cells (e.g., peripheral blood mononuclear cells) are used as somatic cells, the cells are obtained by centrifuging whole blood (density gradient centrifugation). In such a case, by using a centrifuge tube with a syringe mechanism as the settling tube set in the centrifuge, and by using a centrifuge that can extract a specific fraction obtained by centrifugation while maintaining a closed system, the layer containing peripheral blood mononuclear cells, which is the fraction layer obtained by centrifugation, can be moved directly to the first container A1 without coming into contact with the outside air.
本明細書において、体細胞の由来種も特に限定されず、例えば、ラット、マウス、ハムスター、モルモット等のげっ歯類、ウサギ等のウサギ目、ブタ、ウシ、ヤギ、ヒツジ等の有蹄目、イヌ、ネコ等のネコ目、メガネザル、オナガザル、カニクイザル、アカゲザル、ニホンザル、テナガザル、クモザル、オマキザル、オラウータン、ゴリラ、チンパンジー、ヒト(いずれも、直鼻亜目)等や、キツネザル、アイアイ、ロリス(いずれも、曲鼻亜目)等の霊長類などの細胞であってよい。体細胞の由来種として、好ましくはヒトである。 In the present specification, the species of origin of the somatic cells is not particularly limited, and may be, for example, cells from rodents such as rats, mice, hamsters, guinea pigs, etc.; lagomorphs such as rabbits; ungulates such as pigs, cows, goats, sheep, etc.; carnivores such as dogs and cats; tarsiers, long-tailed macaques, cynomolgus monkeys, rhesus monkeys, Japanese macaques, gibbons, spider monkeys, capuchin monkeys, orangutans, gorillas, chimpanzees, humans (all in the Stratorhinidae suborder), etc.; and primates such as lemurs, aye-ayes, and lorises (all in the Strangorhinidae suborder). Humans are preferred as the species of origin of the somatic cells.
本明細書において、特に断りのない限り、「細胞」には、「細胞集団」が含まれるものとする。細胞集団は、1種類の細胞から構成されていてもよく、2種類以上の細胞から構成されていてもよい。 In this specification, unless otherwise specified, "cells" includes "cell populations." A cell population may be composed of one type of cell, or may be composed of two or more types of cells.
本明細書において、「初期化因子」としては、例えば、Oct3/4、Sox2、Sox1、Sox3、Sox15、Sox17、Klf4、Klf2、c-Myc、N-Myc、L-Myc、Nanog、Lin28、Fbx15、ERas、ECAT15-2、Tcl1、beta-catenin、Lin28b、Sall1、Sall4、ESrrb、Nr5a2、Tbx3またはGlis1等が例示され、これらの初期化因子は、単独で用いても良く、組み合わせて用いても良い。初期化因子の組み合わせとしては、WO2007/069666、WO2008/118820、WO2009/007852、WO2009/032194、WO2009/058413、WO2009/057831、WO2009/075119、WO2009/079007、WO2009/091659、WO2009/101084、WO2009/101407、WO2009/102983、WO2009/114949、WO2009/117439、WO2009/126250、WO2009/126251、WO2009/126655、WO2009/157593、WO2010/009015、WO2010/033906、WO2010/033920、WO2010/042800、WO2010/050626、WO2010/056831、WO2010/068955、WO2010/098419、WO2010/102267、WO2010/111409、WO2010/111422、WO2010/115050、WO2010/124290、WO2010/147395、WO2010/147612、Nat Biotechnol,2008.26.795-797、Cell Stem Cell,2008.2.525-528、Stem Cells,2008.26.2467-2474、Nat Biotechnol,2008.26.1269-1275、Cell Stem Cell,2008.3.568-574、Cell Stem Cell,2008.3.475-479、Cell Stem Cell,2008.3.132-135、Nat Cell Biol,2009.11.197-203、Nat Biotechnol,2009.27.459-461、Proc Natl Acad Sci USA,2009.106.8912-8917、Nature,2009.461.643-649、Cell Stem Cell,2009.5.491-503、Cell Stem Cell,2010.6.167-74、Nature,2010.463.1096-1100、Stem Cells,2010.28.713-720、Nature,2011.474.225-229に記載の組み合わせが例示される。 In this specification, examples of "reprogramming factors" include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, ESrrb, Nr5a2, Tbx3, and Glis1, and these reprogramming factors may be used alone or in combination. Combinations of reprogramming factors include those described in WO2007/069666, WO2008/118820, WO2009/007852, WO2009/032194, WO2009/058413, WO2009/057831, WO2009/075119, WO2009/079007, WO2009/091659, WO2009/101084, WO2009/101407, WO2009/102983, WO2009/114949, WO2009/117439, WO2009/126250, WO2009/126251, WO 2009/126655, WO2009/157593, WO2010/009015, WO2010/033906, WO2010/033920, WO2010/042800, WO2010/050626, WO2010/056831, WO2010/0689 55, WO2010/098419, WO2010/102267, WO2010/111409, WO2010/111422, WO2010/115050, WO2010/124290, WO2010/147395, WO2010/147612, Nat Bio technol,2008.26.795-797, Cell Stem Cell,2008.2.525-528, Stem Cells,2008.26.2467-2474, Nat Biotechnol,2008.26.1269-1275, Cell Stem Cell,2008.3.568-574, Cell Stem Cell,2008.3.475-479, Cell Stem Cell,2008.3.132-135, Nat Cell Biol,2009.11.197-203, Nat Bi Examples of combinations include those described in Cell Stem Cell, 2009.27.459-461, Proc Natl Acad Sci USA, 2009.106.8912-8917, Nature, 2009.461.643-649, Cell Stem Cell, 2009.5.491-503, Cell Stem Cell, 2010.6.167-74, Nature, 2010.463.1096-1100, Stem Cells, 2010.28.713-720, and Nature, 2011.474.225-229.
初期化因子の組み合わせとしては、例えば、以下:
(1)Oct3/4、Klf4、Sox2、c-Myc
(ここで、Sox2は、Sox1、Sox3、Sox15、Sox17またはSox18で置換可能である。また、Klf4は、Klf1、Klf2またはKlf5で置換可能である。さらに、c-Mycは、T58A(活性型変異体)、N-Myc、L-Mycで置換可能である。特に、臨床用の場合、L-Mycが好ましい。)
(2)Oct3/4、Klf4、Sox2
(3)Oct3/4、Klf4、c-Myc
(4)Oct3/4、Sox2、Nanog、Lin28
(5)Oct3/4、Klf4、c-Myc、Sox2、Nanog、Lin28
(6)Oct3/4、Klf4、Sox2、bFGF
(7)Oct3/4、Klf4、Sox2、SCF
(8)Oct3/4、Klf4、c-Myc、Sox2、bFGF
(9)Oct3/4、Klf4、c-Myc、Sox2、SCF
が挙げられる。
Combinations of reprogramming factors include, for example, the following:
(1) Oct3/4, Klf4, Sox2, c-Myc
(Here, Sox2 can be replaced by Sox1, Sox3, Sox15, Sox17 or Sox18. Klf4 can be replaced by Klf1, Klf2 or Klf5. Furthermore, c-Myc can be replaced by T58A (active mutant), N-Myc or L-Myc. In particular, L-Myc is preferred for clinical use.)
(2) Oct3/4, Klf4, Sox2
(3) Oct3/4, Klf4, c-Myc
(4) Oct3/4, Sox2, Nanog, Lin28
(5) Oct3/4, Klf4, c-Myc, Sox2, Nanog, Lin28
(6) Oct3/4, Klf4, Sox2, bFGF
(7) Oct3/4, Klf4, Sox2, SCF
(8) Oct3/4, Klf4, c-Myc, Sox2, bFGF
(9) Oct3/4, Klf4, c-Myc, Sox2, SCF
Examples include:
本発明の工程s1では、第1番目の容器A1内において、液体培地中で体細胞に初期化因子を接触させることで、該初期化因子を体細胞内に導入する。体細胞に導入する初期化因子は、タンパク質の形態であってもよく、該タンパク質をコードする核酸(RNAまたはDNA)や該核酸を含む発現ベクターの形態であってもよい。初期化因子をRNAの形態で導入する場合には、細胞に導入された免疫原性のRNAにより細胞防御機構の活性化が生じ得るため、該防御機構を回避するためのRNA(例えば、ワクシニアウイルス由来のE3 mRNA、K3L mRNA、B18 mRNA、これらのmRNAの組合せ等)を体細胞に導入してもよい。また、多能性幹細胞の樹立効率向上のため、各種miRNAまたはそのミミック(例えば、miR-302a-3pまたはそのミミック、miR-302b-3pまたはそのミミック、miR-302c-3pまたはそのミミック、miR-302d-3pまたはそのミミック、miR-367-3pまたはそのミミック、これらのmiRNAまたはそのミミックの組合せ等)を体細胞に導入してもよい。 In step s1 of the present invention, the reprogramming factor is introduced into the somatic cells by contacting the reprogramming factor with the somatic cells in a liquid medium in a first container A1. The reprogramming factor introduced into the somatic cells may be in the form of a protein, or in the form of a nucleic acid (RNA or DNA) encoding the protein or an expression vector containing the nucleic acid. When the reprogramming factor is introduced in the form of RNA, the immunogenic RNA introduced into the cells may activate the cell's defense mechanism, so RNA to circumvent the defense mechanism (e.g., E3 mRNA, K3L mRNA, B18 mRNA derived from vaccinia virus, a combination of these mRNAs, etc.) may be introduced into the somatic cells. In addition, to improve the efficiency of establishing pluripotent stem cells, various miRNAs or mimics thereof (e.g., miR-302a-3p or mimic thereof, miR-302b-3p or mimic thereof, miR-302c-3p or mimic thereof, miR-302d-3p or mimic thereof, miR-367-3p or mimic thereof, combinations of these miRNAs or mimics, etc.) may be introduced into somatic cells.
上述するようなmiRNAを体細胞に導入する場合には、天然型のmiRNAの形態でもよく、核酸に化学修飾が付されたmiRNAでもよく、miRNAミミックでもよく、あるいはmiRNAをコードするDNAや該DNAを含む発現ベクターの形態であってもよいが、好ましくはmiRNAミミックである。miRNAミミックは、二本鎖RNAの形態であり、典型的には、天然型RNAからなるガイド鎖と、化学修飾(例えば、2‘-Oメチル修飾)が付されたパッセンジャー鎖で構成されており、ガイド鎖はRNAi活性を示す一方でパッセンジャー鎖はRNAi活性を示さないため、細胞内の天然型のmiRNAを模倣することとなる。 When the miRNA described above is introduced into somatic cells, it may be in the form of natural miRNA, miRNA with chemical modification of nucleic acid, miRNA mimic, or DNA encoding miRNA or an expression vector containing said DNA, but is preferably a miRNA mimic. A miRNA mimic is in the form of double-stranded RNA, and is typically composed of a guide strand made of natural RNA and a passenger strand with chemical modification (e.g., 2'-O methyl modification). The guide strand exhibits RNAi activity, while the passenger strand does not exhibit RNAi activity, so it mimics natural miRNA in cells.
発現ベクターとしては、例えば、レトロウイルス、レンチウイルス、アデノウイルス、アデノ随伴ウイルス、ヘルペスウイルス、センダイウイルスなどのウイルスベクター、プラスミドベクター、エピソーマルベクター、人工染色体ベクター、トランスポゾンベクター(piggyBac,piggyBat,TolII)などが挙げられる。発現ベクターにおいて使用されるプロモーターとしては、例えば、EF1αプロモーター、ACTBプロモーター、UbqCプロモーター、PGKプロモーター、CAGプロモーター、SRαプロモーター、SV40プロモーター、LTRプロモーター、CMV(サイトメガロウイルス)プロモーター、RSV(ラウス肉腫ウイルス)プロモーター、MoMuLV(モロニーマウス白血病ウイルス)LTR、HIV LTR、HSV-TK(単純ヘルペスウイルスチミジンキナーゼ)プロモーターなどが用いられる。一方で、miRNAをコードするDNAを含む発現ベクターにおいては、プロモーターとしては、pol III系のプロモーター(例、SNR6、SNR52、SCR1、RPR1、U3、U6、H1プロモーター等)が好ましい。 Expression vectors include, for example, viral vectors such as retrovirus, lentivirus, adenovirus, adeno-associated virus, herpes virus, and Sendai virus, as well as plasmid vectors, episomal vectors, artificial chromosome vectors, and transposon vectors (piggyBac, piggyBat, TolII). Promoters used in expression vectors include, for example, EF1α promoter, ACTB promoter, UbqC promoter, PGK promoter, CAG promoter, SRα promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (moloney murine leukemia virus) LTR, HIV LTR, and HSV-TK (herpes simplex virus thymidine kinase) promoter. On the other hand, in an expression vector containing DNA encoding miRNA, the promoter is preferably a pol III promoter (e.g., SNR6, SNR52, SCR1, RPR1, U3, U6, H1 promoter, etc.).
mRNAやmiRNAなどのRNAは、化学合成により作製してもよく、またインビトロ転写反応(IVT)法により作製することもできる。あるいは、生物内(大腸菌や哺乳類培養細胞)で発現させたものを精製することもできる。化学合成方法としては、例えば、ヌクレオシドホスホロアミダイトと固相担体を用いた方法などが挙げられる。また、化学修飾が導入されたヌクレオシドホスホロアミダイト(例えば、糖部の2’-O-メチル化ホスホロアミダイトなど)を用いることで、RNA配列の任意の塩基部位に修飾ヌクレオチドを導入することができる。 RNA such as mRNA and miRNA can be produced by chemical synthesis or by in vitro transcription (IVT). Alternatively, it can be expressed in an organism (Escherichia coli or cultured mammalian cells) and then purified. Chemical synthesis methods include, for example, methods using nucleoside phosphoramidites and solid-phase supports. In addition, modified nucleotides can be introduced into any base site in an RNA sequence by using chemically modified nucleoside phosphoramidites (for example, 2'-O-methylated phosphoramidites in the sugar moiety).
核酸(例えば、上述のmRNA、miRNA、これらをコードするDNA等)、該核酸を含む発現ベクターまたはタンパク質(例えば、初期化因子)の細胞への導入は、公知の種々の方法によって行うことができる。かかる方法として、例えば、リン酸カルシウム仲介性トランスフェクション、エレクトロポレーション、リポソームトランスフェクション、リポフェクション、遺伝子銃、マイクロインジェクション、ウイルスベクター法、ウイルス様粒子法、アグロバクテリウム法、アグロインフィルトレーション法、PEG-カルシウム法、ソノポレーション法、脂質ナノ粒子法などが挙げられる。 Nucleic acids (e.g., the above-mentioned mRNA, miRNA, DNA encoding these, etc.), expression vectors containing the nucleic acids, or proteins (e.g., reprogramming factors) can be introduced into cells by various known methods. Examples of such methods include calcium phosphate-mediated transfection, electroporation, liposome transfection, lipofection, gene gun, microinjection, viral vector method, virus-like particle method, Agrobacterium method, agroinfiltration method, PEG-calcium method, sonoporation method, lipid nanoparticle method, etc.
本発明の製造方法(I)における培養(典型的に、工程s1~工程s4)は、全部または一部の期間、フィーダーフリー条件下および/またはゼノフリー条件下での培養であってもよい。臨床での使用の観点からは、本発明の製造方法は、全期間がフィーダーフリーかつゼノフリー条件下で行われることが好ましい。本明細書において、「フィーダーフリー」とは、培養対象の細胞の培養条件を整えるために用いる、補助役を果たす他の細胞種(即ち、フィーダー細胞)を含まない培地または培養条件を意味する。また、「ゼノフリー」とは、培養対象の細胞の生物種とは異なる生物由来の成分を含まない培地または培養条件を意味する。 The culture in the production method (I) of the present invention (typically, steps s1 to s4) may be cultured under feeder-free conditions and/or xeno-free conditions for all or part of the period. From the viewpoint of clinical use, it is preferable that the production method of the present invention is carried out under feeder-free and xeno-free conditions for the entire period. In this specification, "feeder-free" means a medium or culture conditions that do not contain other cell types (i.e., feeder cells) that play a supporting role and are used to prepare the culture conditions for the cells to be cultured. In addition, "xeno-free" means a medium or culture conditions that do not contain components derived from organisms different from the organism species of the cells to be cultured.
本発明の製造方法(I)で用いる基礎培地としては、特に限定されないが、Essential8培地(CTSTM Essential 8TM Medium、Essential 8TM Medium、Essential 8TM Flex Medium、Essential 6TM Medium)(サーモフィッシャーサイエンティフィック社)、StemFit(登録商標)AK02培地(味の素株式会社)、StemFit(登録商標)AK03培地(味の素株式会社)、StemFit(登録商標)Basic03培地、CTS(登録商標)KnockOut SR XenoFree Medium(Gibco)、mTeSR1培地、TeSR1培地(Stem Cell Technologies)、Iscove‘s modified Dulbecco’s medium(GEヘルスケア社)、Improved MEM(サーモフィッシャーサイエンティフィック社)などが挙げられる。これらの培地は、フィーダーフリーかつゼノフリー条件下での培養に用いることもできる。また、RPMI-1640培地、EagleMEM(EMEM)、ダルベッコ改変MEM、Glasgow’s MEM(GMEM)、α-MEM、199培地、IMDM、DMEM、Hybridoma Serum free培地、KnockOutTM DMEM、AdvancedTM培地(例:Advanced MEM、Advanced RPMI、Advanced DMEM/F-12)、Ham’s Medium F-12、Ham’s Medium F-10、Ham’s Medium F12K、DMEM/F-12、ATCC-CRCM30、DM-160、DM-201、BME、Fischer、McCoy’s 5A、Leibovitz’s L-15、RITC80-7、MCDB105、MCDB107、MCDB131、MCDB153、MCDB201、NCTC109、NCTC135、Waymouth’s Medium(例:Waymouth’s MB752/1)、CMRL培地(例:CMRL-1066)、Williams’mediumE、Brinster’s BMOC-3 Medium、E8 Medium、StemPro 34、MesenPRO RS(以上サーモフィッシャーサイエンティフィック社)、ReproFF2、Primate ES Cell Medium、ReproStem(以上リプロセル株式会社)、ProculAD(ロート製薬株式会社)、MSCBM-CD、MSCGM-CD(以上Lonza社)、EX-CELL302倍地(SAFC社)またはEX-CELL-CD-CHO(SAFC社)、ReproMedTM iPSC Medium(リプロセル株式会社)およびこれらの混合物などが挙げられるが、これらに限定されない。
The basal medium used in the production method (I) of the present invention is not particularly limited, and examples thereof include Essential 8 medium (CTS ™ Essential 8 ™ Medium, Essential 8 ™ Medium, Essential 8 ™ Flex Medium, Essential 6 ™ Medium) (Thermo Fisher Scientific), StemFit (registered trademark) AK02 medium (Ajinomoto Co., Inc.), StemFit (registered trademark) AK03 medium (Ajinomoto Co., Inc.), StemFit (registered trademark) Basic03 medium, CTS (registered trademark) KnockOut SR XenoFree Medium (Gibco), mTeSR1 medium, TeSR1 medium (Stem Cell Technologies), Iscove's modified Dulbecco's Examples of suitable media include Improved MEM (GE Healthcare), and Improved MEM (Thermo Fisher Scientific). These media can also be used for culture under feeder-free and xeno-free conditions. In addition, RPMI-1640 medium, Eagle MEM (EMEM), Dulbecco's modified MEM, Glasgow's MEM (GMEM), α-MEM, 199 medium, IMDM, DMEM, Hybridoma Serum free medium, KnockOut ™ DMEM, Advanced ™ medium (e.g., Advanced MEM, Advanced RPMI, Advanced DMEM/F-12), Ham's Medium F-12, Ham's Medium F-10, Ham's Medium F12K, DMEM/F-12, ATCC-CRCM30, DM-160, DM-201, BME, Fischer, McCoy's 5A, Leibovitz's L-15, RITC80-7, MCDB105, MCDB107, MCDB131,
本発明の製造方法(I)で用いる基礎培地には、必要に応じて細胞の生存または増殖に必要な生理活性物質および栄養因子などを添加できる。これらの添加物は、培地に予め添加されていてもよく、細胞培養中に添加されてもよい。培養中に添加する方法は、1溶液または2種以上の混合溶液などいかなる形態によってでもよく、連続的または断続的な添加であってもよい。 The basal medium used in the production method (I) of the present invention can be supplemented with physiologically active substances and nutritional factors necessary for cell survival or proliferation, if necessary. These additives may be added to the medium in advance, or may be added during cell culture. The method of adding them during culture may be in any form, such as one solution or a mixed solution of two or more types, and may be continuous or intermittent addition.
生理活性物質としては、インシュリン、IGF-1、トランスフェリン、アルブミン、補酵素Q10、各種サイトカイン(インターロイキン類(IL-2、IL-7、IL-15等)、幹細胞因子(SCF)、アクチビン等)、各種ホルモン、各種増殖因子(白血病抑制因子(LIF)、塩基性線維芽細胞増殖因子(bFGF)、TGF-β等)などが挙げられる。
栄養因子としては、糖、アミノ酸、ビタミン、加水分解物または脂質などが挙げられる。
糖としては、グルコース、マンノースまたはフルクトースなどが挙げられ、1種または2種以上を組み合わせて用いられる。
アミノ酸としては、L-アラニン、L-アルギニン、L-アスパラギン、L-アスパラギン酸、L-システイン、L-グルタミン酸、L-グルタミン、グリシン、L-ヒスチジン、L-イソロイシン、L-ロイシン、L-リジン、L-メチオニン、L-フェニルアラニン、L-プロリン、L-セリン、L-スレオニン、L-トリプトファン、L-チロシンまたはL-バリンなどが挙げられ、1種または2種以上を組み合わせて用いられる。
ビタミンとしては、d-ビオチン、D-パントテン酸、コリン、葉酸、myo-イノシトール、ナイアシンアミド、ピロドキサール、リボフラビン、チアミン、シアノコバラミンまたはDL-α―トコフェロールなどが挙げられ、1種または2種以上を組み合わせて用いられる。
加水分解物としては、大豆、小麦、米、えんどう豆、とうもろこし、綿実、酵母抽出物などを加水分解したものが挙げられる。
脂質としては、コレステロール、リノール酸またはリノレイン酸などが挙げられる。
Examples of physiologically active substances include insulin, IGF-1, transferrin, albumin, coenzyme Q10, various cytokines (interleukins (IL-2, IL-7, IL-15, etc.), stem cell factor (SCF), activin, etc.), various hormones, and various growth factors (leukemia inhibitory factor (LIF), basic fibroblast growth factor (bFGF), TGF-β, etc.).
Nutritional factors include sugars, amino acids, vitamins, hydrolysates, lipids, and the like.
Examples of the sugar include glucose, mannose, fructose, and the like, and one or more of them may be used in combination.
Examples of amino acids include L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, and L-valine, and these may be used alone or in combination of two or more.
Examples of vitamins include d-biotin, D-pantothenic acid, choline, folic acid, myo-inositol, niacinamide, pyrodoxal, riboflavin, thiamine, cyanocobalamin, and DL-α-tocopherol, and these may be used alone or in combination of two or more kinds.
Hydrolysates include those derived from hydrolyzed soybeans, wheat, rice, peas, corn, cottonseed, yeast extracts, and the like.
The lipids include cholesterol, linoleic acid, and linolenic acid.
さらに、培地には、カナマイシン、ストレプトマイシン、ペニシリンまたはハイグロマイシンなどの抗生物質を必要に応じて添加してもよい。シアル酸等の酸性物質を培地に添加する場合には、培地のpHを細胞の成育に適した中性域であるpH5~9、好ましくはpH6~8に調整することが望ましい。
Furthermore, antibiotics such as kanamycin, streptomycin, penicillin, or hygromycin may be added to the medium as necessary. When an acidic substance such as sialic acid is added to the medium, it is desirable to adjust the pH of the medium to a neutral range suitable for cell growth, between
本明細書において、培地は、血清(例えば、ウシ胎児血清(FBS)、ヒト血清、ウマ血清)含有培地であっても無血清培地であってもよい。異種動物由来成分の混入防止の観点からは血清を含有しないか、培養される細胞と同種動物由来の血清が用いられることが好ましい。ここで、無血清培地とは、無調整または未精製の血清を含まない培地を意味する。無血清培地は、精製された血液由来成分や動物組織由来成分(例えば、増殖因子)を含有していてもよい。 In this specification, the medium may be a serum-containing medium (e.g., fetal bovine serum (FBS), human serum, horse serum) or a serum-free medium. From the viewpoint of preventing contamination with components derived from different kinds of animals, it is preferable that the medium does not contain serum, or that serum derived from the same kind of animal as the cells to be cultured is used. Here, serum-free medium means a medium that does not contain unconditioned or unpurified serum. The serum-free medium may contain purified blood-derived components or animal tissue-derived components (e.g., growth factors).
本明細書において、培地は、血清と同様に、血清代替物についてもこれを含んでいても含んでいなくともよい。血清代替物としては、例えば、アルブミン、脂質リッチアルブミンおよび組換えアルブミン等のアルブミン代替物、植物デンプン、デキストラン、タンパク質加水分解物、トランスフェリンまたは他の鉄輸送体、脂肪酸、インスリン、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオグリセロールあるいはこれらの均等物などが挙げられる。血清代替物の具体例として、例えば、WO98/30679記載の方法により調製されるものや、市販のknockout Serum Replacement[KSR](Life Technologies社)、Chemically-defined Lipid concentrated(Life Technologies社)およびGlutamax(Life Technologies社)などが挙げられる。また、生体由来因子としては、多血小板血漿(PRP)、ヒト間葉系幹細胞の培養上清成分などが挙げられる。 As used herein, the medium may or may not contain serum substitutes, as well as serum. Serum substitutes include, for example, albumin substitutes such as albumin, lipid-rich albumin, and recombinant albumin, vegetable starch, dextran, protein hydrolysates, transferrin or other iron transporters, fatty acids, insulin, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thioglycerol, or equivalents thereof. Specific examples of serum substitutes include those prepared by the method described in WO98/30679, as well as commercially available products such as Knockout Serum Replacement [KSR] (Life Technologies), Chemically-defined Lipid Concentrated (Life Technologies), and Glutamax (Life Technologies). Examples of biologically derived factors include platelet-rich plasma (PRP) and culture supernatant components of human mesenchymal stem cells.
本明細書において、培地は、細胞の浮遊培養に用いる足場材料(以下、「スキャフォールド」ともいう)を含んでもよく、該足場材料は、細胞培養において細胞の足場として機能する材料または基材を意味する。足場材料は、上述のように細胞の浮遊培養に用いることができれば(換言すれば、培地中に遊離しても良い)特に制限されないが、合成樹脂を含有するあるいは合成樹脂からなるものや、コラーゲン等の柔軟性素材からなるものが例示される。また、一例として、足場材料は、アテロコラーゲンを含むものや、アテロコラーゲンからなるものであってもよく、具体的には、アテロコラーゲンを足場材料として適した形状に成形したものが挙げられる。典型的には、足場材料は、ナノファイバー以外の材料である。 In this specification, the medium may contain a scaffold material (hereinafter also referred to as "scaffold") used for suspension culture of cells, and the scaffold material refers to a material or substrate that functions as a scaffold for cells in cell culture. The scaffold material is not particularly limited as long as it can be used for suspension culture of cells as described above (in other words, it may be free in the medium), but examples include those that contain or are made of synthetic resin, and those that are made of flexible materials such as collagen. As an example, the scaffold material may contain or be made of atelocollagen, and specifically, atelocollagen molded into a shape suitable for use as a scaffold material. Typically, the scaffold material is a material other than nanofibers.
合成樹脂は、重合性モノマー(以下、単に「モノマー」ともいう)を重合(重縮合も含む)して得られるポリマー(以下、単に「ポリマー」ともいう)を主成分とするものを意味する。上記ポリマーは一種または二種以上の重合性モノマーのコポリマーも含む。あるいは、足場材料は、ガラスやシリコーンなどの無機材料を主成分としたものでもよい。 Synthetic resin refers to a material whose main component is a polymer (hereinafter, also simply referred to as "polymer") obtained by polymerizing (including polycondensation) a polymerizable monomer (hereinafter, also simply referred to as "monomer"). The above polymer also includes copolymers of one or more polymerizable monomers. Alternatively, the scaffold material may be one whose main component is an inorganic material such as glass or silicone.
上記ポリマーとしては、例えば、(不)飽和炭化水素、芳香族炭化水素、(不)飽和脂肪酸、芳香族カルボン酸、(不)飽和ケトン、芳香族ケトン、(不)飽和アルコール、芳香族アルコール、(不)飽和アミン、芳香族アミン、(不)飽和チオール、芳香族チオール、有機ケイ素化合物の1種以上の重合性モノマーからなるポリマーが挙げられる。 Examples of the above polymers include polymers made of one or more polymerizable monomers of (un)saturated hydrocarbons, aromatic hydrocarbons, (un)saturated fatty acids, aromatic carboxylic acids, (un)saturated ketones, aromatic ketones, (un)saturated alcohols, aromatic alcohols, (un)saturated amines, aromatic amines, (un)saturated thiols, aromatic thiols, and organosilicon compounds.
具体的な上記ポリマーとしては、例えば、ポリスチレン、ポリオレフィン、ポリエーテル、ポリビニルアルコール、ポリビニルアセタール、ポリエステル、ポリ(メタ)アクリル酸エステル、エポキシ樹脂、ポリアミド、ポリイミド、ポリウレタン、ポリカーボネート、セルロース、デキストラン、ポリペプチド(例:ゼラチン等)などが挙げられる。これらのポリマーは、一種類で用いてもよいし、二種類以上組み合わせて用いてもよい。二種類以上のポリマーを組み合わせる場合は、二種類以上のポリマーを混合して用いてもよいし、二種類以上のポリマーの骨格を化学結合させたポリマーとして用いてもよい。 Specific examples of the above polymers include polystyrene, polyolefin, polyether, polyvinyl alcohol, polyvinyl acetal, polyester, poly(meth)acrylic acid ester, epoxy resin, polyamide, polyimide, polyurethane, polycarbonate, cellulose, dextran, and polypeptide (e.g., gelatin). These polymers may be used alone or in combination of two or more. When combining two or more polymers, the two or more polymers may be mixed and used, or the skeletons of the two or more polymers may be chemically bonded to form a polymer.
足場材料は公知の方法により製造してもよいし、市販品を用いてもよい。市販品としては、例えば、Cytodex-1(GE Healthcare)、Corning(登録商標)低濃度 Synthemax(登録商標)II マイクロキャリア(Corning社)などが挙げられる。 Scaffolding materials may be produced by known methods, or commercially available products may be used. Examples of commercially available products include Cytodex-1 (GE Healthcare) and Corning (registered trademark) low concentration Synthemax (registered trademark) II microcarriers (Corning, Inc.).
典型的には、上記足場材料のうち、アテロコラーゲンを含有する足場材料は、該足場材料の表面の全部または一部をアテロコラーゲンで被覆(コーティング)することで、所望するアテロコラーゲンを含有する足場材料を調製することができる。被覆に用いるアテロコラーゲンの純度は、特に限定されないが、高純度(例えば、90%以上、より好ましくは95%以上、最も好ましくは100%)であることが好ましい。マイクロキャリア等の遊離した足場材料の表面と細胞との接着性を向上させる目的で、アテロコラーゲンに加えて、細胞外マトリックス(ECM)等の任意の細胞支持用基質でコーティングされたものであり得る。また、ネイティブコラーゲンは実質的に含まれない(例えば、10%以下、より好ましく5%以下(例:4%、3%、2%、1%、0%)ことが好ましい。細胞支持用基質は、幹細胞またはフィーダー細胞(用いられる場合)の接着を目的とする任意の物質であり得る。このような細胞支持用基質としては、コラーゲン、ゼラチン、ポリ-L-リジン、ポリ-D-リジン、ラミニン(または、ラミニンの一部構造体)、およびフィブロネクチン並びにそれらの混合物、例えばマトリゲル、並びに溶解細胞膜調製物などが挙げられる(Lancet,2005.365.9471.1636-1641)。本明細書において、「純度」とは、特に断らない限り品質の高さ(不純物の混入割合の低さ)を示す指標としての質量パーセント濃度(以下では、「質量パーセント濃度」を単に「濃度」という)を意味するが、混合物(例えば、アテロコラーゲンとその他の細胞支持用基質との混合物など)中の特定成分(例えば、アテロコラーゲン)の濃度を指す場合もある。 Typically, among the above scaffold materials, a scaffold material containing atelocollagen can be prepared by covering (coating) all or part of the surface of the scaffold material with atelocollagen to prepare a scaffold material containing the desired atelocollagen. The purity of the atelocollagen used for coating is not particularly limited, but it is preferable that it is highly pure (e.g., 90% or more, more preferably 95% or more, and most preferably 100%). In order to improve the adhesiveness between the surface of a free scaffold material such as a microcarrier and cells, the scaffold material may be coated with any cell-supporting substrate such as an extracellular matrix (ECM) in addition to atelocollagen. Furthermore, it is preferable that the material is substantially free of native collagen (e.g., 10% or less, more preferably 5% or less (e.g., 4%, 3%, 2%, 1%, 0%). The cell support substrate can be any material intended for the attachment of stem cells or feeder cells (if used). Such cell support substrates include collagen, gelatin, poly-L-lysine, poly-D-lysine, laminin (or a partial structure of laminin), and fibronectin, as well as mixtures thereof, such as Matrigel, and lysed cell membrane preparations (Lancet, 2005.365.9471.1636-1641). In this specification, unless otherwise specified, "purity" means a mass percent concentration (hereinafter, "mass percent concentration" is simply referred to as "concentration") as an indicator of high quality (low rate of impurity contamination), but may also refer to the concentration of a specific component (e.g., atelocollagen) in a mixture (e.g., a mixture of atelocollagen and other cell support substrates).
上記足場材料の形状は特に限定されないが、例えば、円筒形、長球の形状、球形などが挙げられるが、好ましくは球形である。かかる球形の足場材料としては、具体的には、マイクロキャリアなどが挙げられる。本発明者らは以前、バイオリアクターを用いた培養において、直径が105μm以下、105~250μm、250~425μm、425~600μmのマイクロキャリアのいずれを用いても、多能性幹細胞が増殖できることを見出している。足場材料の大きさも特に限定されないが、マイクロキャリアなどの球形のものを用いる場合には、足場材料の粒径(直径)は、典型的には、50~1000μmであり、70~700μmであってもよく、100~400μmであることが好ましい。また、細胞の増殖率の観点から、好適な一態様において、粒径は600μmである。粒径は、国際規格ISO 13319「粒度分布の測定-電気的検知帯法」に記載のコールターカウンター法により測定することができる。 The shape of the scaffold material is not particularly limited, and examples thereof include cylindrical, oval, and spherical shapes, with spherical shapes being preferred. Specific examples of such spherical scaffold materials include microcarriers. The inventors have previously found that pluripotent stem cells can be grown using microcarriers with diameters of 105 μm or less, 105 to 250 μm, 250 to 425 μm, and 425 to 600 μm in culture using a bioreactor. The size of the scaffold material is also not particularly limited, but when using a spherical scaffold material such as a microcarrier, the particle size (diameter) of the scaffold material is typically 50 to 1000 μm, may be 70 to 700 μm, and is preferably 100 to 400 μm. In addition, in a preferred embodiment, the particle size is 600 μm from the viewpoint of cell proliferation rate. The particle size can be measured by the Coulter counter method described in the international standard ISO 13319 "Measurement of particle size distribution - Electrical detection zone method".
I型コラーゲン分子は、約95%のらせん(ヘリックス)部分と約5%の非らせん部分(テロペプチド)からできている。この非ヘリックス部分は抗原性の強い領域であり、プロテアーゼ(タンパク質分解酵素)により切断される。抗原性の強いテロペプチド部分をペプシンなどのプロテアーゼで消化・除去後に高度精製した抗原性の極めて低い天然高分子材料が、足場材料に含有されるアテロコラーゲンである(Matrix, 1992, 12. 274-281)。一方で、生体内に存在するコラーゲンは、3本のポリペプチド鎖が、らせんを巻いた「3重らせん構造」の不溶性の線維状タンパク質でありNativeコラーゲンとも言われる。本発明で用いるアテロコラーゲンの由来は限定されず、例えば、哺乳動物(例:ヒト、マウス、ラット、サル、ウシ、ウマ、ブタ、イヌ等)由来のものなどが挙げられる。異種動物由来成分の混入防止の観点からは、培養する細胞と同一由来のアテロコラーゲンを用いることが好ましい。かかるアテロコラーゲンは公知の方法により製造してもよく、市販品を用いてもよい。例えば、アテロコラーゲンを含む細胞や組織から抽出したコラーゲンや、培養細胞から分泌させたコラーゲンをプロテアーゼで処理してアテロコラーゲンを精製することができる。 Type I collagen molecules are composed of about 95% helical (helix) parts and about 5% non-helical parts (telopeptides). This non-helical part is a region with high antigenicity and is cleaved by proteases (protein-degrading enzymes). The atelocollagen contained in the scaffold material is a natural polymeric material with extremely low antigenicity that is highly purified after digestion and removal of the highly antigenic telopeptide part with proteases such as pepsin (Matrix, 1992, 12. 274-281). On the other hand, collagen present in the living body is an insoluble fibrous protein with a "triple helix structure" in which three polypeptide chains are wound in a helix, and is also called native collagen. The origin of the atelocollagen used in the present invention is not limited, and examples include those derived from mammals (e.g., humans, mice, rats, monkeys, cows, horses, pigs, dogs, etc.). From the viewpoint of preventing contamination with components derived from different animals, it is preferable to use atelocollagen derived from the same origin as the cells to be cultured. Such atelocollagen may be produced by known methods, or a commercially available product may be used. For example, atelocollagen can be purified by treating collagen extracted from cells or tissues containing atelocollagen, or collagen secreted from cultured cells, with a protease.
足場材料におけるアテロコラーゲンの濃度((アテロコラーゲンの質量/アテロコラーゲンを含有する足場材料の質量)×100)は、細胞に対して細胞死抑制効果を示す濃度である限りにおいて、特に限定されない。このような濃度は、実施例記載の方法および従来公知の方法を用いて当業者が適宜設定することができる。足場材料におけるアテロコラーゲンの濃度は、例えば、0.1%以上(例:0.1%、1%、3%、5%、10%、20%、25%、30%またはそれ以上)であり、100%以下である。足場材料に、アテロコラーゲンを含む細胞支持用基質がコーティングされている場合には、該細胞支持用基質におけるアテロコラーゲンの濃度は、90%以上(例:91%、92%、93%、94%、95%、95.5%、96%、97%、98%、99%または100%)である。本発明の一態様において、足場材料は実質的にアテロコラーゲンからなるが、「実質的にアテロコラーゲンからなる」とは、アテロコラーゲンの濃度が100%の場合だけでなく、100%に近い濃度(例えば、95%以上、好ましくは95.5%以上(例:96%、97%、98%、99%または100%))であることを意味する。 The concentration of atelocollagen in the scaffold material ((mass of atelocollagen/mass of scaffold material containing atelocollagen) x 100) is not particularly limited as long as it is a concentration that exhibits a cell death inhibitory effect on cells. Such a concentration can be appropriately set by a person skilled in the art using the methods described in the Examples and conventionally known methods. The concentration of atelocollagen in the scaffold material is, for example, 0.1% or more (e.g., 0.1%, 1%, 3%, 5%, 10%, 20%, 25%, 30% or more) and 100% or less. When the scaffold material is coated with a cell support substrate containing atelocollagen, the concentration of atelocollagen in the cell support substrate is 90% or more (e.g., 91%, 92%, 93%, 94%, 95%, 95.5%, 96%, 97%, 98%, 99% or 100%). In one aspect of the present invention, the scaffold material is substantially composed of atelocollagen, and "substantially composed of atelocollagen" does not mean that the atelocollagen concentration is 100%, but rather that the concentration is close to 100% (e.g., 95% or more, preferably 95.5% or more (e.g., 96%, 97%, 98%, 99% or 100%)).
また、培地におけるアテロコラーゲンの濃度も特に制限されず、アテロコラーゲンの濃度を適宜設定することで、細胞の増殖率を制御することも可能となる。培地におけるアテロコラーゲンの濃度は、例えば0.01~20%、好ましくは0.05~5%、より好ましくは0.1~2%である。また、培地におけるアテロコラーゲンの濃度は、0.5%~20%、1%~15%または5%~10%であることも好ましい。 In addition, the concentration of atelocollagen in the medium is not particularly limited, and by appropriately setting the concentration of atelocollagen, it is possible to control the cell proliferation rate. The concentration of atelocollagen in the medium is, for example, 0.01 to 20%, preferably 0.05 to 5%, and more preferably 0.1 to 2%. In addition, it is also preferable that the concentration of atelocollagen in the medium is 0.5% to 20%, 1% to 15%, or 5% to 10%.
本発明の製造方法(I)における培養(典型的には、工程s1~工程s4)条件は、特に制限されないが、約30~40℃、好ましくは約37℃であり、CO2含有空気の雰囲気下で培養が行われ、CO2濃度は、好ましくは約2~5%である。例えば、各容器内の温度の制御は、従来公知の温度制御法を参照することができ、例えば、室温の温度制御、各容器へのヒーターの付与、容器内に供給される材料の温度制御などが挙げられる。 The conditions for the culture (typically, steps s1 to s4) in the production method (I) of the present invention are not particularly limited, but are about 30 to 40° C., preferably about 37° C., and culture is performed in an atmosphere of CO 2 -containing air, with a CO 2 concentration of preferably about 2 to 5%. For example, the temperature in each container can be controlled by referring to a conventionally known temperature control method, such as temperature control at room temperature, providing a heater to each container, and temperature control of the material supplied to the container.
本発明の製造方法(I)における培養(典型的には、工程s1~工程s4)における細胞の培養密度は、細胞が増殖できる限り特に限定されない。通常1.0×102~1.0×107細胞/cm2、好ましくは1.0×103~1.0×106細胞/cm2、より好ましくは1.0×104~1.0×105細胞/cm2である。 The cell culture density in the culture (typically, steps s1 to s4) in the production method (I) of the present invention is not particularly limited as long as the cells can grow, and is usually 1.0×10 2 to 1.0×10 7 cells/cm 2 , preferably 1.0×10 3 to 1.0×10 6 cells/cm 2 , and more preferably 1.0×10 4 to 1.0×10 5 cells/cm 2 .
本発明の製造方法(I)における工程s1を行う期間は、工程s3においてiPS細胞が樹立される限り、特に限定されないが、典型的には、例えば、30分~24時間、2時間~6時間等が挙げられる。 The period for performing step s1 in the production method (I) of the present invention is not particularly limited as long as iPS cells are established in step s3, but typically includes, for example, 30 minutes to 24 hours, 2 hours to 6 hours, etc.
本発明の製造方法(I)における工程s2において、液体培地中の初期化因子の濃度を低減させる方法は、特に限定されないが、工程s1で用いた培地を、初期化因子を添加しない培地と交換することで達成し得る。該培地交換は、希釈により培地中の初期化因子の濃度が低減することで、体細胞と初期化因子の接触(頻度)が低減あるいは終了する限り、工程s1で用いた培地の全ての交換でもよく、その一部の交換であってもよい。本明細書において、「初期化因子の濃度が低減する」には、培地中に初期化因子が存在しない状態も含み得る。典型的には、工程s2における培地中の初期化因子の濃度は、工程s1と比して、例えば、1/10以下であり、好ましくは1/100以下、より好ましくは1/300以下、さらに好ましくは1/1000以下、さらにより好ましくは1/3000以下である。また、工程s2から工程s3へは、上述のように培地中の初期化因子が所望の濃度に低減されれば、工程を進めてもよい。 In step s2 of the production method (I) of the present invention, the method of reducing the concentration of the reprogramming factor in the liquid medium is not particularly limited, but can be achieved by replacing the medium used in step s1 with a medium to which no reprogramming factor is added. The medium replacement may be a complete replacement of the medium used in step s1, or a partial replacement, as long as the concentration of the reprogramming factor in the medium is reduced by dilution, thereby reducing or terminating the contact (frequency) between the somatic cells and the reprogramming factor. In this specification, "reducing the concentration of the reprogramming factor" may also include a state in which no reprogramming factor is present in the medium. Typically, the concentration of the reprogramming factor in the medium in step s2 is, for example, 1/10 or less, preferably 1/100 or less, more preferably 1/300 or less, even more preferably 1/1000 or less, and even more preferably 1/3000 or less, compared to step s1. In addition, the process may proceed from step s2 to step s3 if the reprogramming factor in the medium is reduced to a desired concentration as described above.
本発明の製造方法(I)における工程s3は、液体培地中で前記体細胞を培養して、iPS細胞を樹立する工程であり、人工多能性幹細胞の樹立は、自体公知の方法により導入した初期化因子の発現等(例えば、Oct3/4、SOX2、Nanog、TRA-1-60、TRA-1-81、SSEA3、SSEA4、アルカリホスファターゼ等)により適宜確認し得る。
本発明の製造方法(I)における工程s3を行う期間は、iPS細胞が樹立される限り、特に限定されないが、典型的には、例えば、10日間以上であり、14日間以上が好ましい。上限についても特に制限はないが、典型的には40日間以下であり、30日間以下が好ましい。
Step s3 in the production method (I) of the present invention is a step of culturing the somatic cells in a liquid medium to establish iPS cells, and the establishment of induced pluripotent stem cells can be appropriately confirmed by the expression of reprogramming factors introduced by a method known per se (e.g., Oct3/4, SOX2, Nanog, TRA-1-60, TRA-1-81, SSEA3, SSEA4, alkaline phosphatase, etc.).
The period for carrying out step s3 in the production method (I) of the present invention is not particularly limited as long as iPS cells are established, but is typically, for example, 10 days or more, preferably 14 days or more. There is also no particular upper limit, but it is typically 40 days or less, preferably 30 days or less.
各材料供給源(G1~Gn)から各容器に供給される材料は、前記した各工程の実施するために必要な材料(例えば、体細胞、培地、初期化因子等)である。上述した以外の体細胞からiPS細胞を得るために必要な材料それ自体は、従来技術を参照することができる。細胞培養に必要なO2やCO2などのガスも供給すべき材料であるが、容器の壁部がガス透過性であれば、該ガスの供給を省略することができる。以下の他の容器も同様である。 The materials supplied from each material supply source (G1 to Gn) to each container are materials necessary for carrying out each of the above-mentioned steps (e.g., somatic cells, culture medium, reprogramming factors, etc.). For the materials themselves necessary for obtaining iPS cells from somatic cells other than those mentioned above, reference can be made to the prior art. Gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of the gases can be omitted. The same applies to the other containers described below.
当該製造方法(I)は、上記したiPS細胞を樹立する工程s3の後に、iPS細胞の収率の観点から、該iPS細胞を拡大培養する工程s4をさらに有することが好ましい。該拡大培養の回数はp回である(pは1以上の整数、即ち、p≧1)。 From the viewpoint of the yield of iPS cells, it is preferable that the manufacturing method (I) further includes a step s4 of expanding the iPS cells after the step s3 of establishing the iPS cells. The number of times of the expansion is p (p is an integer of 1 or more, i.e., p≧1).
以下に、拡大培養の工程s4を詳細に説明する。
該拡大培養の工程s4で用いられる容器、入出用ポート、接続管路、送り機構のそれぞれの構成は、上記工程s1~s3の説明で示した構成を参照することができ、ここでは説明を省略する。
The expansion culture step s4 will be described in detail below.
The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in the expansion culture step s4 can be referenced from the configurations shown in the explanation of steps s1 to s3 above, and therefore will not be explained here.
工程s4では、工程s3で樹立した、第n番目の密閉容器(An)中の内容物(即ち、iPS細胞を含む培地)の一部または全部を、容器B1に移動し所望する細胞密度になるまで培養する。工程s5、工程s6と拡大培養を継続する場合も、各工程の移動ごとに移動する内容物は工程s3から工程s4と同様である。 In step s4, some or all of the contents (i.e., culture medium containing iPS cells) in the nth sealed container (An) established in step s3 are transferred to container B1 and cultured until the desired cell density is reached. When the expansion culture is continued in steps s5 and s6, the contents transferred at each step are the same as those from step s3 to step s4.
図8に一例を示すように、iPS細胞を拡大培養する工程s4では、上記した工程s3が実施される第n番目の容器Anの後に、該拡大培養の回数pに対応するp個の容器(B1~Bp)が、接続管路を介して直列に接続される。各容器(B1~Bp)の入出用ポートには、各容器に対応付けられた工程が実施されるように、必要な材料供給源(Gb1~Gbp)が接続されている。各容器に接続される材料供給源の数は、図ではそれぞれ1つだけが示されているが、その数は限定されない。図8では、液面に達していない入出用ポートの図示を省略している。上記工程s3のための最後の容器(An)から工程s4のための最後の容器(Bp)まで、それぞれ送り機構FnおよびFb1~Fb(p-1)によって、iPS細胞が一方向に順次移動し、容器(B1~Bp)のそれぞれの内部で1回の拡大培養が実施され、それにより合計p回の拡大培養が実施されて、容器Bpにおいて工程s4が完了し、所望する細胞数のiPS細胞が得られる。 As shown in an example in Figure 8, in step s4 of expanding iPS cells, p containers (B1 to Bp) corresponding to the number p of times of expansion culture are connected in series via connecting pipelines after the nth container An in which the above-mentioned step s3 is carried out. Necessary material supply sources (Gb1 to Gbp) are connected to the input/output ports of each container (B1 to Bp) so that the process corresponding to each container is carried out. Although only one material supply source is shown to be connected to each container in the figure, the number is not limited. Input/output ports that have not reached the liquid level are not shown in Figure 8. From the last container (An) for step s3 to the last container (Bp) for step s4, the iPS cells are moved sequentially in one direction by the feed mechanisms Fn and Fb1 to Fb(p-1), respectively, and one expansion culture is carried out inside each of the containers (B1 to Bp), thereby carrying out a total of p expansion cultures, completing step s4 in container Bp and obtaining the desired number of iPS cells.
(拡大培養の回数p)
工程s4において、拡大培養を何回行なうかは、所望するiPS細胞数を得られれば特に限定はされないが、通常の処理操作では、1~5回程度が好ましく、より好ましくは2~5回程度が挙げられる。当該製造方法(I)における拡大培養の回数p(=容器の数)は、典型的には、1~5程度が好ましい数であり、より好ましくは2~5回程度が挙げられる。
(Number of times of expansion culture p)
In step s4, the number of times expansion culture is performed is not particularly limited as long as the desired number of iPS cells can be obtained, but in a normal processing operation, it is preferably about 1 to 5 times, more preferably about 2 to 5 times. The number of times p (= the number of containers) of expansion culture in the production method (I) is typically preferably about 1 to 5, more preferably about 2 to 5 times.
本発明の製造方法(I)における工程s4を行う期間は、所望するiPS細胞数が得られれば特に限定されないが、典型的には、例えば、1~40日間、3~20日間、5~10日間である。 The period for which step s4 is performed in the production method (I) of the present invention is not particularly limited as long as the desired number of iPS cells can be obtained, but is typically, for example, 1 to 40 days, 3 to 20 days, or 5 to 10 days.
拡大培養において、各材料供給源(Gb1~Gbp)から各容器に供給される材料は、拡大培養に必要なものであり、主として培地である。また、細胞培養に必要なO2やCO2などのガスも供給すべき材料であるが、容器の壁部がガス透過性であれば、該ガスの供給を省略することができる。 In the expansion culture, the materials supplied from each material supply source (Gb1 to Gbp) to each container are necessary for the expansion culture, and are mainly culture media. Gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of these gases can be omitted.
2.分化細胞の製造方法
次に、本発明による分化細胞の製造方法(以下、製造方法(II)ともいう)を、本発明による細胞製造装置の構成例を参照しながら詳細に説明する。装置各部の説明は、後述の細胞製造装置の各部の説明でもある。
2. Method for Producing Differentiated Cells Next, the method for producing differentiated cells according to the present invention (hereinafter also referred to as production method (II)) will be described in detail with reference to an example of the configuration of the cell production device according to the present invention. The description of each part of the device also includes the description of each part of the cell production device described below.
当該製造方法(II)は、1態様では、上記した製造方法(I)における工程s1~s3と、iPS細胞を分化誘導する工程s5とを有する。また、当該製造方法(II)の好ましい態様では、工程s1~s3の後に拡大培養の工程s4が加えられ、よって、当該製造方法(II)は、上記工程s1~s4と、iPS細胞を分化誘導する工程s5とを有する。 In one embodiment, the manufacturing method (II) comprises steps s1 to s3 in the manufacturing method (I) described above, and step s5 of inducing differentiation of iPS cells. In a preferred embodiment of the manufacturing method (II), step s4 of expansion culture is added after steps s1 to s3, and thus the manufacturing method (II) comprises the above steps s1 to s4, and step s5 of inducing differentiation of iPS cells.
また、既に準備したiPS細胞を用いて分化細胞を製造する場合、当該製造方法(II)は、その前段に製造方法(I)を持たない独立した細胞製造方法であってもよい。その場合、分化誘導の工程s6が複数であって、各工程に対応付けられた容器が複数であれば、〔複数の細胞製造工程を複数の容器に分けて、工程の順に容器から容器へと細胞を移動させ、それにより、各容器に対応付けられた工程を順次実施していく〕という本発明の細胞製造方法に該当する。 In addition, when differentiating cells are produced using iPS cells that have already been prepared, the production method (II) may be an independent cell production method that does not have production method (I) as a preceding step. In that case, if there are multiple differentiation induction steps s6 and multiple containers associated with each step, this corresponds to the cell production method of the present invention in which [multiple cell production steps are divided into multiple containers, cells are moved from container to container in the order of the steps, and the steps associated with each container are carried out sequentially].
図9は、当該製造方法(II)を説明するためのブロック図である。図9(a)に示すように、当該製造方法(II)では、上記製造方法(I)で用いられた容器のうちの最後尾の容器X1の後に、工程s5を実施するための容器C1が、接続管路を介してさらに接続される。容器X1は、図1における工程s1~s3の最後の容器Anであってもよいし、図8における工程s4の最後の容器Bpであってもよい。容器C1は、1以上の開閉可能な入出用ポートを有し、該入出用ポートを通じて、工程s5の分化誘導に必要な材料が容器C1の内部に供給される(外部の材料供給源は図示を省略している)。図9では、液面に達していない入出用ポートの図示を省略している。送り機構Fx1によって、容器X1から容器C1へと、iPS細胞が移動し、該容器C1において前記工程s5が実施される。 FIG. 9 is a block diagram for explaining the manufacturing method (II). As shown in FIG. 9(a), in the manufacturing method (II), a container C1 for carrying out step s5 is further connected through a connecting pipeline after the last container X1 among the containers used in the manufacturing method (I). The container X1 may be the last container An of steps s1 to s3 in FIG. 1, or the last container Bp of step s4 in FIG. 8. The container C1 has one or more openable and closable inlet/outlet ports, and materials necessary for differentiation induction in step s5 are supplied to the inside of the container C1 through the inlet/outlet ports (external material supply sources are not shown). In FIG. 9, inlet/outlet ports that have not reached the liquid level are not shown. The iPS cells are transferred from the container X1 to the container C1 by the feed mechanism Fx1, and the above-mentioned step s5 is carried out in the container C1.
該分化誘導の工程s5(後述の工程s5a、s5bを含む)で用いられる容器、入出用ポート、接続管路、送り機構のそれぞれの構成は、上記工程s1~s4の説明で示した構成を参照することができ、ここでは説明を省略する。 The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in differentiation induction step s5 (including steps s5a and s5b described below) can be referenced from the configurations described in steps s1 to s4 above, and will not be described here.
各容器に供給される材料は、分化誘導のために必要なものである。分化誘導に必要な材料それ自体は、後述するものを含め、従来技術も参照することができる。細胞培養に必要なO2やCO2などのガスも供給すべき材料であるが、容器の壁部がガス透過性であれば、該ガスの供給を省略することができる。 The materials supplied to each container are necessary for differentiation induction. For the materials themselves necessary for differentiation induction, reference can be made to the prior art, including those described below. Gases such as O2 and CO2 necessary for cell culture are also materials that should be supplied, but if the walls of the container are gas permeable, the supply of the gases can be omitted.
(分化細胞)
本発明の製造方法(II)では、人工多能性幹細胞を分化誘導させて、所望する細胞またはオルガノイドを製造し得る。得られる細胞は、幹細胞や前駆細胞などの未分化細胞であってもよく、最終分化細胞であってもよい。本明細書において、本発明の製造方法(II)で除去対象となる未分化細胞は、本発明の製造方法(II)により製造することを企図した幹細胞や前駆細胞等以外の細胞を意味する。以下で、本発明の製造方法(II)により製造され得る未分化細胞と最終分化細胞の両方を包含する用語として、「分化細胞」との用語を用いることがある。また、生殖細胞であってもよい。本明細書において、「未分化細胞」とは、細胞系譜において、最終分化に至っていない細胞を意味し、未分化細胞としては、例えば、多能性幹細胞を除く幹細胞、前駆細胞などが挙げられる。幹細胞または前駆細胞としては、例えば、エピブラスト様細胞、始原生殖細胞(「始原生殖細胞様細胞」とも称する)、生殖幹細胞、外胚葉系(列の)細胞である神経堤細胞、神経幹細胞、神経前駆細胞、グリア前駆細胞、網膜幹細胞、角膜幹細胞、ケラチノサイト表皮幹細胞、メラノサイト幹細胞、乳腺幹細胞、中胚葉系(列の)細胞である造血前駆細胞、骨髄系幹細胞、リンパ系幹細胞、B前駆細胞、T前駆細胞、間葉系幹細胞、心臓幹細胞、心臓前駆細胞、血管内皮前駆細胞、血管周皮細胞、血小板前駆細胞(例:巨核球前駆細胞、巨核芽球、前巨核球、成熟巨核球等)、骨格筋幹細胞、脂肪幹細胞、腎前駆細胞、内胚葉系(列の)細胞である肝幹細胞、肝臓前駆細胞(例:肝芽細胞、肝前駆細胞、肝星細胞前駆細胞、肝幹前駆細胞等)、腸幹細胞、気道幹細胞などが挙げられる。
(differentiated cells)
In the production method (II) of the present invention, the desired cells or organoids can be produced by inducing differentiation of artificial pluripotent stem cells. The obtained cells may be undifferentiated cells such as stem cells and precursor cells, or may be terminally differentiated cells. In this specification, the undifferentiated cells to be removed in the production method (II) of the present invention refer to cells other than stem cells and precursor cells intended to be produced by the production method (II) of the present invention. Hereinafter, the term "differentiated cells" may be used as a term that encompasses both undifferentiated cells and terminally differentiated cells that can be produced by the production method (II) of the present invention. They may also be germ cells. In this specification, "undifferentiated cells" refers to cells that have not reached terminal differentiation in the cell lineage, and examples of undifferentiated cells include stem cells and precursor cells excluding pluripotent stem cells. Examples of stem or progenitor cells include epiblast-like cells, primordial germ cells (also referred to as "primordial germ cell-like cells"), germline stem cells, ectodermal (row) cells such as neural crest cells, neural stem cells, neural progenitor cells, glial progenitor cells, retinal stem cells, corneal stem cells, keratinocyte epidermal stem cells, melanocyte stem cells, mammary stem cells, mesodermal (row) cells such as hematopoietic progenitor cells, bone marrow stem cells, lymphatic stem cells, B progenitor cells, T progenitor cells, mesenchymal stem cells, cardiac stem cells, cardiac progenitor cells, vascular endothelial progenitor cells, vascular pericytes, platelet progenitor cells (e.g., megakaryocyte progenitor cells, megakaryoblasts, promegakaryocytes, mature megakaryocytes, etc.), skeletal muscle stem cells, adipose stem cells, kidney progenitor cells, endodermal (row) cells such as hepatic stem cells, liver progenitor cells (e.g., hepatoblasts, hepatic progenitor cells, hepatic stellate cell progenitors, hepatic stem progenitor cells, etc.), intestinal stem cells, and airway stem cells.
本明細書において、「最終分化細胞」とは、細胞系譜において、最終分化に至った細胞を意味し、最終分化細胞としては、特に限定されないが、例えば、骨芽細胞、軟骨細胞、脂肪細胞、肝細胞、肝中皮細胞、胆管上皮細胞、肝星細胞、肝類洞内皮細胞、クッパー細胞、ピット細胞、血管内皮細胞、血液細胞(例:赤血球、血小板、白血球、肥満細胞、樹状細胞等)、膵管上皮細胞、膵導管細胞、腺房中心細胞、腺房細胞、ランゲルハンス島、心筋細胞、線維芽細胞、平滑筋細胞、I型肺胞上皮細胞、II型肺胞上皮細胞、クララ細胞、線毛上皮細胞、基底細胞、杯細胞、神経内分泌細胞、クルチッキー細胞、尿細管上皮細胞、尿路上皮細胞、円柱上皮細胞、糸球体上皮細胞、糸球体内皮細胞、蛸足細胞、メサンギウム細胞、神経細胞、グリア細胞(例:アストロサイト、ミクログリア、オリゴデンドロサイト、上衣細胞、シュワン細胞等)などが挙げられる。白血球としては、リンパ球(例:B細胞、T細胞、NK細胞等)、顆粒球(例:好中球、好酸球、好塩基球等)、単球などが挙げられる。 As used herein, the term "terminally differentiated cells" refers to cells that have reached terminal differentiation in the cell lineage. Examples of terminally differentiated cells include, but are not limited to, osteoblasts, chondrocytes, adipocytes, hepatic mesothelial cells, bile duct epithelial cells, hepatic stellate cells, hepatic sinusoidal endothelial cells, Kupffer cells, pit cells, vascular endothelial cells, blood cells (e.g., red blood cells, platelets, white blood cells, mast cells, dendritic cells, etc.), pancreatic ductal epithelial cells, pancreatic ductal cells, acinar center cells, acinar cells, laminar cells, and endothelial cells. These include islets of Gerhans, cardiac muscle cells, fibroblasts, smooth muscle cells, type I alveolar epithelial cells, type II alveolar epithelial cells, Clara cells, ciliated epithelial cells, basal cells, goblet cells, neuroendocrine cells, Kruczyk cells, renal tubular epithelial cells, urothelial cells, columnar epithelial cells, glomerular epithelial cells, glomerular endothelial cells, octopus podocytes, mesangial cells, nerve cells, glial cells (e.g., astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells, etc.). White blood cells include lymphocytes (e.g., B cells, T cells, NK cells, etc.), granulocytes (e.g., neutrophils, eosinophils, basophils, etc.), monocytes, etc.
本発明の一実施態様において、本発明の製造方法(II)により得られる細胞やオルガノイド(目的の細胞またはオルガノイド)は、神経堤細胞、神経前駆細胞、神経細胞、大脳皮質オルガノイド、造血前駆細胞、血小板、T細胞、エピブラスト様細胞、始原生殖細胞または心筋細胞である。また、内エナメル上皮、エナメル芽細胞、中間層細胞、星状網細胞、外エナメル上皮、歯乳頭細胞、象牙芽細胞でもある。 In one embodiment of the present invention, the cells or organoids (target cells or organoids) obtained by the production method (II) of the present invention are neural crest cells, neural precursor cells, nerve cells, cerebral cortical organoids, hematopoietic precursor cells, platelets, T cells, epiblast-like cells, primordial germ cells, or cardiomyocytes. They may also be inner enamel epithelium, ameloblasts, stratum intermedium cells, stellate reticulum cells, outer enamel epithelium, dental papilla cells, or odontoblasts.
また、本明細書において、「オルガノイド」とは、細胞が集積して形成された構造体を意味し、典型的には、生体内の臓器と類似した構造及び機能を有する。本発明の分化誘導法により得られるオルガノイドとして、例えば、神経オルガノイド(例:大脳皮質オルガノイド、小脳オルガノイド、脊髄オルガノイド、中脳オルガノイド、脈絡叢オルガノイド、海馬オルガノイド、視床下部オルガノイド、脳下垂体前葉オルガノイド及び大脳基底核オルガノイド等)、肺オルガノイド、肝オルガノイド、気道上皮オルガノイド、腸オルガノイド、膵臓オルガノイド、腎臓オルガノイド、気道オルガノイド、胃オルガノイド、甲状腺オルガノイド、胸腺オルガノイド、精巣オルガノイド、食道オルガノイド、皮膚オルガノイド、卵管オルガノイド、卵巣オルガノイド、唾液腺オルガノイド、眼胞オルガノイド、眼杯オルガノイド、膀胱オルガノイド、前立腺オルガノイド、軟骨オルガノイド、心臓オルガノイド、骨組織オルガノイド、筋組織オルガノイド、がんオルガノイドなどが挙げられるが、一実施態様において、大脳皮質オルガノイドなどの神経オルガノイドである。 In addition, as used herein, "organoid" refers to a structure formed by the accumulation of cells, and typically has a structure and function similar to that of an organ in the body. Organoids obtained by the differentiation induction method of the present invention include, for example, neural organoids (e.g., cerebral cortex organoids, cerebellar organoids, spinal cord organoids, midbrain organoids, choroid plexus organoids, hippocampal organoids, hypothalamic organoids, anterior pituitary organoids, and basal ganglia organoids, etc.), lung organoids, liver organoids, airway epithelial organoids, intestinal organoids, pancreatic organoids, kidney organoids, airway organoids, stomach organoids, thyroid organoids, thymus organoids, testicular organoids, esophageal organoids, skin organoids, fallopian tube organoids, ovarian organoids, salivary gland organoids, ocular vesicle organoids, optic cup organoids, bladder organoids, prostate organoids, cartilage organoids, cardiac organoids, bone tissue organoids, muscle tissue organoids, and cancer organoids. In one embodiment, the organoids are neural organoids such as cerebral cortex organoids.
ある構造体がオルガノイドであるかどうかは、例えば、顕微鏡観察により層構造形成の有無の確認や、マーカータンパク質の発現を調べることにより確認することができる。具体的には、大脳皮質オルガノイドの場合には、Foxg1が全体に発現したドーム状の神経上皮が認められ、該神経上皮は層構造を形成する。該構造において、上皮の内側にPax6、Sox2陽性のventricular zoneに相当する神経前駆細胞の層が、その外側に、Reelin/Carletinin陽性のCajal Retzius細胞からなる第1層や、Ctip2/Tbr1陽性のdeep layerなどが認められる。また、肝オルガノイドでは、HHEX、SOX2、HNF4A、AFP、ALBなどがマーカーになり、膵臓オルガノイドでは、PDX1、SOX17、SOX9などがマーカーになり、腸管に分化するオルガノイドでは、CDX2、SOX9などがマーカーになり、腎臓オルガノイドでは、Pax2、Six2などがマーカーになる。 Whether a certain structure is an organoid can be confirmed, for example, by observing with a microscope to see whether a layer structure is formed or not, or by examining the expression of marker proteins. Specifically, in the case of cerebral cortical organoids, a dome-shaped neuroepithelium with Foxg1 expressed throughout is observed, and this neuroepithelium forms a layer structure. In this structure, a layer of neural precursor cells corresponding to the Pax6- and Sox2-positive ventricular zone is observed inside the epithelium, and on the outside of this, a first layer consisting of Reelin/Carletinin-positive Cajal-Retzius cells and a Ctip2/Tbr1-positive deep layer are observed. In addition, in liver organoids, markers include HHEX, SOX2, HNF4A, AFP, and ALB; in pancreatic organoids, markers include PDX1, SOX17, and SOX9; in organoids that differentiate into the intestine, markers include CDX2 and SOX9; and in kidney organoids, markers include Pax2 and Six2.
目的の細胞またはオルガノイドを得るための分化誘導方法は、公知の方法を用いることができる。例えば、多能性幹細胞から神経堤細胞への分化誘導は、Fukuta M. et al., PLoS One, 2014, 9(12): e112291やKamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47に記載の方法などにより、行うことができる。具体的には、多能性幹細胞を培養容器に播種して接着培養(足場材料を用いた浮遊培養)した後、TGFβ阻害剤及びGSK3β阻害剤を含む培地で接着培養(足場材料を用いた浮遊培養)することで神経堤細胞に分化させることができる。 A known method can be used for inducing differentiation to obtain the desired cells or organoids. For example, differentiation of pluripotent stem cells into neural crest cells can be induced by the methods described in Fukuta M. et al., PLoS One, 2014, 9(12): e112291 and Kamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47. Specifically, pluripotent stem cells can be seeded in a culture vessel and cultured as adherent cells (suspension culture using a scaffold material), and then differentiated into neural crest cells by culture as adherent cells (suspension culture using a scaffold material) in a medium containing a TGFβ inhibitor and a GSK3β inhibitor.
神経堤細胞から、間葉系幹細胞、神経前駆細胞、神経細胞、グリア細胞、骨細胞、軟骨細胞、角膜細胞、メラノサイトなどの細胞を製造することもできる。例えば、これらの細胞への分化誘導は、Fukuta M. et al., PLoS One, 2014, 9(12): e112291、Horikiri T. et al., PLoS One, 2017, 12(1): e0170342、Kamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47に記載の方法などに基づいて行うことができる。具体的には、例えば、神経堤細胞をFibronectinでコーティングしたプレートに播種し、N-2 Supplement、BDNF、GDNF、NT-3、NGFを添加したDMEM/F12に交換し、37℃、5%CO2下で約14日間培養することで、神経前駆細胞および神経細胞を得ることができる。あるいは、神経堤細胞をプレートに播種し、SB431542およびCHIR99021を含むCDM培地で1日間培養した後、B-27 Supplement、N-2 Supplement、L-glutamine、Penicillin/Streptomycin、BDNF、GDNF、NT-3、NGFを添加したNeurobasal mediumに交換し、37℃、5%CO2下で約35日間培養することで、神経前駆細胞および神経細胞を得ることができる。 From neural crest cells, it is also possible to produce cells such as mesenchymal stem cells, neural progenitor cells, nerve cells, glial cells, bone cells, chondrocytes, corneal cells, and melanocytes. For example, differentiation into these cells can be induced based on the methods described in Fukuta M. et al., PLoS One, 2014, 9(12): e112291, Horikiri T. et al., PLoS One, 2017, 12(1): e0170342, and Kamiya D, et al., NPJ Regen Med., 2022 Sep 15;7(1):47. Specifically, for example, neural crest cells are seeded onto a plate coated with fibronectin, the medium is replaced with DMEM/F12 supplemented with N-2 Supplement, BDNF, GDNF, NT-3, and NGF, and the cells are cultured at 37°C under 5% CO2 for approximately 14 days to obtain neural progenitor cells and neurons. Alternatively, neural crest cells can be plated and cultured in CDM medium containing SB431542 and CHIR99021 for one day, after which the medium is replaced with Neurobasal medium supplemented with B-27 Supplement, N-2 Supplement, L-glutamine, Penicillin/Streptomycin, BDNF, GDNF, NT-3, and NGF, and cultured at 37°C in 5% CO2 for approximately 35 days to obtain neural progenitor cells and neurons.
また、間葉系間質細胞への分化誘導は、例えば、次の方法などが挙げられる。神経堤細胞を培養容器に播種し、SB431542およびCHIR99021を含むCDM培地で1日間培養する。1日後、培地を、FBSを含むαMEMに交換する。分化誘導開始約14日後に、間葉系間質細胞を得ることができる。 In addition, the following method can be used to induce differentiation into mesenchymal stromal cells. Neural crest cells are seeded in a culture vessel and cultured for one day in CDM medium containing SB431542 and CHIR99021. After one day, the medium is replaced with αMEM containing FBS. Mesenchymal stromal cells can be obtained about 14 days after the start of differentiation induction.
多能性幹細胞をT細胞に分化させる方法として、例えば、(1)多能性幹細胞を、造血前駆細胞に分化させる工程、及び(2)該造血前駆細胞をT細胞に分化させる工程を含む方法が挙げられる。前記工程(1)は、例えば、WO2013/075222、WO2016/076415、Liu S.et al., Cytotherapy, 17 (2015); 344-358などに記載されているように、造血前駆細胞への誘導培地中で多能性幹細胞を培養する工程であり得る。また、前記工程(2)は、WO2016/076415などに記載されているような、(2-1)造血前駆細胞からCD4CD8両陽性T細胞を誘導する工程、及び(2-2)CD4CD8両陽性T細胞からCD8陽性T細胞を誘導する工程であり得る。 A method for differentiating pluripotent stem cells into T cells includes, for example, a method comprising (1) a step of differentiating pluripotent stem cells into hematopoietic progenitor cells, and (2) a step of differentiating the hematopoietic progenitor cells into T cells. The step (1) may be, for example, a step of culturing pluripotent stem cells in an induction medium for hematopoietic progenitor cells, as described in WO2013/075222, WO2016/076415, Liu S. et al., Cytotherapy, 17 (2015); 344-358, etc. In addition, the step (2) may be, for example, a step (2-1) of inducing CD4CD8 bi-positive T cells from hematopoietic progenitor cells, and a step (2-2) of inducing CD8 positive T cells from CD4CD8 bi-positive T cells, as described in WO2016/076415, etc.
多能性幹細胞を血小板に分化誘導させる工程は、例えば、(1)多能性幹細胞を造血前駆細胞に分化させる工程、および造血前駆細胞を血小板に分化させる工程を含む方法が挙げられる。前記工程(2)は、例えば、WO2012/157586、US2014/127815、Nakamura, Eto, et al. Cell Stem Cell 14, 535-548 (2014)などに記載されているように、TPOおよび/またはSCFを含む培地で造血前駆細胞を7~15日間程度培養する工程であり得る。この工程により、巨核球および血小板を含む細胞集団を得ることができる。 The step of inducing differentiation of pluripotent stem cells into platelets can be, for example, (1) a method including a step of differentiating pluripotent stem cells into hematopoietic progenitor cells and a step of differentiating hematopoietic progenitor cells into platelets. The step (2) can be, for example, a step of culturing hematopoietic progenitor cells in a medium containing TPO and/or SCF for about 7 to 15 days, as described in WO2012/157586, US2014/127815, Nakamura, Eto, et al. Cell Stem Cell 14, 535-548 (2014), etc. This step can produce a cell population containing megakaryocytes and platelets.
多能性幹細胞を始原生殖細胞に分化誘導させる工程は、例えば、WO2017/002888などに記載されているように、(1)多能性幹細胞を、アクチビンAおよびGSK3β阻害剤を含む培養液中で40時間~60時間程度培養してエピブラスト様細胞に分化させる工程、および(2)エピブラスト様細胞を、BMPを含む培養液中で4~8日間程度培養して始原生殖細胞(始原生殖細胞様細胞)に分化させる工程を含む方法が挙げられる。 The process of inducing differentiation of pluripotent stem cells into primordial germ cells includes, for example, a method comprising the steps of: (1) culturing pluripotent stem cells in a culture medium containing activin A and a GSK3β inhibitor for approximately 40 to 60 hours to differentiate them into epiblast-like cells; and (2) culturing the epiblast-like cells in a culture medium containing BMP for approximately 4 to 8 days to differentiate them into primordial germ cells (primordial germ cell-like cells), as described in WO2017/002888 and the like.
多能性幹細胞から心筋細胞への分化誘導としては、例えば、WO2015/141827、Laflamme MA and Murry CE, Nature. 473(7347):326-35 (2011)などに記載の方法などが挙げられる。この他にも、例えば、人工多能性幹細胞を浮遊培養により胚様体を形成させて心筋細胞を製造する方法、BMPシグナル伝達を抑制する物質の存在下で心筋細胞を製造する方法(WO2005/033298)、Activin AとBMPを順に添加させて心筋細胞を製造する方法(WO2007/002136)、カノニカル(古典的)Wntシグナル経路の活性化を促す物質の存在下で心筋細胞を製造する方法(WO2007/126077)などが挙げられる。典型的には、例えば、心筋細胞のマーカータンパク質としては、NKX2.5(心筋特異的転写因子)、TNNT2(トロポニンT)が、心筋前駆細胞のマーカータンパク質としては、KDR(血管内皮細胞増殖因子(VEGF)の受容体)やISL1(LIMホメオドメイン転写因子)が挙げられる。 Examples of methods for inducing differentiation of pluripotent stem cells into cardiomyocytes include the methods described in WO2015/141827 and Laflamme MA and Murry CE, Nature. 473(7347):326-35 (2011). Other examples include a method for producing cardiomyocytes by forming embryoid bodies through suspension culture of induced pluripotent stem cells, a method for producing cardiomyocytes in the presence of a substance that suppresses BMP signaling (WO2005/033298), a method for producing cardiomyocytes by sequentially adding Activin A and BMP (WO2007/002136), and a method for producing cardiomyocytes in the presence of a substance that promotes activation of the canonical (classical) Wnt signaling pathway (WO2007/126077). Typically, for example, marker proteins for cardiomyocytes include NKX2.5 (a cardiac muscle-specific transcription factor) and TNNT2 (troponin T), while marker proteins for cardiac progenitor cells include KDR (a receptor for vascular endothelial growth factor (VEGF)) and ISL1 (a LIM homeodomain transcription factor).
また、複数種類の細胞を用いて、オルガノイドを製造することもできる。例えば、肝オルガノイドの場合には、WO2013/047639などに記載されているように、多能性幹細胞から、肝前駆細胞(臓器細胞)、間葉系幹細胞、および血管内皮細胞を誘導し、これらの混合物を浮遊培養することで、肝オルガノイドを製造することができる。 It is also possible to produce organoids using multiple types of cells. For example, in the case of liver organoids, as described in WO2013/047639, liver progenitor cells (organ cells), mesenchymal stem cells, and vascular endothelial cells can be induced from pluripotent stem cells, and the mixture can be cultured in suspension to produce liver organoids.
本発明の製造方法(II)は、全部または一部の期間、フィーダーフリー条件下および/またはゼノフリー条件下での培養であってもよい。臨床での使用の観点からは、本発明の分化誘導法は、全期間がフィーダーフリーかつゼノフリー条件下で行われることが好ましい。 The production method (II) of the present invention may involve culturing under feeder-free conditions and/or xeno-free conditions for all or part of the period. From the viewpoint of clinical use, it is preferable that the differentiation induction method of the present invention is carried out under feeder-free and xeno-free conditions for the entire period.
また、別の態様において、本発明の製造方法(II)により得られた細胞またはオルガノイド(以下、「本発明の分化細胞またはオルガノイド」とも称することがある。)が提供される。 In another aspect, there is provided a cell or organoid obtained by the production method (II) of the present invention (hereinafter, also referred to as the "differentiated cell or organoid of the present invention").
本発明の製造方法(II)は、得られた目的の細胞またはオルガノイドを回収する工程を含んでいてもよい。回収した細胞は、細胞凍結保存液を用いて凍結保存してもよい。また、容器に回収した細胞は、セルカンターにてセルカウントを行ってもよく、細胞の表面マーカーに対する抗体により標識し、フローサイトメトリーやマスサイトメトリー、磁気細胞分離法などにより精製してもよい。 The manufacturing method (II) of the present invention may include a step of recovering the obtained target cells or organoids. The recovered cells may be cryopreserved using a cell cryopreservation solution. The cells recovered in the container may be subjected to cell counting using a cell counter, or may be labeled with an antibody against a cell surface marker and purified by flow cytometry, mass cytometry, magnetic cell separation, or the like.
本発明の分化誘導法における、用いられる細胞、培養期間や用いられる培地の種類を含む培養方法や培養条件等の具体例や定義等は、上記「1.人工多能性幹細胞の製造方法」で記載した内容がすべて援用される。 Specific examples and definitions of the cells used, the culture method and culture conditions including the culture period and the type of medium used in the differentiation induction method of the present invention are all those described in "1. Method for producing induced pluripotent stem cells" above.
当該製造方法(II)の好ましい態様では、図9(a)に示した工程s5は、iPS細胞を、外胚葉系細胞、中胚葉系細胞、または内胚葉系細胞へと分化誘導する工程s5aであってもよい。また、当該製造方法(II)の好ましい他の態様では、図9(b)に示すように、前記工程s5aの後に、さらなる分化誘導の工程s5bを有してもよい。該工程s5bは、前記工程s5aで得られた外胚葉系細胞等に対して分化誘導を行い、さらなる他の細胞を得る工程である。 In a preferred embodiment of the manufacturing method (II), step s5 shown in FIG. 9(a) may be step s5a of inducing differentiation of iPS cells into ectodermal cells, mesodermal cells, or endodermal cells. In another preferred embodiment of the manufacturing method (II), as shown in FIG. 9(b), step s5a may be followed by step s5b of further differentiation induction. Step s5b is a step of inducing differentiation of the ectodermal cells, etc. obtained in step s5a to obtain further other cells.
図9(b)に示すように、前記工程s5aが実施される容器C1の後には、さらなる分化誘導の工程s5bを実施するための容器C2が、接続管路Jc1を介してさらに接続される。該容器C2は、1以上の開閉可能な入出用ポート(符号は省略する)を有する。該入出用ポートを通じて、工程s5bの分化誘導に必要な材料が該容器C2の内部に供給される。送り機構Fc1によって、容器C1から容器C2へと、細胞(前記の外胚葉系細胞、中胚葉系細胞、または内胚葉系細胞)が移動し、該容器C2において工程s5bが実施される。 As shown in FIG. 9(b), a container C2 for carrying out a further differentiation induction step s5b is further connected via a connecting pipe Jc1 after the container C1 in which the step s5a is carried out. The container C2 has one or more openable and closable input/output ports (reference numbers omitted). Materials necessary for the differentiation induction in step s5b are supplied to the inside of the container C2 through the input/output ports. The cells (the ectodermal, mesodermal, or endodermal cells) are moved from the container C1 to the container C2 by a feed mechanism Fc1, and the step s5b is carried out in the container C2.
前記工程s5bの後には、必要に応じて、さらなる分化誘導の工程が追加されてもよく、その場合には、追加される工程に対応付けられた容器が上記説明と同様に接続される。 After step s5b, a further differentiation induction step may be added as necessary, in which case the containers corresponding to the added step are connected in the same manner as described above.
(未分化細胞の除去)
当該製造方法(II)の好ましい態様では、上記工程s5の後に、未分化細胞を除去する工程s6がさらに加えられ、該工程s6に対応付けられた容器D1が上記説明と同様に接続される。図10に示すように、上記分化誘導の工程s5(上記工程s5a、s5bを含む)で用いられた容器のうち最後尾の容器X2(例えば、容器C1またはC2)の後に、該工程s6を実施するための容器D1が、接続管路Jx2を介してさらに接続される。送り機構Fx2によって、容器X2から容器D1へと分化細胞が移動する。容器D1は、1以上の開閉可能な入出用ポートを有し、該入出用ポートを通じて、該工程s6に必要な材料が容器D1内へと供給され、該工程s6が実施される。これにより、容器D1内には、未分化細胞が除去された分化細胞が残る。
(Removal of undifferentiated cells)
In a preferred embodiment of the manufacturing method (II), a step s6 of removing undifferentiated cells is added after the step s5, and a container D1 associated with the step s6 is connected in the same manner as described above. As shown in FIG. 10, a container D1 for carrying out the step s6 is further connected through a connecting pipe Jx2 after the last container X2 (e.g., container C1 or C2) among the containers used in the differentiation induction step s5 (including the steps s5a and s5b). Differentiated cells are moved from the container X2 to the container D1 by the feed mechanism Fx2. The container D1 has one or more openable inlet/outlet ports, and materials necessary for the step s6 are supplied into the container D1 through the inlet/outlet ports, and the step s6 is carried out. As a result, differentiated cells from which undifferentiated cells have been removed remain in the container D1.
工程s6で用いられる容器、入出用ポート、接続管路、送り機構のそれぞれの構成は、上記工程s1~s5の説明で示した構成を参照することができる。 The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in step s6 can be referenced from the configurations described in steps s1 to s5 above.
(未分化細胞を除去する方法)
本発明の製造方法(II)において、未分化細胞の除去方法は、該製造方法により製造される細胞以外の細胞を除去し得れば特に限定されず、自体公知の未分化細胞除去剤等を培地中に添加することで行い得る(例えば、Di Mao., et al. Angewandte Chemie International Edition; 9 January 2017、Ben-David, U., et al. Cell Stem Cell, 12, 167 (2013)、WO2019/187918、特開2016-93178、Yoshiki Nakashima, et. al., Molecular Therapy Vol. 26 No 7 July 2018等)。
(Method for Removing Undifferentiated Cells)
In the production method (II) of the present invention, the method for removing undifferentiated cells is not particularly limited as long as it can remove cells other than the cells produced by the production method, and can be performed by adding a known undifferentiated cell removing agent or the like to the medium (for example, Di Mao., et al. Angewandte Chemie International Edition; 9 January 2017, Ben-David, U., et al. Cell Stem Cell, 12, 167 (2013), WO2019/187918, JP2016-93178, Yoshiki Nakashima, et. al., Molecular Therapy Vol. 26 No. 7 July 2018, etc.).
(品質検査)
当該製造方法(II)の好ましい態様では、上記工程s6の後に、分化細胞の品質検査を行うためのサンプルを取り出す工程s7がさらに加えられ、該工程s7に対応付けられた容器E1が上記説明と同様に接続される。図10に示すように、上記した容器D1の後には、該工程s7を実施するための容器E1が、接続管路Jd1を介してさらに接続される。該容器E1は、1以上の開閉可能な入出用ポートを有する。送り機構Fd1によって、容器D1から容器E1へと内容物が移動し、該容器E1の入出用ポートを通じて、検査に必要なサンプルが外部に取り出される。また、品質検査用のサンプルは、上記工程s6の後に、容器D1から取り出してもよい。
(Quality Inspection)
In a preferred embodiment of the manufacturing method (II), step s7 is added after step s6 to take out a sample for quality testing of differentiated cells, and a container E1 associated with step s7 is connected in the same manner as described above. As shown in FIG. 10, a container E1 for carrying out step s7 is further connected after the container D1 via a connecting pipe Jd1. The container E1 has one or more openable and closable input/output ports. The feed mechanism Fd1 moves the contents from the container D1 to the container E1, and the sample required for testing is taken out through the input/output port of the container E1. The sample for quality testing may be taken out of the container D1 after step s6.
工程s7で用いられる容器、入出用ポート、接続管路、送り機構のそれぞれの構成は、上記工程s1~s6の説明で示した構成を参照することができる。 The configurations of the container, inlet/outlet port, connecting pipeline, and feed mechanism used in step s7 can be referenced from the configurations described in steps s1 to s6 above.
品質検査の目的は、本発明の製造方法(II)で製造された細胞やオルガノイド等が所望するものであるかを確認することである。品質検査の検査項目は、特に限定はされないが、細胞やオルガノイドの形態や細胞表面マーカー等の発現の有無、無菌試験、エンドトキシン試験、細胞生存率の評価などの基本的な試験などが挙げられ、各項目に応じた検査装置が利用可能である。 The purpose of the quality test is to confirm whether the cells, organoids, etc. produced by the production method (II) of the present invention are the desired ones. The test items for the quality test are not particularly limited, but include basic tests such as the morphology of the cells or organoids, the presence or absence of expression of cell surface markers, sterility tests, endotoxin tests, and evaluation of cell viability, and testing equipment suitable for each item can be used.
(ユーザーへの分化細胞の提供)
工程s7が完了し、細胞が良品であるとの認定を受けた後、容器E1に接続された接続管路や配管などを除去して、容器E1と内部の分化細胞(細胞懸濁液になっている)をそのままユーザー(細胞療法を受ける患者等)に提供(出荷等)してもよいし、シリンジ型の容器やバイアルなどに移してユーザーに提供してもよい。また、容器E1の入出用ポートに分化細胞を取り出すためシリンジを接続した状態で、ユーザーに提供してもよい。また、後述するような医薬等として提供してもよい。
(Provision of differentiated cells to users)
After step s7 is completed and the cells are certified as non-defective, the connecting lines and piping connected to the container E1 may be removed, and the container E1 and the differentiated cells (in the form of a cell suspension) therein may be provided (shipped, etc.) as is to a user (such as a patient undergoing cell therapy), or may be transferred to a syringe-type container or vial and provided to the user. Also, the container E1 may be provided to the user with a syringe connected to the inlet/outlet port for removing the differentiated cells. Also, the container E1 may be provided as a medicine, etc., as described below.
3.分化細胞またはオルガノイドの用途
本発明の分化細胞またはオルガノイドは、免疫療法や再生医療に好適に用いることができるため、別の態様において、本発明の分化細胞またはオルガノイドを含有してなる医薬(以下、「本発明の医薬」と称することがある。)が提供される。本発明の医薬としては、例えば、免疫療法剤や細胞移植剤の形態の剤で提供される。また、本発明の分化細胞またはオルガノイドの有効量を、治療の対象とする霊長類動物に投与または移植する、疾患の治療方法も、本発明に包含される。霊長類動物の具体例は、上記「1.多能性幹細胞の製造方法」に記載した通りであるが、好ましくはヒトである。
3. Uses of Differentiated Cells or Organoids The differentiated cells or organoids of the present invention can be suitably used in immunotherapy and regenerative medicine, so in another aspect, a pharmaceutical comprising the differentiated cells or organoids of the present invention (hereinafter, sometimes referred to as the "pharmaceutical of the present invention") is provided. The pharmaceutical of the present invention is provided, for example, in the form of an immunotherapy agent or a cell transplantation agent. In addition, the present invention also includes a method for treating a disease in which an effective amount of the differentiated cells or organoids of the present invention is administered or transplanted into a primate to be treated. Specific examples of primates are as described above in "1. Method for producing pluripotent stem cells", but are preferably humans.
本発明の分化細胞またはオルガノイドは、それを必要とする対象に生体内に投与または移植して用いることができる。移植は、細胞を一定の位置で固定できる生体内領域に行うことが好ましく、例えば、皮下、腹腔内、腹膜上皮、大網、脂肪組織、筋肉組織や膵臓、腎臓等の各臓器の被膜下などに行うことできる。好ましくは、侵襲度の低い皮下移植である。移植される細胞は、治療上有効量を投与すればよく、移植対象の、年齢、体重、移植部位の大きさ、疾患の重篤度等の要因により変化し得、特に限定されないが、例えば、10×104細胞~10×1011細胞程度とすることができる。 The differentiated cells or organoids of the present invention can be administered or transplanted into the body of a subject in need thereof. The transplantation is preferably performed in a region of the body where the cells can be fixed at a certain position, for example, subcutaneously, intraperitoneally, peritoneal epithelium, omentum, adipose tissue, muscle tissue, or under the capsule of each organ such as the pancreas or kidney. Preferably, the transplantation is subcutaneous, which is less invasive. The cells to be transplanted may be administered in a therapeutically effective amount, which may vary depending on factors such as the age, weight, size of the transplantation site, and severity of the disease of the transplantation subject, and are not particularly limited, but may be, for example, about 10 x 10 4 cells to 10 x 10 11 cells.
本発明の分化細胞またはオルガノイドを、医薬として用いる場合、拒絶反応が起こらないという観点から、移植先の個体のHLA遺伝子型が同一若しくは実質的に同一である体細胞から樹立したiPS細胞に由来する細胞またはオルガノイドを用いることが望ましい。ここで、「実質的に同一」とは、移植した細胞に対して免疫抑制剤により免疫反応が抑制できる程度にHLA遺伝子型が一致していることであり、例えば、HLA-A、HLA-BおよびHLA-DRの3遺伝子座あるいはHLA-Cを加えた4遺伝子座が一致するHLA型を有する体細胞である。年齢や体質などの理由から充分な細胞が得られない場合には、ポリエチレングリコールやシリコーンのようなカプセル、多孔性の容器などに包埋して拒絶反応を回避した状態で移植することも可能である。 When the differentiated cells or organoids of the present invention are used as medicines, it is desirable to use cells or organoids derived from iPS cells established from somatic cells with the same or substantially the same HLA genotype of the recipient individual, from the viewpoint of preventing rejection reactions. Here, "substantially the same" means that the HLA genotype matches the transplanted cells to such an extent that the immune response can be suppressed with an immunosuppressant, for example, somatic cells with an HLA type that matches the three loci of HLA-A, HLA-B, and HLA-DR, or four loci including HLA-C. If sufficient cells cannot be obtained due to age, constitution, or other reasons, they can be embedded in capsules such as polyethylene glycol or silicone, or in porous containers, to avoid rejection reactions, and then transplanted.
本発明の分化細胞またはオルガノイドは、常套手段にしたがって医薬上許容される担体と混合するなどして、注射剤、懸濁剤、点滴剤等の非経口製剤として製造される。従って、一態様において、本発明の分化細胞またはオルガノイドを製剤化する工程を含む、免疫療法剤または細胞移植療法剤の製法も提供される。かかる製法は、本発明の分化細胞またはオルガノイドを準備する工程を含んでいてもよい。さらに、本発明の分化細胞またはオルガノイドを保存する工程を含むこともできる。 The differentiated cells or organoids of the present invention are prepared as parenteral preparations such as injections, suspensions, or drops by mixing with a medicamentically acceptable carrier according to conventional methods. Thus, in one embodiment, there is also provided a method for producing an immunotherapy agent or cell transplantation therapy agent, which includes a step of formulating the differentiated cells or organoids of the present invention. Such a method may include a step of preparing the differentiated cells or organoids of the present invention. Furthermore, it may also include a step of preserving the differentiated cells or organoids of the present invention.
当該非経口製剤に含まれ得る医薬上許容される担体としては、例えば、生理食塩水、ブドウ糖やその他の補助薬を含む等張液(例えば、D-ソルビトール、D-マンニトール、塩化ナトリウムなど)などの注射用の水性液を挙げることができる。本発明の細胞は、例えば、緩衝剤(例えば、リン酸塩緩衝液、酢酸ナトリウム緩衝液)、無痛化剤(例えば、塩化ベンザルコニウム、塩酸プロカインなど)、安定剤(例えば、ヒト血清アルブミン、ポリエチレングリコールなど)、保存剤、酸化防止剤などと配合してもよい。 Examples of pharma- ceutically acceptable carriers that may be included in the parenteral formulation include aqueous solutions for injection, such as physiological saline, isotonic solutions containing glucose and other adjuvants (e.g., D-sorbitol, D-mannitol, sodium chloride, etc.). The cells of the present invention may be compounded with, for example, buffers (e.g., phosphate buffer, sodium acetate buffer), soothing agents (e.g., benzalkonium chloride, procaine hydrochloride, etc.), stabilizers (e.g., human serum albumin, polyethylene glycol, etc.), preservatives, antioxidants, etc.
本発明の免疫療法剤または細胞移植療法剤は、細胞の凍結保存に通常使用される条件で凍結保存された状態で提供され、用時融解して用いることもできる。その場合、血清若しくはその代替物、有機溶剤(例、DMSO)等をさらに含んでいてもよい。この場合、血清若しくはその代替物の濃度は、特に限定されるものではないが約1~約30%(v/v)、好ましくは約5~約20%(v/v)であり得る。有機溶剤の濃度は、特に限定されるものではないが0~約50%(v/v)、好ましくは約5~約20%(v/v)であり得る。 The immunotherapy agent or cell transplantation therapy agent of the present invention is provided in a frozen state under conditions normally used for cryopreservation of cells, and can be thawed when used. In this case, it may further contain serum or a substitute thereof, an organic solvent (e.g., DMSO), etc. In this case, the concentration of serum or a substitute thereof is not particularly limited, but may be about 1 to about 30% (v/v), preferably about 5 to about 20% (v/v). The concentration of the organic solvent is not particularly limited, but may be 0 to about 50% (v/v), preferably about 5 to about 20% (v/v).
また、本発明の分化細胞またはオルガノイドは、疾患の治療または予防に有用な薬剤である候補薬剤をスクリーニングする方法にも用いることができる。 The differentiated cells or organoids of the present invention can also be used in methods for screening candidate drugs that are useful for treating or preventing disease.
4.細胞製造装置
次に、本発明による細胞製造装置(以下、当該装置ともいう)の構成を説明する。当該装置の全体的な特徴や、各容器、入出用ポート、接続管路、送り機構のそれぞれの構成は、上記製造方法(I)、(II)の説明で詳細に述べたとおりであり、ここでは詳細な説明を省略する。
4. Cell manufacturing device Next, the configuration of the cell manufacturing device according to the present invention (hereinafter, also referred to as the device) will be described. The overall features of the device and the configurations of each vessel, inlet/outlet port, connecting pipeline, and feed mechanism are as described in detail in the explanation of the above manufacturing methods (I) and (II), so detailed explanations will be omitted here.
当該装置は、
iPS細胞を製造する部分(以下、iPS細胞製造部ともいう)、
iPS細胞を拡大培養する部分(以下、拡大培養部ともいう)、
iPS細胞を分化誘導して分化細胞を形成する部分(以下、分化誘導部ともいう)、
未分化細胞を除去する部分(以下、未分化細胞除去部ともいう)、
分化細胞を検査する部分(以下、検査部ともいう)
に分けることができる。
The device comprises:
The part that produces iPS cells (hereinafter referred to as the iPS cell production department),
A portion for expanding iPS cells (hereinafter also referred to as the expansion culture portion),
A part for inducing differentiation of iPS cells to form differentiated cells (hereinafter also referred to as a differentiation induction part),
A part for removing undifferentiated cells (hereinafter also referred to as an undifferentiated cell removing part),
The part where differentiated cells are examined (hereinafter referred to as the examination part)
It can be divided into:
(iPS細胞製造部)
当該装置におけるiPS細胞製造部は、上記製造方法(I)においてn=3の場合の工程を実施する部分である。図11に示すように、当該装置は、iPS細胞製造部として、上記工程s1を実施するための容器A1と、上記工程s2を実施するための容器A2と、上記工程s3を実施するための容器A3とを有する。各容器A1~A3は、接続管路J1、J2をそれぞれ介して、前記工程の順に直列に接続されているか、または前記工程の順に直列に接続可能な状態となっている。各容器は、連通状態に切り替えられた接続管路J1、J2をそれぞれ通じて、内容物を次段の容器へと移動させる送り機構F1を有する。各材料供給源G1~G3は、それぞれの容器で実施される工程に応じて適宜に選択される。
(iPS cell manufacturing department)
The iPS cell production section in the device is a section that performs the step in the above-mentioned production method (I) when n=3. As shown in FIG. 11, the device performs the above-mentioned step s1 as the iPS cell production section. The containers A1 to A3 have connecting pipes J1 and J2, respectively. The containers are connected in series in the order of the steps through the connecting pipes J1 and J2, respectively, which are switched to a communicating state. The container has a feed mechanism F1 for moving the contents to the next container. Each of the material supply sources G1 to G3 is appropriately selected according to the process to be carried out in the respective container.
iPS細胞製造部を用いて、工程s1~s3を順に実施し、体細胞からiPS細胞を製造する操作については、上記製造方法(I)で説明したとおりである。 The steps s1 to s3 are carried out in order using the iPS cell production section to produce iPS cells from somatic cells, as described in the production method (I) above.
(容器の数)
上記製造方法(I)では、液体培地中の初期化因子の濃度を低減させる工程s2が複数の処理段階(複数の工程)に分かれていてもよく、その場合には、工程s2のために複数の容器A2~A(n-1)を用いることができる。
これに対して、当該装置では、好ましい態様の1つとして、工程s2が単一の工程であって、該工程s2を実施するための単一の容器A2を有する構成となっている。
しかしながら、当該装置においても該工程s2は複数の処理段階(複数の工程)に分かれていてもよく、その場合には、図1に示す構成と同様に、該工程s2のために複数の容器A2~A(n-1)を必要な数だけ追加し挿入することができる。さらなる容器が工程s2のために挿入されても、また、容器A1~A3の直列接続の間に付加的な工程のための他の容器が挿入されても、容器A1~A3は直列に接続されているものとする。
(Number of containers)
In the above-mentioned production method (I), step s2 of reducing the concentration of the reprogramming factor in the liquid medium may be divided into multiple processing steps (multiple steps), in which case multiple containers A2 to A(n-1) can be used for step s2.
In contrast, in one preferred embodiment of the apparatus, step s2 is a single step, and the apparatus has a configuration including a single vessel A2 for carrying out step s2.
However, in this apparatus, step s2 may be divided into a plurality of processing stages (a plurality of steps), in which case a necessary number of containers A2 to A(n-1) can be added and inserted for step s2, similar to the configuration shown in Fig. 1. Even if further containers are inserted for step s2, or if other containers for additional steps are inserted between the series connection of containers A1 to A3, containers A1 to A3 are still considered to be connected in series.
(拡大培養部)
当該装置における拡大培養部は、上記製造方法(I)における工程s4を実施するための部分である。図8に示すように、当該装置は、拡大培養部として、iPS細胞を液体培地中でp回拡大培養する工程s4を実施するためのp個の容器B1~Bpをさらに有する。ここで、pは1以上の整数、即ち、p≧1である。容器B1~Bpのそれぞれは、1以上の開閉可能な入出用ポートを有する。
(Expansion Culture Section)
The expansion culture section in the device is a section for carrying out step s4 in the above-mentioned production method (I). As shown in Fig. 8, the device further has p containers B1 to Bp as the expansion culture section for carrying out step s4 of expanding iPS cells in a liquid medium p times. Here, p is an integer of 1 or more, i.e., p ≥ 1. Each of the containers B1 to Bp has one or more inlet/outlet ports that can be opened or closed.
拡大培養の回数(p回)が1回の場合と2回以上の場合における、それぞれの容器の接続の態様は次のとおりである。
(i)図8の例において、拡大培養の回数pが1である場合、用いられる容器は容器B1だけである。容器B1は、接続管路Jnを介して、容器A3に接続されているか、または接続可能な状態となっている。当該装置は、連通状態に切り替えられた接続管路Jnを通じて、容器A3の内容物を容器B1へと移動させる送り機構Fnを有する。
(ii)図8の例において、拡大培養の回数pが2以上である場合、用いられる容器は、2個以上の容器B1~Bpである。容器B1は、接続管路を介して、容器A3に接続されているか、または接続可能な状態となっている。密閉容器B1~Bpは、それぞれ、接続管路Jb1~Jb(p-1)を介して、工程s4の順に直列に接続されているか、または接続可能な状態となっている。当該装置は、連通状態に切り替えられた接続管路Jn、および、Jb1~Jb(p-1)をそれぞれ通じて、容器A3の内容物を容器B1~Bpへと順に移動させる送り機構Fn、およびFb1~Fb(p-1)を有する。
The connection modes of the containers when the number of times of expansion culture (p times) is one and when it is two or more are as follows.
(i) In the example of Fig. 8, when the number of times p of expansion culture is 1, the only container used is container B1. Container B1 is connected or connectable to container A3 via a connecting pipe Jn. The device has a feed mechanism Fn that moves the contents of container A3 to container B1 through the connecting pipe Jn that is switched to a communicating state.
(ii) In the example of Fig. 8, when the number p of expansion cultures is 2 or more, two or more containers B1 to Bp are used. Container B1 is connected to container A3 via a connecting pipeline, or is in a state where it can be connected. The sealed containers B1 to Bp are connected in series in the order of step s4 via connecting pipelines Jb1 to Jb(p-1), respectively, or are in a state where they can be connected. The device has a feed mechanism Fn and Fb1 to Fb(p-1) that move the contents of container A3 to containers B1 to Bp in order through connecting pipelines Jn, which is switched to a communicating state, and Jb1 to Jb(p-1), respectively.
拡大培養部において工程s4を実施する操作については、上記製造方法(I)で説明したとおりである。iPS細胞は、工程s1~s3を実施することで得られたものであってもよいし、既に準備された(例えば、市販)iPS細胞であってもよい。 The procedure for carrying out step s4 in the expansion culture section is as described in the above manufacturing method (I). The iPS cells may be those obtained by carrying out steps s1 to s3, or they may be iPS cells that have already been prepared (e.g., commercially available).
(分化誘導部)
当該装置における分化誘導部は、上記製造方法(II)における工程s5を実施するための部分である。上記製造方法(II)の説明では、図9を参照して、容器C1、C2を用いた例を具体的に示したが、当該装置の分化誘導部では、図12に示すように、該工程s5を実施するためのq個の密閉容器C1~Cqがさらに用いられる。ここで、qは1以上の整数、即ち、q≧1である。容器C1~Cqのそれぞれは、1以上の開閉可能な入出用ポートを有する。
(Differentiation Induction Part)
The differentiation induction section of the device is a section for carrying out step s5 in the above manufacturing method (II). In the explanation of the above manufacturing method (II), an example using containers C1 and C2 was specifically shown with reference to Fig. 9, but the differentiation induction section of the device further uses q sealed containers C1 to Cq for carrying out step s5, as shown in Fig. 12. Here, q is an integer of 1 or more, that is, q ≥ 1. Each of the containers C1 to Cq has one or more openable/closable inlet/outlet ports.
分化細胞を形成するための容器の数(即ち、順次分化誘導する工程の数)が1回の場合と2回以上の場合における、それぞれの容器の接続の態様は次のとおりである。
(i)図12の例において、q個の容器が1個の容器C1である場合、該容器C1は、接続管路Jn(またはJbp)を介して、容器A3に対して(もしくは容器B1~Bpのうちの最後尾の容器Bpに対して)、接続されているかまたは接続可能な状態となっている。当該装置は、連通状態に切り替えられた接続管路Jn(またはJbp)を通じて、容器A3(または容器Bp)の内容物を容器C1へと移動させる送り機構Fn(または)Fbnを有する。
(ii)図12の例において、q個の容器が2個以上の容器C1~Cqである場合、該容器C1~Cqは、接続管路を介して、工程s5の順に直列に接続されているか、または接続可能な状態となっている。該容器C1は、接続管路Jn(またはJbp)を介して、容器A3(または、容器B1~Bpのうちの最後尾の容器Bp)に対して、接続されているかまたは接続可能な状態となっている。当該装置の分化誘導部は、連通状態に切り替えられた接続管路Jn(またはJbp)を通じて、およびJc1~Jc(q-1)を通じて、容器A3の内容物(または容器Bpの内容物)を、容器C1~Cqへと順に移動させる送り機構Fn(またはFbp)および送り機構Jc1~Jc(q-1)を有する。
When the number of containers for forming differentiated cells (i.e., the number of steps for sequentially inducing differentiation) is one time and when it is two or more times, the connection modes of the respective containers are as follows.
12, when the q containers are one container C1, the container C1 is connected or connectable to the container A3 (or to the last container Bp among the containers B1 to Bp) via a connecting pipe Jn (or Jbp). The device has a feed mechanism Fn (or) Fbn that moves the contents of the container A3 (or the container Bp) to the container C1 through the connecting pipe Jn (or Jbp) that is switched to a communicating state.
(ii) In the example of Figure 12, when the q containers are two or more containers C1 to Cq, the containers C1 to Cq are connected in series or are connectable through the connecting pipeline in the order of step s5. The container C1 is connected or is connectable to the container A3 (or the last container Bp among the containers B1 to Bp) through the connecting pipeline Jn (or Jbp). The differentiation induction unit of the device has a feed mechanism Fn (or Fbp) and feed mechanisms Jc1 to Jc(q-1) that move the contents of the container A3 (or the contents of the container Bp) to the containers C1 to Cq in order through the connecting pipeline Jn (or Jbp) switched to the communicating state and through Jc1 to Jc(q-1).
(容器の数q)
当該装置の分化誘導部において、容器の数(=分化誘導の段階の数)qは、特に限定はされないが、iPS細胞から目的の分化細胞を得るための分化誘導の段階の数を考慮すると、1~6程度(自家移植では、例えば、3~6程度)が有用な数である。
(Number of containers q)
In the differentiation induction section of the device, the number of containers (= the number of differentiation induction stages) q is not particularly limited, but taking into consideration the number of differentiation induction stages to obtain the desired differentiated cells from iPS cells, a useful number is approximately 1 to 6 (for example, approximately 3 to 6 in the case of autologous transplantation).
当該装置の分化誘導部において工程s5を実施する操作については、上記製造方法(II)で説明したとおりである。 The operation for carrying out step s5 in the differentiation induction section of the device is as described in the manufacturing method (II) above.
既に準備された(例えば、市販)iPS細胞を用いて分化誘導を行う場合、当該分化誘導部は、独立した細胞製造装置であってもよく、その前段には、iPS細胞製造部や拡大培養部が無くともよい。この場合、分化誘導の工程が複数であって、各工程に対応付けられた容器が複数であれば、〔複数の細胞製造工程が複数の容器に分けられ、工程の順に容器から容器へと接続管路を通じて細胞が移動するように構成され、それにより、各容器に対応付けられた工程が順次実施される〕という本発明の細胞製造装置に該当する。 When differentiation induction is performed using already prepared (e.g., commercially available) iPS cells, the differentiation induction section may be an independent cell manufacturing device, and there may be no iPS cell production section or expansion culture section in the preceding stage. In this case, if there are multiple differentiation induction steps and multiple containers associated with each step, this corresponds to the cell manufacturing device of the present invention in which [multiple cell manufacturing steps are divided into multiple containers, and cells are configured to move from container to container through connecting pipes in the order of the steps, whereby the steps associated with each container are performed sequentially].
(未分化細胞除去部)
当該装置における未分化細胞除去部は、上記製造方法(II)における工程6を実施する部分である。図10に示すように、当該装置は、未分化細胞除去部として、工程s6を実施するための容器D1をさらに有する。該容器D1では、前記分化誘導部(工程s5)の容器C1~Cqのうちの最後尾の容器Cq(図10では、X2)の内容物から、未分化細胞が除去される。該容器D1は、1以上の開閉可能な入出用ポートを有する。該容器D1は、接続管路(図10では、Jx2)を介して、前記最後尾の容器Cq(図10では、容器X2)に接続されているか、または接続可能な状態となっている。当該装置の未分化細胞除去部は、連通状態に切り替えられた接続管路Jx2を通じて、前記容器Cqの内容物を容器D1へと移動させる送り機構(図10では、Fx2)を有する、
(Undifferentiated cell removal section)
The undifferentiated cell removal unit in the device is a part that performs
未分化細胞除去部において工程s6を実施する操作については、上記製造方法(II)で説明したとおりである。 The procedure for carrying out step s6 in the undifferentiated cell removal section is as described in the manufacturing method (II) above.
(検査部)
当該装置における検査部は、上記製造方法(II)における工程s7を実施する部分である。図10に示すように、当該装置は、検査部として容器E1をさらに有する。該容器E1は、上記工程s6によって得られる分化細胞を検査するためのサンプルを取り出すための容器である。該容器E1は、1以上の開閉可能な入出用ポートを有する。該容器E1は、接続管路Jd1を介して、容器D1に接続されているか、または、接続可能な状態となっている。当該装置の検査部は、連通状態に切替えられた接続管路Jd1を通じて、容器D1の内容物を容器E1へと移動させる送り機構Fd1を有する。
(Inspection Department)
The inspection section of the device is a section that carries out step s7 in the manufacturing method (II). As shown in FIG. 10, the device further has a container E1 as an inspection section. The container E1 is a container for taking out a sample for inspecting the differentiated cells obtained by the above step s6. The container E1 has one or more openable/closable inlet/outlet ports. The container E1 is connected to the container D1 via a connecting pipe Jd1 or is in a state where it can be connected. The inspection section of the device has a feed mechanism Fd1 that moves the contents of the container D1 to the container E1 through the connecting pipe Jd1 that has been switched to a communicating state.
検査部において工程s7を実施する操作については、上記製造方法(II)で説明したとおりである。 The operation for carrying out step s7 in the inspection section is as described in the manufacturing method (II) above.
(容器を基板上に配置する態様)
図13は、当該装置の好ましい態様の一例を示す図である。図13の例では、当該装置は、当該装置に用いられる容器を配置するための基板をさらに有する。図13に示すように、該基板Y10上には工程の実施に必要な容器が配置され、各容器は該基板Y10に固定されている。図13の例では、計10個の容器(A1~A3、B1、B2、C1~C3、D1、E1)が工程の順に基板面に固定されている。同図の例では、容器A1の入出用ポート111には、体細胞を供給するための材料供給源であるシリンジG1が接続されており、容器E1の入出用ポートには、最終製品である分化細胞を取り出すためのシリンジが接続されている。各容器は、開閉可能な入出用ポート(例えば、符号111、112、113で示す部分)を有する。また、各容器は、連通状態と非連通状態に切り替え可能な上記接続管路(例えば、符号J1で示す部分)を介して、上記工程の順に接続されているか、または接続可能な状態となっている。細胞は図中の矢印の方向に順に移動し、各容器内でその容器に対応した処理を受ける。図13では、説明のために、接続管路を連通状態と非連通状態に切り替える機構や、接続可能な状態とする機構についての図示を省略している。また、連通状態に切り替えられた該接続管路を通じて、容器の内容物を次段の容器へと移動させる上記送り機構についても、図示を省略している。
(Aspect of placing the container on the substrate)
FIG. 13 is a diagram showing an example of a preferred embodiment of the device. In the example of FIG. 13, the device further has a substrate for arranging containers used in the device. As shown in FIG. 13, containers necessary for carrying out the steps are arranged on the substrate Y10, and each container is fixed to the substrate Y10. In the example of FIG. 13, a total of 10 containers (A1 to A3, B1, B2, C1 to C3, D1, E1) are fixed to the substrate surface in the order of the steps. In the example of the same figure, a syringe G1, which is a material supply source for supplying somatic cells, is connected to the input/
基板上に容器を配置した構成であれば、多数の容器を用いる場合であっても、取り扱いが容易になる。また、容器が工程の順番に基板上に配置されるので、複数のチューブ配管の混乱が抑制される。また、基板上に配置すると、製造工程の目視確認、および位置情報の取得にも適しており、作業者が作業工程を認識・識別する目印となり、さらには、位置情報の付与によって電子的データでの製造工程管理の情報処理が行いやすく成る利点を有するので、好ましい。 If the containers are arranged on a board, they are easy to handle, even when a large number of containers are used. In addition, because the containers are arranged on the board in the order of the process, confusion among the multiple tube piping is suppressed. Arranging them on a board is also preferable because it is suitable for visually checking the manufacturing process and obtaining position information, and serves as a mark for workers to recognize and identify the work process. Furthermore, adding position information makes it easier to process information for manufacturing process management using electronic data.
基板の材料は、特に限定されず、金属やプラスチックなどの硬質材料や、プラスチック製の可撓性材料などが例示され、塵、微粒子、揮発性ガスなどが発生せず、アルコールを用いた清拭清掃が可能な材料が好ましい。 The material of the substrate is not particularly limited, and examples include hard materials such as metals and plastics, and flexible plastic materials. It is preferable for the material to be one that does not generate dust, fine particles, or volatile gases, and that can be wiped clean with alcohol.
(2つ折り可能な基板)
図13に例示するように、当該装置の好ましい態様では、基板Y10が、折り曲げ中心線Y11を軸として2つ折り可能である。図13の例では、基板Y10の外周形状は、折り曲げ中心線Y11について線対称形である。この折り曲げ中心線Y11によって分けられた基板面の2つの領域e1、e2のうちの一方の領域e1には、上記容器のうちの所定数の容器(図13の例では、容器(A1、A2、A3、B1、B2))が、該折り曲げ中心線Y11に沿って、一つの向きd1に、順に並ぶように配置される。該折り曲げ中心線Y11によって分けられた基板面の2つの領域e1、e2のうちの他方の領域e2には、上記容器のうちの残りの容器(図13の例では、容器(C1、C2、C3、D1、E1))が、該折り曲げ中心線Y11に沿って、前記向きd1とは逆の向きd2に、順に並ぶように配置されている。そして、一方の領域e1の容器のうちの最後尾の容器(図13の例では、容器B2)と、他方の領域e2の容器のうちの先頭の容器(図13の例では、容器C1)とが、接続管路Jb2によって接続されているか、または接続可能な状態となっている。
(Foldable substrate)
As illustrated in FIG. 13, in a preferred embodiment of the device, the substrate Y10 can be folded in two with the folding center line Y11 as an axis. In the example of FIG. 13, the outer peripheral shape of the substrate Y10 is linearly symmetrical with respect to the folding center line Y11. In one area e1 of the two areas e1 and e2 of the substrate surface divided by the folding center line Y11, a predetermined number of the above-mentioned containers (in the example of FIG. 13, containers (A1, A2, A3, B1, B2)) are arranged so as to be lined up in order along the folding center line Y11 in one direction d1. In the other area e2 of the two areas e1 and e2 of the substrate surface divided by the folding center line Y11, the remaining containers (in the example of FIG. 13, containers (C1, C2, C3, D1, E1)) are arranged so as to be lined up in order along the folding center line Y11 in a direction d2 opposite to the direction d1. The last container among the containers in one area e1 (in the example of Figure 13, container B2) and the first container among the containers in the other area e2 (in the example of Figure 13, container C1) are connected or can be connected by a connecting pipe Jb2.
前記のように2つ折り可能な基板上に容器を配置すると、図14に例示するように、基板を2つ折りすると、各容器の入出用ポートが同じ方向(図14では上方)を向いて互いに接近する。これにより、容器に接続される各チューブの長さは最短となり、かつ、どのチューブの長さも同じ(または同様)となる。結果として、本発明に用いられる全ての容器は、いずれも同様のチューブ付きの同様の規格の製品を一種類用意するだけでよくなるので、低コスト化が実現される。また、チューブ長さが同じであれば、材料供給源から容器までの材料到達時間の差を小さくすることができる。これにより、製造ロット間での、細胞の品質の差を小さくすることができるので好ましい。さらに、チューブ長さが最短となることで、チューブ内の無駄なスペースが減り、製造ロスも減少する。 When containers are placed on a foldable substrate as described above, as shown in FIG. 14, when the substrate is folded in half, the inlet/outlet ports of each container face in the same direction (upward in FIG. 14) and approach each other. This minimizes the length of each tube connected to the container, and all tubes are the same (or similar) in length. As a result, all containers used in the present invention only require the preparation of one type of product with similar specifications and similar tubes, thereby realizing low costs. In addition, if the tube lengths are the same, the difference in the time it takes for the material to reach the container from the material supply source can be reduced. This is preferable because it reduces the difference in cell quality between production lots. Furthermore, by minimizing the tube length, wasted space in the tube is reduced, and production losses are also reduced.
(基板の2つ折り)
図14は、上記基板を2つ折りする場合の態様を例示する図である。2つ折りは、断面がV字状のシャープな折り目が形成されるような態様であってもよい。しかし、図14に示すように、断面がU字状であるような湾曲した折り目であれば、2つの領域e1、e2に位置する容器同士が互いに過度に接近しないので好ましい。
(Folding the board in half)
Fig. 14 is a diagram illustrating an example of a case where the substrate is folded in two. The folding may be performed in such a manner that a sharp crease having a V-shaped cross section is formed. However, as shown in Fig. 14, a curved crease having a U-shaped cross section is preferable because the containers located in the two regions e1 and e2 are not excessively close to each other.
基板を2つ折りにしたとき、図14(a)に示すように、容器の入出用ポートが折り目から遠ざかる向きに延びていてもよいし、図14(b)に示すように、容器の入出用ポートが折り目に近づく向きに延びていてもよい。また、基板を2つ折りにしたとき、容器は2つ折りされた基板に挟まれる位置にあってもよい。基板が容器を覆わず、容器内の観察や容器への外部チューブのアクセスが容易である点からは、図14に示すように、容器は2つ折りされた基板の外側にある態様が好ましい。 When the substrate is folded in half, the inlet/outlet port of the container may extend away from the fold as shown in FIG. 14(a), or may extend toward the fold as shown in FIG. 14(b). Furthermore, when the substrate is folded in half, the container may be located in a position sandwiched between the folded substrates. In order to facilitate observation of the inside of the container and access to the container with an external tube without the substrate covering the container, it is preferable that the container be located on the outside of the folded substrate as shown in FIG. 14.
(複数の容器が、2枚の可撓性シート間に形成される態様)
図15は、当該装置の好ましい態様の他の例を示す図である。図15の例では、当該装置は、2枚の互いに重ね合わせられた可撓性シートY31、T32をさらに有する。この態様では、当該装置の全ての容器となる領域と、1以上の入出用ポートとなる領域と、接続管路となる領域とが、前記2枚の可撓性シートY31、T32の間の所定の位置に形成されている。即ち、これらの領域を非接合領域として残して、前記2枚の可撓性シートは互いに接合されている。図15では、説明のために、容器A1と接続管路J1だけを示している。図15(a)に示すように、2枚の可撓性シートY31、T32が、それらの間に容器A1と接続管路J1を形成しており、これらの領域の外周囲において、2枚の可撓性シートY31、T32が互いに接合されて1枚のシートY30となっている。可撓性シートY31、T32が互いに接合される領域は、容器や管路などの形成すべき領域の外形線に隣接する帯状の領域だけであってもよいし、該形成すべき領域以外の全域であってもよい。
(An embodiment in which multiple containers are formed between two flexible sheets)
FIG. 15 is a diagram showing another example of a preferred embodiment of the device. In the example of FIG. 15, the device further has two flexible sheets Y31 and T32 stacked on each other. In this embodiment, the area that will become all the containers of the device, the area that will become one or more input/output ports, and the area that will become the connecting pipeline are formed at predetermined positions between the two flexible sheets Y31 and T32. That is, the two flexible sheets are joined to each other, leaving these areas as non-jointed areas. In FIG. 15, only the container A1 and the connecting pipeline J1 are shown for the purpose of explanation. As shown in FIG. 15(a), the two flexible sheets Y31 and T32 form the container A1 and the connecting pipeline J1 between them, and the two flexible sheets Y31 and T32 are joined to each other around the outer periphery of these areas to form one sheet Y30. The area where the flexible sheets Y31 and T32 are joined to each other may be only a strip-shaped area adjacent to the outline of the area where a container, pipeline, etc. is to be formed, or it may be the entire area other than the area where the container, pipeline, etc. is to be formed.
図15(b)は、図15(a)の図を、切断面w1-w1において切断したときの端面図である。図15(b)には、説明のために、背後に見える容器A1の外形を破線で示している。該2枚の可撓性シートY31、Y32の外面には、該接続管路を開閉するためのアクチュエーターJ1aが設けられている。該押圧用アクチュエーターJ1aは、例えば、ピンチバルブなどの直動型のプレス装置であって、接続管路の側方の領域においてシートY30を貫通するように設けられており、それにより、接続管路を表裏から挟み込んで押圧することができるようになっている。該押圧用アクチュエーターJ1aは、押圧ポジション(接続管路となる領域を該可撓性シートY31、T32の外側から押圧して非連通状態とするポジション)と、非押圧ポジション(該接続管路となる領域を押圧せず連通状態とするポジション)とを取るように作動する。図15の例では、該押圧用アクチュエーターJ1aは、非押圧ポジションを取っており、接続管路J1は連通状態にある。該押圧用アクチュエーターJ1aの作動により、接続管路となる領域は、連通状態と非連通状態とに切り替え可能な接続管路として機能する。各容器において1以上の入出用ポートとなる領域は、図13に示すように、各容器(A1、A2、A3など)となる領域から可撓性シート(図13では基板Y10)の外周縁まで延びて開口端部となっている。該開口端部には、開閉可能な入出用ポートのための構成(図示せず)が付与されている。 FIG. 15(b) is an end view of FIG. 15(a) cut at the cut surface w1-w1. For the purpose of explanation, FIG. 15(b) shows the outline of the container A1 seen in the background in broken lines. An actuator J1a for opening and closing the connection pipeline is provided on the outer surfaces of the two flexible sheets Y31 and Y32. The pressing actuator J1a is, for example, a direct acting press device such as a pinch valve, and is provided so as to penetrate the sheet Y30 in the area to the side of the connection pipeline, thereby making it possible to pinch and press the connection pipeline from the front and back. The pressing actuator J1a operates to take a pressing position (a position in which the area to become the connection pipeline is pressed from the outside of the flexible sheets Y31 and T32 to make it non-connected) and a non-pressing position (a position in which the area to become the connection pipeline is not pressed and made connected). In the example of FIG. 15, the pressing actuator J1a is in a non-pressing position, and the connection pipe J1 is in a connected state. By actuating the pressing actuator J1a, the region that becomes the connection pipe functions as a connection pipe that can be switched between a connected state and a non-connected state. As shown in FIG. 13, the region that becomes one or more input/output ports in each container extends from the region that becomes each container (A1, A2, A3, etc.) to the outer edge of the flexible sheet (substrate Y10 in FIG. 13) to form an open end. The open end is provided with a structure (not shown) for an openable input/output port.
前記のように、2枚の可撓性シートの間に、工程の数に対応した複数の容器と接続管路と入出用ポートを設けると、簡単な構成によって、多数の容器を持った当該装置が得られる。このような構成は、コストの点から、1回の使用ごとに使い捨てが可能である。 As described above, by providing multiple containers corresponding to the number of processes, connecting pipelines, and inlet/outlet ports between two flexible sheets, the device can be constructed with a large number of containers in a simple manner. From the standpoint of cost, this type of construction can be disposable after one use.
(2つ折り可能なシート)
当該装置の好ましい態様では、図15の互いに重ね合わせられ互いに接合された2枚の可撓性シートY31、Y32の外周形状は、図13に示すように、折り曲げ中心線Y11を軸として2つ折り可能な形状である。より好ましい態様では、該シートの外周形状は、折り曲げ中心線Y11について線対称形である。
この折り曲げ中心線Y11によって分けられた2つの領域e3、e4のうちの一方の領域e3には、上記容器のうちの所定数の密閉(図13の例では、容器(A1、A2、A3、B1、B2))となる領域が、該折り曲げ中心線Y11に沿って、一つの向きd3に、順に並ぶように形成される。該所定数の容器となる領域の各外周のうち、該折り曲げ中心線Y11から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域(111、112、113)が、該折り曲げ中心線Y11から離れる方向に延びている。該入出用ポートとなる領域(111、112、113)は、接合された可撓性シートY31、Y32の外周縁まで延びて開口端部となっている。容器となる領域同士の間には、該容器となる領域同士を接続する接続管路となる領域が形成されている。
(Sheet that can be folded in two)
In a preferred embodiment of the device, the outer peripheral shape of the two overlapping and joined flexible sheets Y31 and Y32 in Fig. 15 is a shape that can be folded in two around the folding center line Y11 as an axis, as shown in Fig. 13. In a more preferred embodiment, the outer peripheral shape of the sheets is symmetrical with respect to the folding center line Y11.
In one of the two regions e3 and e4 separated by the folding center line Y11, a predetermined number of regions that will become sealed containers (in the example of FIG. 13, containers (A1, A2, A3, B1, B2)) are formed in order along the folding center line Y11 in one direction d3. From the outer periphery of each of the regions that will become the predetermined number of containers, the one or more inlet/outlet port regions (111, 112, 113) extend in a direction away from the folding center line Y11. The inlet/outlet port regions (111, 112, 113) extend to the outer periphery of the joined flexible sheets Y31 and Y32 to form open ends. Between the regions that will become containers, a region that will become a connecting pipe that connects the regions that will become containers is formed.
該折り曲げ中心線Y11によって分けられた2つの領域e3、e4のうちの他方の領域e4には、上記容器のうちの残りの容器(図13の例では、容器(C1、C2、C3、D1、E1))となる領域が、該折り曲げ中心線Y11に沿って、前記一つの向きd3とは逆の向きd4に、順に並ぶように形成されている。また、残りの容器となる領域の各外周のうち、折り曲げ中心線Y11から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域が、該折り曲げ中心線Y11から離れる方向に延び、かつ、前記2枚の可撓性シートの外周縁まで延びて開口端部となっている。前記残りの容器となる領域同士の間には、該容器となる領域同士を接続する接続管路となる領域(図13の例では符号J1の部分など)が形成されている。前記一方の領域e3の容器のうちの最後尾の密閉容器(図13の例では、容器B2)と、前記他方の領域e4の容器のうちの先頭の容器(図13の例では、容器C1)とは、前記折り曲げ中心線を横切る接続管路Jb2となる領域によって接続されている。 In the other region e4 of the two regions e3, e4 separated by the folding center line Y11, regions which will become the remaining containers (in the example of Figure 13, containers (C1, C2, C3, D1, E1)) are formed in order along the folding center line Y11 in a direction d4 opposite to the one direction d3. Furthermore, from the outer periphery of each of the regions which will become the remaining containers, which is located farther from the folding center line Y11, regions which will become the one or more input/output ports extend in a direction away from the folding center line Y11 and extend to the outer periphery of the two flexible sheets to form an open end. Between the regions which will become the remaining containers, regions which will become connecting pipelines connecting the regions which will become the containers (such as the part indicated by symbol J1 in the example of Figure 13) are formed. The rearmost sealed container among the containers in the one region e3 (in the example of FIG. 13, container B2) and the first container among the containers in the other region e4 (in the example of FIG. 13, container C1) are connected by a region that becomes the connecting pipe Jb2 that crosses the folding center line.
このような2つ折り可能な態様によれば、上記の2つ折り可能な基板の説明においても述べたとおり、各容器の入出用ポートが同じ方向を向いて互いに接近するので、容器に接続される各チューブの長さが最短、かつ、どのチューブの長さも同じ(または同様)となるので好ましい。 As described above in the description of the foldable substrate, this type of foldable configuration is preferable because the inlet/outlet ports of each container face in the same direction and are close to each other, making the length of each tube connected to the container the shortest and all of the tubes the same (or similar) length.
図16は、基板上に複数の容器を配列して固定するための好ましい態様の一例を示す図であり、図17は、図16に示す基板の実施例品およびその使用状態を示す図である。図16、図17に示す態様では、屈曲可能な柔軟なシートで形成された基板Y10の主面上の、容器を配置すべき位置に、容器を収容し得るポケットY20が設けられている。同図の例では、基板Y10は、その主面が垂直面となるように用いられ、折り曲げ中心線Y11の両側の各領域e1、e2には、それぞれ垂直方向に5つのポケットが配置されている。各ポケットは、容器を適切に収容できる大きさに構成されている。領域e1の5つのポケットには、上から下へ順に容器A1、A2、A3、B1、B2が挿入され、領域e2の5つのポケットには、下から上へ順に容器(C1、C2、C3、D1、E1)が挿入されている。この配置によって、10個の容器が2つの列(容器A1、A2、A3、B1、B2と、容器C1、C2、C3、D1、E1)をなしてコンパクトになり、容器B2と容器C1とが接近しており、両者の連結も、他の容器同士の連結と同様に容易である。 16 is a diagram showing an example of a preferred embodiment for arranging and fixing a plurality of containers on a substrate, and FIG. 17 is a diagram showing an embodiment of the substrate shown in FIG. 16 and its use state. In the embodiment shown in FIG. 16 and FIG. 17, a pocket Y20 capable of accommodating a container is provided at a position where the container is to be arranged on the main surface of a substrate Y10 formed from a bendable flexible sheet. In the example shown in the figure, the substrate Y10 is used so that its main surface is a vertical surface, and five pockets are arranged vertically in each of the regions e1 and e2 on both sides of the folding center line Y11. Each pocket is configured to a size that can appropriately accommodate a container. Containers A1, A2, A3, B1, and B2 are inserted into the five pockets of the region e1 in order from top to bottom, and containers (C1, C2, C3, D1, and E1) are inserted into the five pockets of the region e2 in order from bottom to top. This arrangement allows the ten containers to be compactly arranged in two rows (containers A1, A2, A3, B1, B2, and containers C1, C2, C3, D1, E1), with containers B2 and C1 close to each other and easy to connect to each other, just like connecting other containers to each other.
基板面に容器を挿入するためのポケットを設けたことにより、基板に対して容器を速やかに着脱できるので、好ましい。また、図17に示すように、容器を垂直方向に配置したことによって、広い場所を占有しないコンパクトな装置を得ることができる。 Providing a pocket on the substrate surface for inserting the container is preferable because it allows the container to be quickly attached to and detached from the substrate. Also, as shown in Figure 17, by arranging the container vertically, a compact device that does not occupy a large space can be obtained.
図18(a)は、基板上に複数の容器を配列して固定するための好ましい他の態様例を示す図である。図18(b)は、図18(a)に示す基板の実施例品およびその使用状態を示す図である。図18に示す態様では、図16、図17に示す態様と同様に、柔軟なシートで形成された基板Y10に、容器を収容し得るポケットY20が設けられている。ただし、同図の例では、折り曲げ中心線Y11の両側の各領域e1、e2に設けられた5つのポケットの開口部がいずれも折り曲げ中心線Y11の方向を向いている。両頭の矢印は、各ポケットに容器を入出する方向を示している。領域e1の5つのポケットには、図の右から左へ順に容器A1、A2、A3、B1、B2が挿入され、領域e2の5つのポケットには、図の左から上から順に容器(C1、C2、C3、D1、E1)が挿入されている。折り曲げ中心線Y11において、基板を図14(b)に示すように2つに折り曲げることによって、図18(b)に示すように、各容器の入出用ポートが同じ方向(図14では上方)を向いて互いに接近するので好ましい。 18(a) is a diagram showing another preferred embodiment for arranging and fixing a plurality of containers on a substrate. FIG. 18(b) is a diagram showing an embodiment of the substrate shown in FIG. 18(a) and its use state. In the embodiment shown in FIG. 18, similar to the embodiments shown in FIG. 16 and FIG. 17, a pocket Y20 capable of accommodating a container is provided on a substrate Y10 formed of a flexible sheet. However, in the example shown in the figure, the openings of the five pockets provided in each of the regions e1 and e2 on both sides of the folding center line Y11 all face the direction of the folding center line Y11. The double-headed arrows indicate the direction in which a container is inserted and removed from each pocket. Containers A1, A2, A3, B1, and B2 are inserted into the five pockets of the region e1 in order from right to left in the figure, and containers (C1, C2, C3, D1, and E1) are inserted into the five pockets of the region e2 in order from left to top in the figure. By folding the substrate in two at the folding center line Y11 as shown in FIG. 14(b), the inlet/outlet ports of each container face in the same direction (upward in FIG. 14) and approach each other, as shown in FIG. 18(b), which is preferable.
当該装置における容器の入出用ポートの開閉、外部の材料供給源による材料供給の開始と停止、接続管路の開閉、送り機構の作動と停止、温度管理、時間管理などは、手動で行ってもよいし、制御装置(制御プログラムを実行するコンピューターや、シーケンス回路など)によって自動で行なってもよいし、これらを組み合わせた半自動で行ってもよい。 The opening and closing of the container's inlet and outlet ports, starting and stopping the supply of material from an external material supply source, opening and closing of the connecting pipelines, starting and stopping the feed mechanism, temperature control, time control, etc. in the device can be performed manually, automatically by a control device (a computer that executes a control program, a sequence circuit, etc.), or semi-automatically by combining these.
(上記細胞製造装置を用いた細胞製造方法)
当該細胞製造方法は、本発明の細胞製造装置を用いてiPS細胞を製造する方法であって、上記製造方法(I)で説明したとおり、工程s1~s3を各容器A1~A3で実施し、容器A3においてiPS細胞を得る方法である。
(Cell manufacturing method using the above cell manufacturing device)
This cell manufacturing method is a method for producing iPS cells using the cell manufacturing apparatus of the present invention, and as described in the above manufacturing method (I), steps s1 to s3 are carried out in each of containers A1 to A3, and iPS cells are obtained in container A3.
当該製造方法は、図11に示すように、次の工程s1~s3を少なくとも有する。
(i)上記細胞製造装置の容器A1内において、液体培地中で体細胞に初期化因子を接触させる工程s1。
(ii)前記工程s1の完了後に、連通状態に切り替えられた接続管路J1を通じて、該容器A1の内容物を容器A2内に移動させ、該容器A2内において、前記液体培地中の初期化因子の濃度を低減させる工程s2。
(iii)前記工程s2の完了後に、連通状態に切り替えられた接続管路J2を通じて、該容器A2の内容物を容器A3内に移動させ、該容器A3内において、液体培地中でiPS細胞を樹立する工程s3。
As shown in FIG. 11, the manufacturing method includes at least the following steps s1 to s3.
(i) Step s1 of contacting somatic cells with reprogramming factors in a liquid medium in a container A1 of the cell manufacturing apparatus.
(ii) after completion of step s1, a step s2 is performed in which the contents of container A1 are transferred into container A2 through connecting pipe J1 that has been switched to a communicating state, and the concentration of the reprogramming factor in the liquid medium is reduced in container A2.
(iii) after completion of step s2, a step s3 of transferring the contents of container A2 into container A3 through connecting pipeline J2 switched to a communicating state, and establishing iPS cells in liquid medium in container A3.
工程s1~s3および上記細胞製造装置の詳細については、上記説明のとおりである。当該製造方法は、iPS細胞から分化細胞を製造する工程、分化細胞が含まれた懸濁液から未分化細胞を除去する工程、さらには、得られた分化細胞を検査する工程を有してもよい。 Details of steps s1 to s3 and the cell manufacturing apparatus are as described above. The manufacturing method may include a step of manufacturing differentiated cells from iPS cells, a step of removing undifferentiated cells from the suspension containing the differentiated cells, and a step of inspecting the obtained differentiated cells.
以下に、本発明の細胞製造装置を実際に製作し、該装置を用いて本発明によるiPS細胞の製造方法を実施した様子、および、得られたiPS細胞の評価を示す。製作した細胞製造装置の各部の動作(材料供給動作、入出用ポートおよび接続管路の開閉動作、送り機構のポンプ動作など)は、実験用のため全て手動による動作であるが、いずれもコンピューター制御可能な開閉機構やポンプ装置などを用いることによって、自動化が可能な動作である。 Below, we show how the cell manufacturing device of the present invention was actually manufactured and how the method for manufacturing iPS cells of the present invention was carried out using the device, as well as an evaluation of the iPS cells obtained. The operations of each part of the manufactured cell manufacturing device (material supply operations, opening and closing operations of input/output ports and connecting pipes, pump operation of the feed mechanism, etc.) are all manual operations for experimental purposes, but all of these operations can be automated by using computer-controlled opening and closing mechanisms and pump devices, etc.
実施例1
(ヒト全血からのiPS細胞の製造)
本実施例では、ヒト全血を遠心分離して末梢血単核球(PBMC)を得、該PBMCからiPS細胞を製造した。本発明による細胞製造装置として、図2に示した自作の密閉容器を用い、該容器を図19に示すように、ボトルラック(試験管立てに類するもの)に直列的に配置し、直列的に接続可能な状態として、当該細胞製造装置を構成した。各容器の容積は、いずれも20mlである。各容器は、いずれも、ガス用の第1入出用ポート、汎用の第2入出用ポート、汎用の第3入出用ポートを有し、汎用の入出用ポートは、材料供給用の管路として、また、接続管路として用いられるものである。
Example 1
(Production of iPS cells from human whole blood)
In this example, human whole blood was centrifuged to obtain peripheral blood mononuclear cells (PBMCs), and iPS cells were produced from the PBMCs. As the cell production device according to the present invention, a self-made sealed container shown in FIG. 2 was used, and the containers were arranged in series in a bottle rack (similar to a test tube stand) as shown in FIG. 19, so that the cell production device was configured so that the containers could be connected in series. The volume of each container was 20 ml. Each container had a first inlet/outlet port for gas, a general-purpose second inlet/outlet port, and a general-purpose third inlet/outlet port, and the general-purpose inlet/outlet port was used as a material supply line and as a connection line.
iPS細胞の製造工程は、次のとおりである。
工程s1:容器A1内において、液体培地中でPBMCに初期化因子を接触させる工程。
工程s2:容器A2内において、液体培地中の初期化因子の濃度を低減させる工程。
工程s3:容器A3において、14日間の培養を行い、iPS細胞を樹立する工程。
工程s4-1:容器B1において、1回目の拡大培養を行う工程。
工程s4-2:容器B2において、2回目の拡大培養を行う工程。(工程s4-2は、工程s4-1と操作は共通。)
The process for producing iPS cells is as follows.
Step s1: A step of contacting PBMCs with reprogramming factors in a liquid medium in container A1.
Step s2: A step of reducing the concentration of the reprogramming factor in the liquid medium in container A2.
Step s3: A step of culturing in container A3 for 14 days to establish iPS cells.
Step s4-1: A step of performing a first expansion culture in container B1.
Step s4-2: A step of performing a second expansion culture in the container B2. (Step s4-2 is the same as step s4-1 in terms of the operation.)
(ヒト全血液からPBMCを取得する処理(遠心分離))
テルモ血液バッグMAP液(TERUMO BB-QM200J8A)に入った40mlのヒト全血から、4mlのヒト全血を採取し、単核球分離用採血管(BDバキュテイナ(登録商標)CPT(BD 362760)へ4ml入れる操作を行なった。
(Process for obtaining PBMC from human whole blood (centrifugation))
From 40 ml of human whole blood contained in a Terumo blood bag MAP liquid (TERUMO BB-QM200J8A), 4 ml of human whole blood was collected and placed in a blood collection tube for mononuclear cell separation (BD Vacutainer (registered trademark) CPT (BD 362760)).
安全キャビネット内で、採血用バッグの排出口に、薬液注入吸引用のコネクターであるケモクレーブ(登録商標)バッグスパイクを接続した。該バッグスパイクにシリンジ5mlを接続し、血液4mlを該シリンジへ吸引した。該バッグスパイクからシリンジを外し、該バッグスパイクはキャップ(Combi-Stopper)で閉鎖した。
ヒト全血4mlを収容したシリンジへ注射針(23G)を安全キャビネット内で装着した。該注射針を、BDバキュテイナCPTのゴム栓に穿刺し、該ヒト全血を陰圧で引き注入した。
BDバキュテイナCPTを遠心分離機内にセットし、遠心分離により(1500~1800G、15分(ヘパリン)、20分(クエン酸))、PBMCを分離した。
In a safety cabinet, a Chemoclave (registered trademark) bag spike, which is a connector for injecting and suctioning a drug solution, was connected to the outlet of the blood collection bag. A 5 ml syringe was connected to the bag spike, and 4 ml of blood was sucked into the syringe. The syringe was removed from the bag spike, and the bag spike was closed with a cap (Combi-Stopper).
An injection needle (23G) was attached to a syringe containing 4 ml of human whole blood in a safety cabinet. The injection needle was pierced into the rubber stopper of a BD Vacutainer CPT, and the human whole blood was drawn and injected by negative pressure.
The BD Vacutainer CPT was placed in a centrifuge and PBMCs were separated by centrifugation (1500-1800 G, 15 minutes (heparin), 20 minutes (citric acid)).
(工程s1:容器A1において、PBMCに初期化因子を接触させる工程)
(i)容器A1へのPBMCの注入
ロックコネクター(テルモ社製TS-LC11)を介在させて、容器A1の第2入出用ポートと、容器A2の第3入出用ポートを安全キャビネット内で接続した。実際の製造においては、事前に接続済みであってもよい。
容器A1と容器A2のそれぞれのガス用の入出用ポートを閉鎖した。
図20に示すように、吸引ポンプ用のシリンジを容器A2の第2入出用ポートへ接続し、BDバキュテイナ(登録商標)ルアーロックアクセスデバイス(以下、ルアーロックアクセスデバイス)を容器A1の第3入出用ポートへ接続し、該ルアーロックアクセスデバイスに、遠心分離されたPBMCを含む液体を収容する上記のBDバキュテイナCPTを接続した。
容器A2の第2入出用ポートに接続した吸引ポンプ用のシリンジによって、容器A2内の空気を吸引した。吸引ポンプ用のシリンジの操作によって、BDバキュテイナCPT内のPBMCをシリンジ内に流入させ、その後、該シリンジ内のPBMCを容器A1内に流入させた。
(Step s1: contacting PBMC with reprogramming factors in container A1)
(i) Injection of PBMCs into container A1 The second inlet/outlet port of container A1 was connected to the third inlet/outlet port of container A2 in a safety cabinet via a lock connector (TS-LC11 manufactured by Terumo Corporation). In actual production, the connection may be made in advance.
The gas inlet and outlet ports of vessel A1 and vessel A2 were closed.
As shown in Figure 20, a syringe for a suction pump was connected to the second input/output port of container A2, a BD Vacutainer (registered trademark) Luer lock access device (hereinafter referred to as the Luer lock access device) was connected to the third input/output port of container A1, and the above-mentioned BD Vacutainer CPT containing a liquid containing centrifuged PBMCs was connected to the Luer lock access device.
Air was sucked out from inside the container A2 using a syringe for a suction pump connected to the second inlet/outlet port of the container A2. By operating the syringe for the suction pump, the PBMCs in the BD Vacutainer CPT were made to flow into the syringe, and then the PBMCs in the syringe were made to flow into the container A1.
(ii)容器A1への初期化因子の注入
市販のベクターはバイアルに充填されているので、該バイアルからシリンジを用いてベクターを採取した。臨床用の細胞の製造においては、該ベクターは、無菌的に接続可能なコネクターを備えた容器に封入されて提供される。シリンジ(テルモ社製)2.5mlと、注射針(テルモ社製、23G×1)を用いて、初期化因子としてSRV iPS-2 Vector (4.6×107 CIU/ml 0.1ml)を0.1ml(全量)吸い上げた。
容器A1の第3入出用ポートに接続されていた上記のルアーロックアクセスデバイスを外し、その第3入出用ポートに、針を外した前記シリンジを接続し、SRV iPS-2 Vector全量を容器A1の第3入出用ポートを通じて容器A1内に注入した。これにより、PBMCと初期化因子を接触させた。
(ii) Injection of reprogramming factor into container A1 Commercially available vectors are filled in vials, so the vector was extracted from the vial using a syringe. In the production of cells for clinical use, the vector is provided sealed in a container equipped with a connector that can be connected aseptically. Using a 2.5 ml syringe (Terumo Corporation) and an injection needle (Terumo Corporation, 23G x 1), 0.1 ml (total amount) of SRV iPS-2 Vector (4.6 x 10 7 CIU/ml 0.1 ml) was drawn up as a reprogramming factor.
The above-mentioned luer lock access device connected to the third inlet/outlet port of container A1 was removed, the syringe without the needle was connected to the third inlet/outlet port, and the entire amount of SRV iPS-2 Vector was injected into container A1 through the third inlet/outlet port of container A1. This allowed the PBMCs to come into contact with the reprogramming factors.
容器A1を含んだ装置全体を、37℃のホットプレートを設置した安全キャビネット内で、2時間インキュベートした。 The entire apparatus, including container A1, was incubated for 2 hours in a safety cabinet equipped with a hot plate at 37°C.
(工程s2:液体培地中の初期化因子の濃度を低減させる工程)
ロックコネクター(テルモ社製、TS-LC11)を用いて、容器A1の第2入出用ポートと、容器A2の第3入出用ポートとを、安全キャビネット内において、接続管路で接続した(実際の製造においては事前に接続済みである)。
次に容器A1のガス用の入出用ポートを閉鎖し、該容器A1の第3入出用ポートに接続されていたシリンジを外し、図21に示すように、StemFit AK03培地が20ml以上入ったシリンジを容器A1の第3入出用ポートに接続して、該StemFit AK03培地を20ml注入した。その結果、容器A1と容器A2には、それぞれ元のバイアルに対して半分の量の細胞(PBMC)と、元のバイアルに対して200倍に希釈された初期化ベクターが収容された状態となった。
(Step s2: Reducing the concentration of reprogramming factors in the liquid medium)
Using a lock connector (Terumo Corporation, TS-LC11), the second input/output port of container A1 and the third input/output port of container A2 were connected with a connecting pipe inside a safety cabinet (in actual production, the connection was already made in advance).
Next, the gas inlet/outlet port of container A1 was closed, the syringe connected to the third inlet/outlet port of container A1 was removed, and a syringe containing 20 ml or more of StemFit AK03 medium was connected to the third inlet/outlet port of container A1, and 20 ml of the StemFit AK03 medium was injected, as shown in Figure 21. As a result, container A1 and container A2 each contained half the amount of cells (PBMCs) compared to the original vial, and reprogramming vectors diluted 200 times compared to the original vial.
(工程s3:14日間の培養によるiPS細胞の樹立)
図22に示すように、容器A3の第3入出用ポートより、シリンジにて、5mLのアテロコラーゲンビーズ溶液(KOKEN:MIC-00)を安全キャビネット内で注入した。同シリンジを外し、ロックコネクターを用いて容器A2の第2入出用ポートと容器A3の第3入出用ポートを安全キャビネット内で接続した(実際の製造では、事前に接続済みである)。
容器A2の第1入出用ポート(ガス用)を閉鎖し、容器A2の第3入出用ポートに接続されているロックコネクターを安全キャビネット内で外した。
使用済みとなった容器A1については、第1入出用ポートの閉鎖を確認し、容器A1の第2入出用ポートは、接続済みのロックコネクターの開口部を、流体ディスペンシングコネクターを利用して閉鎖した。また、容器A1の第3入出用ポートも流体ディスペンシングコネクターのキャップを用いて閉鎖した。
(Step s3: Establishment of iPS cells by culturing for 14 days)
22, 5 mL of atelocollagen bead solution (KOKEN: MIC-00) was injected from the third inlet/outlet port of container A3 with a syringe in a safety cabinet. The syringe was removed, and the second inlet/outlet port of container A2 and the third inlet/outlet port of container A3 were connected in the safety cabinet using a lock connector (in actual production, they were connected in advance).
The first inlet/outlet port (for gas) of vessel A2 was closed, and the lock connector connected to the third inlet/outlet port of vessel A2 was removed in a safety cabinet.
For the used container A1, the first inlet/outlet port was confirmed to be closed, and the opening of the connected lock connector for the second inlet/outlet port of the container A1 was closed using a fluid dispensing connector. The third inlet/outlet port of the container A1 was also closed using the cap of the fluid dispensing connector.
図23に示すように、液体培地(StemFit AK03)が10mL以上入ったシリンジを容器A2の第3入出用ポートへ接続して、該液体培地を10mL注入する。その結果、容器A3の液体培地の量は10mLとなった。この段階で、容器A3には、元のバイアルに対して1/4の量のPBMCと、元のバイアルに対して400倍に希釈された初期化ベクターが収容された状態となった。 As shown in Figure 23, a syringe containing 10 mL or more of liquid medium (StemFit AK03) is connected to the third inlet/outlet port of container A2, and 10 mL of the liquid medium is injected. As a result, the amount of liquid medium in container A3 becomes 10 mL. At this stage, container A3 contains 1/4 the amount of PBMC compared to the original vial, and initialization vector diluted 400 times compared to the original vial.
容器A3の第3入出用ポートに接続されているロックコネクターを安全キャビネット内で外した。また、容器A3の第3入出用ポートも流体ディスペンシングコネクター(BRAUN:415080)のキャップを用いて閉鎖した。使用済みとなった容器A2については、第1入出用ポート(ガス用)の閉鎖を確認し、容器A2の第2入出用ポートは接続済みのロックコネクターの開口を流体ディスペンシングコネクターのコネクターを用いて閉鎖した。また、容器A2の第3入出用ポートも流体ディスペンシングコネクターのキャップを用いて閉鎖した。 The lock connector connected to the third inlet/outlet port of container A3 was removed inside the safety cabinet. The third inlet/outlet port of container A3 was also closed using the cap of a fluid dispensing connector (BRAUN: 415080). For container A2, which had been used, it was confirmed that the first inlet/outlet port (for gas) was closed, and the opening of the connected lock connector for the second inlet/outlet port of container A2 was closed using the connector of the fluid dispensing connector. The third inlet/outlet port of container A2 was also closed using the cap of the fluid dispensing connector.
当該装置全体を、37℃のホットプレートを設置した安全キャビネット内で14日間インキュベートした。これにより、iPS細胞が樹立した。 The entire device was incubated in a safety cabinet equipped with a hot plate at 37°C for 14 days. This resulted in the establishment of iPS cells.
なお、液体培地を継続的に供給する場合には、容器A3の第3入出用ポートを閉鎖せずに、エクステンションチューブ(シーマンCTL1001S2)を介して液体培地(StemFit AK03)が入ったシリンジを容器A3の第3入出用ポートに接続して、該液体培地を継続的に供給すればよい。また、廃液の排出のために、容器A3の第3入出用ポートを閉鎖せずに、エクステンションチューブを介して、シリンジを接続してもよいし、廃液バッグを接続してもよい。該廃液バッグは、吸湿性の素材(例えば、吸水性樹脂等、より具体的には、ポリアクリル酸、ポリアクリル酸ナトリウム、ポリアクリル酸共重合体等)や、該素材を含む吸収性物品(例えば、吸水パッド、吸水シート等)を封入した吸湿性の廃液バッグであってもよい。本実施例では、液体培地の継続的な供給を行なった(0.1ml/h)。自動送液ポンプとして、クーデックシリンジポンプ(大研医器株式会社製、CSP-120)を用い、該シリンジポンプの機器を固定するために、ポンプユナイタースタンド(大研医器株式会社製、PUS-200S)と、ポンプユナイター(大研医器株式会社製、PU3-200S)を用いた。 When liquid medium is to be continuously supplied, the third inlet/outlet port of container A3 is not closed, and a syringe containing liquid medium (StemFit AK03) is connected to the third inlet/outlet port of container A3 via an extension tube (Seaman CTL1001S2) to continuously supply the liquid medium. In addition, to discharge waste liquid, a syringe or a waste liquid bag may be connected via an extension tube without closing the third inlet/outlet port of container A3. The waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.). In this example, liquid medium was continuously supplied (0.1 ml/h). A Coudec Syringe Pump (Daiken Medical Co., Ltd., CSP-120) was used as the automatic liquid delivery pump, and a Pump Uniter Stand (Daiken Medical Co., Ltd., PUS-200S) and a Pump Uniter (Daiken Medical Co., Ltd., PU3-200S) were used to secure the syringe pump.
(工程s4:iPS細胞の拡大培養)
前記工程s3における14日間のインキュベートを停止すべく、シリンジポンプを停止した。シリンジ(テルモ社製、50mL)を外し、容器A3のガス用ポートである第1入出用ポートを解放し、続いて、容器A3の第2入出用ポートに接続した廃液用シリンジを引いて、容器A3の陽圧を解放した。
(Step s4: Expansion culture of iPS cells)
To stop the 14-day incubation in step s3, the syringe pump was stopped. The syringe (Terumo Corporation, 50 mL) was removed to open the first inlet/outlet port, which was a gas port of the container A3, and then the waste liquid syringe connected to the second inlet/outlet port of the container A3 was pulled to release the positive pressure of the container A3.
ロックコネクター(テルモ社製、TS-LC11)、三方活栓(テルモ社製、テルフュージョン)、サンプリング用のシリンジ(テルモ社製、2.5ml)を用いた。ロックコネクターに三方活栓を接続し、拡大培養用の第1番目の容器B1の第3入出用ポートに接続した。三方活栓の側枝に、前記サンプリング用のシリンジを接続した。 A lock connector (Terumo, TS-LC11), a three-way stopcock (Terumo, Telfusion), and a sampling syringe (Terumo, 2.5 ml) were used. The three-way stopcock was connected to the lock connector, and then to the third inlet/outlet port of the first expansion culture container B1. The sampling syringe was connected to the side branch of the three-way stopcock.
先ず、図24に示すように、容器B1の第3入出用ポート方向の三方活栓を閉鎖し、次いで、容器A3の第1入出用ポートのクリップを閉鎖した。
次に、容器B1の第3入出用ポートに接続した三方活栓(テルモ社製、テルフュージョン)を全方向ONにし、容器A3の第3入出用ポートに接続したシリンジ(テルモ社製、50mL)に入った液体培地を全て押し出した。その結果、容器A3の内容物が、容器B1へ移動した。
First, as shown in FIG. 24, the three-way stopcock in the direction of the third inlet/outlet port of the container B1 was closed, and then the clip in the first inlet/outlet port of the container A3 was closed.
Next, a three-way stopcock (Terumo Corporation, Terufusion) connected to the third inlet/outlet port of container B1 was turned on in all directions, and all of the liquid medium contained in a syringe (Terumo Corporation, 50 mL) connected to the third inlet/outlet port of container A3 was pushed out. As a result, the contents of container A3 were transferred to container B1.
三方活栓に接続したサンプリング用のシリンジを引き、細胞懸濁液をサンプリングした。本実施例では、樹立したiPS細胞が収容された容器A3の内容物の全量の1/10の量(1.5ml)をサンプリングした。その後、図25に示すように、該三方活栓を操作し、前記サンプリング用のシリンジの方向をOFFにした。 The sampling syringe connected to the three-way stopcock was pulled to sample the cell suspension. In this example, 1/10 of the total volume (1.5 ml) of the contents of container A3 containing the established iPS cells was sampled. After that, as shown in Figure 25, the three-way stopcock was operated to turn the direction of the sampling syringe OFF.
サンプリングした細胞懸濁液に対して、1/50量のHuman GloLIVE TRA-1-60(R) NorthernLightsTM NL557-conjugated Antibodyを添加し、30分後に液体培地(StemFit AK03)を用いて洗浄を2回繰り返した。蛍光顕微鏡を用いてTRA-1-60陽性細胞を検出した。 To the sampled cell suspension, 1/50 volume of Human GloLIVE TRA-1-60(R) NorthernLights ™ NL557-conjugated Antibody was added, and 30 minutes later, the cells were washed twice with liquid medium (StemFit AK03). TRA-1-60 positive cells were detected using a fluorescent microscope.
容器A3の第3入出用ポートに接続したシリンジ(テルモ社製、50mL)が付いたエクステンションチューブ(シーマン、CTL1001S2)を安全キャビネット内で外した。容器A3の第3入出用ポートを流体ディスペンシングコネクターのキャップを用いて閉鎖した。次に、容器A3の第2入出用ポートに接続した廃液用シリンジに接続したエクステンションチューブを安全キャビネット内で外した。容器A3の第2入出用ポートを流体ディスペンシングコネクターのキャップを用いて閉鎖した。使用済みとなった容器A3のガス用ポート(第1入出用ポート)の閉鎖を確認した。 The extension tube (Seaman, CTL1001S2) with the attached syringe (Terumo, 50 mL) connected to the third input/output port of container A3 was removed inside the safety cabinet. The third input/output port of container A3 was closed using the cap of the fluid dispensing connector. Next, the extension tube connected to the waste liquid syringe connected to the second input/output port of container A3 was removed inside the safety cabinet. The second input/output port of container A3 was closed using the cap of the fluid dispensing connector. It was confirmed that the gas port (first input/output port) of the used container A3 was closed.
次に、容器B1の第3入出用ポートに接続した三方活栓(テルモ社製、テルフュージョン)を安全キャビネット内で外した。エクステンションチューブを介して、液体培地(StemFit AK03)が入ったシリンジ(テルモ社製、50ml)を、容器B1の第3入出用ポートに接続し、該液体培地を継続的に供給した。廃液の排出のために、容器B1の第3入出用ポートに、エクステンションチューブを介して、シリンジ(テルモ社製、50ml)を接続した(廃液バッグを接続しても良く、該廃液バッグは、吸湿性の素材(例えば、吸水性樹脂等、より具体的には、ポリアクリル酸、ポリアクリル酸ナトリウム、ポリアクリル酸共重合体等)や、該素材を含む吸収性物品(例えば、吸水パッド、吸水シート等)を封入した吸湿性の廃液バッグであってもよい)。 Next, the three-way stopcock (Terumo, Terufusion) connected to the third inlet/outlet port of container B1 was removed inside the safety cabinet. A syringe (Terumo, 50 ml) containing liquid medium (StemFit AK03) was connected to the third inlet/outlet port of container B1 via an extension tube, and the liquid medium was continuously supplied. To discharge the waste liquid, a syringe (Terumo, 50 ml) was connected to the third inlet/outlet port of container B1 via an extension tube (a waste liquid bag may be connected, and the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., absorbent pad, absorbent sheet, etc.)).
37℃のホットプレートを設置した安全キャビネット内に装置全体を配置し、7日間の拡大培養を行なった。 The entire device was placed in a safety cabinet equipped with a 37°C hot plate, and expansion culture was carried out for 7 days.
(拡大培養後のiPS細胞の評価)
上記拡大培養後の細胞懸濁液15mlから、10mlのサンプルを取得し、Anti-TRA-1-60,Mouse-Mono(TRA-1-60),NL557,GloLIVE(R&D)を用いてライブ染色を行なった。SRVTM iPSC-2 Vectorからの蛍光タンパク質であるGFPと合わせて、蛍光顕微鏡でコロニー数を目視により確認した。図26に、TRA-1-60陽性+GFP陽性コロニー(iPS細胞のコロニー)数を示す。
(Evaluation of iPS cells after expansion culture)
A 10 ml sample was taken from 15 ml of the cell suspension after the above expansion culture, and live staining was performed using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope together with GFP, a fluorescent protein from the SRV TM iPSC-2 Vector. Figure 26 shows the number of TRA-1-60 positive + GFP positive colonies (iPS cell colonies).
(結果)
図26に示すように、全血4mlから採取したPBMC(1.5×107細胞程度)を用いた拡大培養1回目(P2)で確認できたTRA-1-60陽性+GFP陽性コロニー数は1つであった。
(result)
As shown in FIG. 26, the number of TRA-1-60 positive + GFP positive colonies confirmed in the first expansion culture (P2) using PBMCs (about 1.5×10 7 cells) collected from 4 ml of whole blood was one.
培地容量15ml中の10mlに含まれるiPS細胞のコロニー数は1~2であった。この結果より、全血4mlから採取したPBMC(1.5×107細胞程度)を用いたiPS細胞の樹立コロニー数は1~2であると考えられる。 The number of iPS cell colonies contained in 10 ml of the 15 ml medium volume was 1 to 2. From this result, it is considered that the number of iPS cell colonies established using PBMCs (approximately 1.5 x 107 cells) collected from 4 ml of whole blood is 1 to 2.
実施例2
(市販のPBMCからのiPS細胞の製造)
本実施例では、PBMCとして市販品を用いたこと(即ち、全血の遠心分離工程が無いこと)以外は、上記実施例1と同様に、工程s1~s4を実施し、iPS細胞を得た。
Example 2
(Production of iPS cells from commercially available PBMCs)
In this example, iPS cells were obtained by carrying out steps s1 to s4 in the same manner as in Example 1 above, except that a commercially available PBMC was used (i.e., the centrifugation step of whole blood was omitted).
(工程s1:体細胞(PBMC)に初期化因子を接触させる工程)
バイアルに収容された市販のPBMCをシリンジで採取した。また、市販ベクターはバイアルに充填されているので、これもシリンジで採取した。どちらの操作も臨床用細胞の製造の場面においては、無菌的に接続可能なコネクターを備えた容器に封入されて提供される。
(Step s1: Contacting somatic cells (PBMCs) with reprogramming factors)
Commercially available PBMCs contained in a vial were collected with a syringe. Commercially available vectors, which were also filled in vials, were also collected with a syringe. In both cases, in the production of clinical cells, the vectors are provided sealed in containers equipped with connectors that can be connected aseptically.
シリンジ(テルモ社製、5mL)と注射針(ニプロ社製、NIPRO 18G×1-1/2)を用いて、SRV iPS-2 Vector(4.6×107 CIU/mL)を0.1mLと、市販のPBMC(Human PBMC 10M(PRECISION 93210-10M))とを全量吸い上げた。これを、容器A1の第3入出用ポートに接続されたルアーロックに接続して、該容器1内に注入した。これにより、PBMCに初期化因子を接触させた。
Using a syringe (Terumo, 5 mL) and an injection needle (Nipro, NIPRO 18G×1-1/2), 0.1 mL of SRV iPS-2 Vector (4.6×10 7 CIU/mL) and the entire amount of commercially available PBMC (Human PBMC 10M (PRECISION 93210-10M)) were sucked up. This was connected to a Luer lock connected to the third input/output port of container A1 and injected into said
容器A1を含んだ装置全体を、37℃のホットプレートを設置した安全キャビネット内で2時間インキュベートし、工程s1を完了した。 The entire apparatus, including container A1, was incubated for 2 hours in a safety cabinet equipped with a 37°C hot plate to complete step s1.
工程s2(容器A2も初期化因子の濃度の低減)、工程s3(容器A3でのiPS細胞の樹立)、工程s4(容器B1での拡大培養)を、上記実施例1と同様の条件および同様の操作にて実施した。 Step s2 (reducing the concentration of reprogramming factors in vessel A2), step s3 (establishment of iPS cells in vessel A3), and step s4 (expansion culture in vessel B1) were carried out under the same conditions and with the same procedures as in Example 1 above.
(工程s3完了時点(樹立)でのiPS細胞の評価)
工程s3における14日の培養後(iPS細胞の樹立後)に、容器A3から細胞懸濁液のサンプル1.5mlを取得した。該サンプルに対して、Anti-TRA-1-60, Mouse-Mono(TRA-1-60), NL557, GloLIVE(R&D)を用いてライブ染色を行なった。SRVTM iPSC-2 Vectorからの蛍光タンパク質であるGFPと合わせて蛍光顕微鏡でコロニー数を目視確認した。図27に、TRA-1-60陽性+GFP陽性コロニー(iPS細胞のコロニー)数を示す。
(Evaluation of iPS cells at the completion of step s3 (establishment))
After 14 days of culture in step s3 (after establishment of iPS cells), a 1.5 ml sample of cell suspension was obtained from container A3. The sample was subjected to live staining using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope together with GFP, a fluorescent protein from the SRV ™ iPSC-2 Vector. Figure 27 shows the number of TRA-1-60 positive + GFP positive colonies (iPS cell colonies).
(結果)
図27に示すように、市販のPBMC(1×107cells)を用いた、樹立後で確認できたTRA-1-60陽性+GFP陽性コロニー数は1つであった。
(result)
As shown in FIG. 27, the number of TRA-1-60 positive + GFP positive colonies confirmed after establishment using commercially available PBMC (1×10 7 cells) was one.
(考察)
上記サンプル1.5mlに含まれていたiPSコロニーの数は1つであった。この結果から、市販のPBMCを用いたiPS細胞の樹立コロニーの数は10であったと考えられる。
(Discussion)
The number of iPS colonies contained in the 1.5 ml sample was 1. From this result, it is considered that the number of colonies established from iPS cells using commercially available PBMCs was 10.
(拡大培養後のiPS細胞の評価)
SRVTM iPSC-2 Vector(ときわバイオ)100μLを感染後、Day14の樹立後に2回継代培養(2×Day7)を行なった細胞懸濁液50m1から、10mlのサンプルを取得し、Anti-TRA-1-60, Mouse-Mono(TRA-1-60), NL557, GloLIVE(R&D)を用いてライブ染色を行なった。SRVTM iPSC-2 Vectorからの蛍光タンパク質であるGFPと合わせて、蛍光顕微鏡でコロニー数を目視により確認した。図28に、TRA-1-60陽性+GFP陽性コロニー数を示す。
(Evaluation of iPS cells after expansion culture)
After infection with 100 μL of SRV TM iPSC-2 Vector (Tokiwa Bio), 50 ml of cell suspension was subcultured twice (2 x Day 7) after establishment on Day 14. A 10 ml sample was obtained and live stained using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope along with GFP, a fluorescent protein from the SRV TM iPSC-2 Vector. Figure 28 shows the number of TRA-1-60 positive + GFP positive colonies.
(結果)
図28に示すように、PBMC(1×107cells)を用いた拡大培養2回目(P3)で確認できたTRA-1-60陽性+GFP陽性コロニー数は1つであった。
(result)
As shown in FIG. 28, the number of TRA-1-60 positive + GFP positive colonies confirmed in the second expansion culture (P3) using PBMC (1×10 7 cells) was one.
(考察)
液体培地15ml中の10mlに含まれるiPSコロニー数は1つであった。この結果から市販のPBMC(1×107cells)を用いたiPS細胞の樹立コロニー数は1~2であると考察する。
(Discussion)
The number of iPS colonies contained in 10 ml of the 15 ml liquid medium was 1. From this result, it is considered that the number of iPS cell colonies established using commercially available PBMCs (1 x 107 cells) is 1 to 2.
実施例3
(ヒト全血からのiPS細胞の製造(1))
本実施例では、本発明による細胞製造装置を構成する密閉容器として、GREX 10M-CS(Wilson Wolf Corporation製)を用いたこと以外は、実施例1と同様の工程にて、ヒト全血を遠心分離してPBMCを得、該PBMCからiPS細胞を製造した。
Example 3
(Production of iPS cells from human whole blood (1))
In this example, human whole blood was centrifuged to obtain PBMCs, and iPS cells were produced from the PBMCs in the same manner as in Example 1, except that a GREX 10M-CS (manufactured by Wilson Wolf Corporation) was used as the sealed container constituting the cell production apparatus according to the present invention.
GREX 10M-CSは、容器本体と蓋(マルチポートキャップ)とを有する、概して円筒状の密閉容器である。容器本体の底面は、ガス透過性膜で構成されている。当該容器の容積は、100mlである。本実施例では、この容器を直列的に接続し、工程s1~s4を実施した。 The GREX 10M-CS is a generally cylindrical sealed container having a container body and a lid (multi-port cap). The bottom surface of the container body is made of a gas permeable membrane. The volume of the container is 100 ml. In this example, the containers were connected in series and steps s1 to s4 were carried out.
GREX 10M-CSのマルチポートキャップには、以下の入出用ポートが設けられている。
Sample Line Tubing(該Sample Line Tubingの先端には、MicroClave(登録商標) Connectorが設けられている)。以下、サンプル用ポートともいう。
Reduction Line Tubing:Reduction Line Tubingからは、Weldable Reduction Lineが分岐している。
Harvest Line Tubing:以下、収穫用ポートともいう。該以下、収穫用ポートからは、Weldable Harvest Lineが分岐している。
Pall Versapor Vent Filterが接続されたガス用の入出用ポート:以下、ガス用ポートともいう。
The GREX 10M-CS multi-port cap has the following input and output ports:
Sample Line Tubing (a MicroClave (registered trademark) Connector is provided at the tip of the Sample Line Tubing). Hereinafter, this will also be referred to as the sample port.
Reduction Line Tubing: The Weldable Reduction Line branches off from the Reduction Line Tubing.
Harvest Line Tubing: Hereinafter referred to as the harvest port. Hereinafter, the weldable harvest line branches off from the harvest port.
Gas inlet/outlet port to which a Pall Versapor Vent Filter is connected: hereinafter also referred to as the gas port.
(工程s1:容器A1において、PBMCに初期化因子を接触させる工程)
ヒト全血からのPBMCの取得は、上記実施例1と同様である。
図29に示すように、PBMCを遠心分離後の分画として収容するBD Vacutainer CPT、容器GREX 10M-CS、ルアーロックアクセスデバイス、シリンジ(テルモ社製、50ml)、注射針(テルモ社製、23G×1)、三方活栓R型(テルモ社製、テルフュージョン)を準備した。
(Step s1: contacting PBMC with reprogramming factors in container A1)
The acquisition of PBMCs from human whole blood was carried out in the same manner as in Example 1 above.
As shown in Figure 29, a BD Vacutainer CPT for storing PBMCs as a fraction after centrifugation, a container GREX 10M-CS, a Luer lock access device, a syringe (Terumo Corporation, 50 ml), an injection needle (Terumo Corporation, 23G x 1), and a three-way stopcock type R (Terumo Corporation, Terufusion) were prepared.
容器A1のサンプル用ポートに装着されたMicroClave Connectorに、三方活栓R型とルアーロックアクセスデバイスを接続した。 A three-way stopcock type R and a Luer lock access device were connected to the MicroClave Connector attached to the sample port of container A1.
市販のベクターはバイアルに充填されているので、バイアルからシリンジを用いて該ベクターを採取した。臨床用細胞の製造の場面においては、該初期化用のベクターは、無菌コネクターを介して接続可能な容器に封入された状態で提供される。シリンジは事前に25ml程度引いておく。該シリンジに注射針を装着して、SRV iPS-2 Vector(4.6×107 CIU/ml 0.1ml)を0.1ml(全量)取った。 Since commercially available vectors are filled in vials, the vectors were extracted from the vials using a syringe. In the case of producing clinical cells, the initialization vector is provided sealed in a container that can be connected via a sterile connector. Approximately 25 ml of the syringe is drawn up in advance. An injection needle was attached to the syringe, and 0.1 ml (total amount) of SRV iPS-2 Vector (4.6 x 10 7 CIU/ml 0.1 ml) was extracted.
シリンジから注射針を外し、シリンジを三方活栓R型の側方流路へ接続する。該三方活栓は、容器A1のMicroClave Connector方向の流路をOFFにした。PBMCを遠心分離したBD Vacutainer CPTを、前記ルアーロックアクセスデバイスへセットした。BD Vacutainer CPTを、ルアーロックアクセスデバイスへ押し込んだ。三方活栓R型の側方流路へ接続したシリンジで吸引することで、BD Vacutainer CPT内容液が該シリンジ内へ流入して、事前に封入していたベクターと混和された。三方活栓のコックを回し、ルアーロックアクセスデバイス方向の流路をOFFにした。 The injection needle was removed from the syringe and the syringe was connected to the side flow path of the R-type three-way stopcock. The three-way stopcock was turned off in the flow path toward the MicroClave Connector of container A1. The BD Vacutainer CPT, which had been centrifuged to remove PBMCs, was set in the Luer lock access device. The BD Vacutainer CPT was pushed into the Luer lock access device. By aspirating with the syringe connected to the side flow path of the R-type three-way stopcock, the contents of the BD Vacutainer CPT flowed into the syringe and were mixed with the vector that had been sealed in beforehand. The stopcock of the three-way stopcock was turned off in the flow path toward the Luer lock access device.
血清とPBMCの入ったシリンジを90度回転させて垂直姿勢とし、内容液をシリンジ下底に保持したままシリンジを押し込み、細胞懸濁液を容器A1内へ流入させた。
ルアーロックアクセスデバイスからBD Vacutainer CPTを外した。
CO2インキュベーター内で2時間インキュベートした。
The syringe containing the serum and PBMCs was rotated 90 degrees to a vertical position, and the syringe was pushed in while the content liquid was held at the bottom of the syringe, causing the cell suspension to flow into container A1.
The BD Vacutainer CPT was removed from the Luer lock access device.
Incubated in a CO2 incubator for 2 hours.
(工程s2:容器A2において、液体培地中の初期化因子の濃度を低減させる工程)
容器A1のReduction Line Tubingのみを解放し、ガス用ポートと収穫用ポートを閉鎖した。
液体培地StemFit AK03が50ml入ったシリンジを容器A1のReduction LineTubingへ接続して液体培地を50ml注入した。その結果、容器A1の内容物は、注入した50mlの液体培地と共に容器A2へ流入した。
容器A1のReduction Line Tubingを閉鎖した。
細胞(PBMC)が容器A2内で自然沈下するまでCO2インキュベーター内で30分間静置した。
この段階で、容器A2には、元のバイアルに対して1/1量のPBMCと、元のバイアルに対して500倍に希釈された初期化ベクターが収容された状態となった。
(Step s2: Reducing the concentration of reprogramming factors in the liquid medium in container A2)
Only the Reduction Line Tubing of the vessel A1 was opened, and the gas port and the harvest port were closed.
A syringe containing 50 ml of StemFit AK03 liquid medium was connected to the Reduction Line Tubing of the container A1, and 50 ml of the liquid medium was injected. As a result, the contents of the container A1 flowed into the container A2 together with the injected 50 ml of liquid medium.
The Reduction Line Tubing of vessel A1 was closed.
The cells (PBMC) were allowed to stand in a CO 2 incubator for 30 minutes until they naturally settled in container A2.
At this stage, container A2 contained PBMCs in an amount equal to 1/1 of the amount in the original vial, and the initialization vector diluted 500-fold compared to the amount in the original vial.
30分後に装置をCO2インキュベーターから取り出し、安全キャビネット内で容器内の上清を除去する操作を行なった。容器A2のWeldable Reduction Lineのチューブを解放し、廃液回収用の空のシリンジ(50ml)を接続した(廃液バッグを用いても良く、該廃液バッグは、吸湿性の素材(例えば、吸水性樹脂等、より具体的には、ポリアクリル酸、ポリアクリル酸ナトリウム、ポリアクリル酸共重合体等)や、該素材を含む吸収性物品(例えば、吸水パッド、吸水シート等)を封入した吸湿性の廃液バッグであってもよい)。 After 30 minutes, the device was removed from the CO2 incubator, and the supernatant in the container was removed in a safety cabinet. The tube of the Weldable Reduction Line of the container A2 was opened, and an empty syringe (50 ml) for collecting waste liquid was connected (a waste liquid bag may be used, and the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.).
容器A2のガス用ポートの開口部へ空気を50ml注入するために、プランジャーを引いたシリンジ(50ml)を、流体ディスペンシングコネクターを介して接続した。
容器A2のガス用ポートのチューブを解放した。
容器A2のガス用ポートへ接続したシリンジから空気を容器A2内に押し込んだ。その結果、容器A2内の液体培地の上清(容器本体内の上端から8cmまでの領域に存在する液体)が、容器A2のWeldable Reduction Lineに接続された廃液回収用のシリンジ(50ml)へ廃液として回収された。
A syringe (50 ml) with a retracted plunger was connected via the fluid dispensing connector to inject 50 ml of air into the opening of the gas port of container A2.
The tube of the gas port of vessel A2 was opened.
Air was forced into container A2 from a syringe connected to the gas port of container A2. As a result, the supernatant of the liquid medium in container A2 (the liquid present in the region from the top end of the container body to 8 cm) was collected as waste liquid into a waste liquid collecting syringe (50 ml) connected to the weldable reduction line of container A2.
容器A2内には底から2cm程度までの高さに存在するPBMCを含む液体培地が残った。
容器A2のガス用ポートのチューブ、および、Weldable Reduction Lineのチューブを閉鎖した。
この段階で、容器A2には、元のバイアルに対して1/1の量のPBMCと、元のバイアルに対して500倍に希釈された初期化ベクター(元バイアルに対して)が収容された状態となった。
In container A2, the liquid medium containing PBMCs remained at a height of about 2 cm from the bottom.
The gas port tube of vessel A2 and the weldable reduction line tube were closed.
At this stage, container A2 contained PBMCs in an amount equal to 1/1 of the amount in the original vial, and the initialization vector (relative to the original vial) diluted 500-fold compared to the original vial.
(工程s3:14日間の培養によるiPS細胞の樹立)
容器A2のWeldable Harvest Lineを容器A3のMicroClave Connectorへ接続する。MicroClave Connectorは、一般環境においても1回限りであればコネクターを接続可能である(再接続はできない)。
容器A2の収穫用ポートから、シリンジに収容された10mlのアテロコラーゲンビーズ溶液(KOKEN:MIC-00)を安全キャビネット内で注入した。
液体培地StemFit AK03が50ml入ったシリンジを、容器A2のReduction Line Tubingに接続して、該液体培地を50ml注入した。
その結果、容器A2の内容液は、注入した50ml液体培地と共に、容器A3へ流入した。その後、容器A2のReduction Line Tubingを閉鎖した。装置を、配管接続を外すことなく、CO2インキュベーターへ収納し、14日間の培養を開始した。
(Step s3: Establishment of iPS cells by culturing for 14 days)
The Weldable Harvest Line of the container A2 is connected to the MicroClave Connector of the container A3. The MicroClave Connector can be connected only once in a general environment (it cannot be reconnected).
10 ml of an atelocollagen bead solution (KOKEN: MIC-00) contained in a syringe was injected from the harvesting port of container A2 in a safety cabinet.
A syringe containing 50 ml of the liquid medium StemFit AK03 was connected to the Reduction Line Tubing of the container A2, and 50 ml of the liquid medium was injected.
As a result, the liquid content of the container A2 flowed into the container A3 together with the injected 50 ml liquid medium. After that, the Reduction Line Tubing of the container A2 was closed. The device was placed in a CO2 incubator without disconnecting the piping, and a 14-day culture was started.
本実施例では、14日間の培養の途中の7日目に、容器A3の培地液量を100ml量まで追加の注入を行なった。この段階で、容器A3には元のバイアルに対して1/1量のPBMCと、元のバイアルに対して3000倍に希釈された初期化ベクターが収容された状態となった。 In this example, on the 7th day of the 14-day culture, additional medium was injected to increase the volume of the medium in container A3 to 100 ml. At this stage, container A3 contained 1/1 the amount of PBMCs compared to the original vial and initialization vector diluted 3000 times compared to the original vial.
安全キャビネット内で作業者が実施した各種の接続は、実際の製造では事前に実施されている。また、液体培地の注入は、エクステンションチューブ(シーマン CTL1001S2)を介してシリンジから供給した。そのために、図32に示すように、シリンジポンプとしてク-デックシリンジポンプCSP-120(大研医器株式会社 CSP-120)を用い、シリンジポンプの機器を固定するためにポンプユナイタースタンド(大研医器株式会社: PUS-200S)とポンプユナイター(大研医器株式会社: PU3-200S)を用いた。 All the connections made by the operator in the safety cabinet were made in advance in the actual production. The liquid medium was injected from a syringe via an extension tube (Seaman CTL1001S2). For this purpose, as shown in Figure 32, a Coudec Syringe Pump CSP-120 (Daiken Medical Co., Ltd. CSP-120) was used as the syringe pump, and a Pump Uniter Stand (Daiken Medical Co., Ltd.: PUS-200S) and a Pump Uniter (Daiken Medical Co., Ltd.: PU3-200S) were used to secure the syringe pump equipment.
(14日間の培養の途中の7日目に液体培地を追加注入する操作)
容器A3のReduction Line Tubingに接続されたチューブのクリップを解放し、50mLの液体培地(StemFit AK03)が入ったシリンジ(テルモ社製、50ml)を接続した。容器A2のWeldable Harvest Lineをクリップで閉鎖した。図33に示すように、シリンジから液体培地を容器A3へ入れた。Reduction Line Tubingのチューブのクリップを閉鎖した。
(Operation of injecting additional liquid medium on the 7th day of the 14-day culture)
The clip of the tube connected to the Reduction Line Tubing of the container A3 was released, and a syringe (Terumo, 50 ml) containing 50 mL of liquid medium (StemFit AK03) was connected. The Weldable Harvest Line of the container A2 was closed with a clip. As shown in FIG. 33, the liquid medium was poured from the syringe into the container A3. The clip of the tube of the Reduction Line Tubing was closed.
(工程s4:iPS細胞の拡大培養)
容器A3のWeldable Reduction Lineへシリンジ(JMS、100ml)を接続した(廃液バッグでも代替え可能であり、該廃液バッグは、吸湿性の素材(例えば、吸水性樹脂等、より具体的には、ポリアクリル酸、ポリアクリル酸ナトリウム、ポリアクリル酸共重合体等)や、該素材を含む吸収性物品(例えば、吸水パッド、吸水シート等)を封入した吸湿性の廃液バッグであってもよい)。容器A3のガス用ポートに流体ディスペンシングコネクターを接続して、空気を満たしたシリンジ(テルモ社製、50ml)を接続した。該容器A3のMicroClave Connectorへ接続した容器A2のWeldable HarvestLineのクリップを閉鎖した。
シリンジから空気を送り、容器A3内にある上清の培地を廃液用のシリンジへ排出する。この操作を2回繰り返し、流体ディスペンシングコネクターに接続したシリンジを外して空気を再度満たす。容器A3内にある上清の全てを廃液用のシリンジへ排出した。その後、逆流防止のために容器A3のガス用ポートのクリップを閉鎖した。また、廃液の逆流を防ぐために、容器A3のWeldable ReductionLineをクリップで閉鎖した。
(Step s4: Expansion culture of iPS cells)
A syringe (JMS, 100 ml) was connected to the Weldable Reduction Line of the container A3 (a waste liquid bag can be used instead, and the waste liquid bag may be a hygroscopic waste liquid bag containing a hygroscopic material (e.g., water-absorbent resin, more specifically, polyacrylic acid, sodium polyacrylate, polyacrylic acid copolymer, etc.) or an absorbent article containing the material (e.g., water-absorbent pad, water-absorbent sheet, etc.). A fluid dispensing connector was connected to the gas port of the container A3, and a syringe filled with air (Terumo, 50 ml) was connected. The clip of the Weldable Harvest Line of the container A2 connected to the MicroClave Connector of the container A3 was closed.
Air was sent from the syringe, and the supernatant medium in the container A3 was discharged into the syringe for waste liquid. This operation was repeated twice, and the syringe connected to the fluid dispensing connector was removed and filled with air again. All of the supernatant in the container A3 was discharged into the syringe for waste liquid. After that, the clip of the gas port of the container A3 was closed to prevent backflow. In addition, the Weldable Reduction Line of the container A3 was closed with a clip to prevent backflow of the waste liquid.
図34に示すように、容器A3のWeldable Harvest Lineと、三方活栓(テルフュージョンTERUMO)を介して、容器B1のMicroClave Connectorとを接続した。三方活栓には、サンプル取得用のシリンジ(テルモ社製、2.5ml)が接続されている。MicroClave Connector は、一般環境においても1回限りであればコネクターを接続可能である(再接続はできない)。 As shown in Figure 34, the Weldable Harvest Line of container A3 was connected to the MicroClave Connector of container B1 via a three-way stopcock (Terufusion TERUMO). A syringe (Terumo, 2.5 ml) for obtaining samples was connected to the three-way stopcock. The MicroClave Connector can be connected once even in a general environment (it cannot be reconnected).
容器A3内に残留した20ml程度の培地をそのまま、容器B1へ移動させることも可能であるが、アテロコラーゲンビーズを10ml含む液体は、流路を閉塞させる可能性があるので、先に液体培地(StemFit AK03)を50ml入れて作製した細胞懸濁液を容器A3から容器B1へ移動させる。容器A3内の液体培地を流入させるために、容器B1のガス用ポートのクリップを解放した。容器A3内へ収穫用ポートから液体培地を50ml入れた。該液体培地を入れた後、容器A3の収穫用ポートのクリップを閉鎖した。容器B1のMicroClave Connectorを全方向ONにした。容器A3のガス用ポートに流体ディスペンシングコネクターを接続して空気を満たしたシリンジ(テルモ社製、50ml)を接続した。容器A3内へガス用ポートからシリンジで空気を入れた。 It is possible to transfer the approximately 20 ml of medium remaining in container A3 directly to container B1, but since the liquid containing 10 ml of atelocollagen beads may clog the flow path, a cell suspension prepared by first adding 50 ml of liquid medium (StemFit AK03) is transferred from container A3 to container B1. To allow the liquid medium in container A3 to flow in, the clip on the gas port of container B1 was released. 50 ml of liquid medium was added to container A3 through the harvesting port. After the liquid medium was added, the clip on the harvesting port of container A3 was closed. The MicroClave Connector of container B1 was turned ON in all directions. A fluid dispensing connector was connected to the gas port of container A3, and an air-filled syringe (Terumo, 50 ml) was connected. Air was introduced into container A3 through the gas port using the syringe.
容器A3に空気を入れた後に、ガス用ポートのクリップを閉鎖した。容器A3から容器B1へ内容液を移動させた後、サンプル取得用のシリンジにより細胞懸濁液を採取した。採取後には三方活栓を操作し、サンプル取得用のシリンジの方向をOFFにした。その後、容器A3のWeldableHarvest Lineをクリップで閉鎖した。 After air was introduced into container A3, the clip on the gas port was closed. After transferring the contents from container A3 to container B1, a cell suspension was collected using a sample collection syringe. After collection, the three-way stopcock was operated to turn the direction of the sample collection syringe OFF. The Weldable Harvest Line of container A3 was then closed with a clip.
サンプル取得した細胞懸濁液に対して、1/50量のHuman GloLIVE TRA-1-60(R) NorthernLightsTM NL557-conjugated Antibodyを添加し、30分後に液体培地(StemFit AK03)を用いて洗浄を2回繰り返した。蛍光顕微鏡を用いてTRA-1-60陽性細胞を検出した。 A 1/50 volume of Human GloLIVE TRA-1-60® NorthernLights ™ NL557-conjugated Antibody was added to the cell suspension obtained from the sample, and after 30 minutes, the cells were washed twice with liquid medium (StemFit AK03). TRA-1-60 positive cells were detected using a fluorescent microscope.
装置の配管を外すことなく、CO2インキュベーター内に格納し、7日間の培養を行なった。 Without removing the piping from the device, the device was stored in a CO 2 incubator and cultured for 7 days.
(拡大培養後のiPS細胞の評価)
上記拡大培養(1回継代培養(1×Day7))を行なった細胞懸濁液50mlから、10mlのサンプルを取得した。Anti-TRA-1-60, Mouse-Mono(TRA-1-60), NL557, GloLIVE(R&D)を用いてライブ染色を行なった。SRVTM iPSC-2 Vectorからの蛍光タンパク質であるGFPと合わせて蛍光顕微鏡でコロニー数を目視により確認した。図35に、TRA-1-60陽性+GFP陽性コロニー(iPS細胞のコロニー)数を示す。
(Evaluation of iPS cells after expansion culture)
A 10 ml sample was obtained from 50 ml of the cell suspension after the above expansion culture (single subculture (1x Day 7)). Live staining was performed using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope together with GFP, a fluorescent protein from the SRV ™ iPSC-2 Vector. Figure 35 shows the number of TRA-1-60 positive + GFP positive colonies (iPS cell colonies).
(結果)
図35に示すように、全血4mlから採取したPBMC(1.5×107細胞程度)を用いた拡大培養1回目(P2)で確認できたTRA-1-60陽性+GFP陽性コロニー数は1つであった。
(result)
As shown in FIG. 35, the number of TRA-1-60 positive + GFP positive colonies confirmed in the first expansion culture (P2) using PBMCs (about 1.5×10 7 cells) collected from 4 ml of whole blood was one.
(考察)
培地容量50ml中の10mlに含まれるiPSコロニー数は1つであった。この結果より、全血4mlから採取したPBMC(1.5×107細胞程度)を用いたiPS細胞の樹立ロニー数は、5つであると考えられる。
(Discussion)
The number of iPS colonies contained in 10 ml of the 50 ml medium volume was 1. From this result, it is considered that the number of Ronnies of iPS cells established using PBMCs (approximately 1.5 × 10 7 cells) collected from 4 ml of whole blood was 5.
(ヒト全血からのiPS細胞の製造(2))
上記実施例3の製造(1)の結果を受けて、さらに、初期化因子の量を5倍に上げた態様でも実施した。
製造(2)では、(工程s1:容器A1において、PBMCに初期化因子を接触させる工程)でPBMCに接触させるベクター量を5倍に増量し(SRV iPS-2 Vector(4.6×107 CIU/ml 0.1ml)を0.5ml使用)、(工程s3:14日間の培養によるiPS細胞の樹立)終了後の細胞懸濁液50mlから上清を除去した10mlのサンプルを取得し、iPS細胞の評価を行った。
その結果、全血4mlから採取したPBMC(1.5×107細胞程度)を用いた樹立直後(Day14)に確認できたTRA-1-60陽性+GFP陽性コロニー数は3つであった。よって、実施条件を変えれば、初期化効率はさらに上昇することが示された。
(Production of iPS cells from human whole blood (2))
Based on the results of the production (1) in Example 3 above, an embodiment was also carried out in which the amount of the reprogramming factor was increased by 5 times.
In production (2), the amount of vector contacted with PBMCs in step s1: contacting PBMCs with reprogramming factors in container A1 was increased five-fold (0.5 ml of SRV iPS-2 Vector (4.6 x 10 CIU/ml, 0.1 ml) was used), and a 10 ml sample was obtained by removing the supernatant from 50 ml of cell suspension after step s3: establishment of iPS cells by culturing for 14 days, and the iPS cells were evaluated.
As a result, the number of TRA-1-60 positive + GFP positive colonies confirmed immediately after establishment (Day 14) using PBMCs (approximately 1.5 × 10 cells) collected from 4 ml of whole blood was 3. This indicates that the reprogramming efficiency can be further increased by changing the implementation conditions.
実施例4
(市販のPBMCからのiPS細胞の製造)
本実施例では、PBMCとして市販品を用いたこと(即ち、全血の遠心分離工程が無いこと)以外は、上記実施例3と同様に、工程s1~s4を実施し、iPS細胞を得た。
Example 4
(Production of iPS cells from commercially available PBMCs)
In this example, iPS cells were obtained by carrying out steps s1 to s4 in the same manner as in Example 3 above, except that a commercially available PBMC was used (i.e., the centrifugation step of whole blood was omitted).
(拡大培養後のiPS細胞の評価)
SRVTM iPSC-2 Vector(ときわバイオ)100μlを感染後、Day14の樹立後に2回継代培養(2×Day7)を行なった細胞懸濁液50mLから、10Mlサンプルを取得し、Anti-TRA-1-60, Mouse-Mono(TRA-1-60), NL557, GloLIVE(R&D)を用いてライブ染色を行なった。SRVTM iPSC-2 Vectorからの蛍光タンパク質であるGFPと合わせて、蛍光顕微鏡でコロニー数を目視により確認した。図36に、TRA-1-60陽性+GFP陽性コロニー(iPS細胞のコロニー)数を示す。
(Evaluation of iPS cells after expansion culture)
After infection with 100 μl of SRV TM iPSC-2 Vector (Tokiwa Bio), 50 mL of cell suspension was subcultured twice (2×Day 7) after establishment on Day 14. A 10 mL sample was obtained and live stained using Anti-TRA-1-60, Mouse-Mono (TRA-1-60), NL557, and GloLIVE (R&D). The number of colonies was visually confirmed under a fluorescent microscope together with GFP, a fluorescent protein from the SRV TM iPSC-2 Vector. Figure 36 shows the number of TRA-1-60 positive + GFP positive colonies (colonies of iPS cells).
(結果)
図36の図に示すように、PBMC(1×107cells)を用いた拡大培養2回目(P3)で確認できたTRA-1-60陽性+GFP陽性コロニー数は1つであった。
(result)
As shown in the diagram of FIG. 36, the number of TRA-1-60 positive + GFP positive colonies confirmed in the second expansion culture (P3) using PBMC (1×10 7 cells) was one.
(考察)
液体培地50ml中の10mlに含まれるiPSコロニー数は1つであった。この結果から市販のPBMC(1×107cells)を用いたiPS細胞の樹立コロニー数は5であると考察する。
(Discussion)
The number of iPS colonies contained in 10 ml of the 50 ml liquid medium was 1. From this result, it is considered that the number of iPS cell colonies established using commercially available PBMCs (1×10 7 cells) was 5.
実施例5
(iPS細胞から心筋細胞の製造・評価(iPS細胞を分化誘導する工程s5))
本実施例では、本発明による細胞製造装置を用い、本発明による分化細胞の製造方法を実施し、実際にiPS細胞を分化誘導して心筋細胞を製造した。
本発明による分化細胞の製造方法では、好ましい態様として、本発明の製造方法によって得られたiPS細胞が用いられるが、本実施例では、京都大学iPS細胞研究財団提供のiPS細胞(ヒト臨床用iPS細胞の研究用株(15M66))を用いて、本発明による分化細胞の製造方法(上記工程s5)を実施した。
Example 5
(Production and evaluation of cardiomyocytes from iPS cells (step s5 of inducing differentiation of iPS cells))
In this example, the cell production device according to the present invention was used to carry out the method for producing differentiated cells according to the present invention, and the differentiation of iPS cells was actually induced to produce cardiomyocytes.
In a preferred embodiment of the method for producing differentiated cells according to the present invention, iPS cells obtained by the production method of the present invention are used. In this example, the method for producing differentiated cells according to the present invention (step s5 above) was carried out using iPS cells provided by the iPS Cell Research Foundation, Kyoto University (a research strain of human clinical iPS cells (15M66)).
本実施例では、iPS細胞を分化誘導する工程s5が、さらに3つの工程(s5-1、s5-2、s5-3)に分かれており、これらの各工程を、下記のように、3つの密閉容器(C1、C2、C3)に1対1で対応付け、容器から容器へと内容物を送りながら、各容器内で各工程を順次実施した。
工程s5-1:容器C1内において、iPS細胞を分化誘導し、中胚葉を経て、心臓中胚葉を得る工程。
工程s5-2:心臓中胚葉を容器C2へと移動させ、該容器C2内において、前記の心臓中胚葉を分化誘導し、心筋前駆細胞を得る工程。
工程s5-3:心筋前駆細胞を容器C3へと移動させ、該容器C3内において、前記の心筋前駆細胞を分化誘導し、心筋細胞を得る工程。
In this example, step s5 of inducing differentiation of iPS cells is further divided into three steps (s5-1, s5-2, s5-3), and each of these steps is assigned one-to-one to three sealed containers (C1, C2, C3) as described below. Each step is carried out sequentially in each container while the contents are transferred from container to container.
Step s5-1: A step of inducing differentiation of iPS cells in the container C1 to obtain cardiac mesoderm via mesoderm.
Step s5-2: A step of transferring the cardiac mesoderm to a container C2, and inducing differentiation of the cardiac mesoderm in the container C2 to obtain cardiac progenitor cells.
Step s5-3: a step of transferring myocardial precursor cells to a container C3, and inducing differentiation of the myocardial precursor cells in the container C3 to obtain cardiomyocytes.
本実施例では、前記3つの密閉容器として、上記実施例3および4で用いた容器と同様のGREX 10M-CSを用いた。
工程s5-1、s5-2、s5-3を実施するための細胞製造装置の概要は、図12に示すとおりであり、同図の構成におけるq=3の場合に該当する。また、密閉容器GREX 10M-CSの外観は、図29に示したとおりである。
In this example, the three sealed containers were made of GREX 10M-CS, which was the same as the containers used in Examples 3 and 4 above.
The outline of the cell manufacturing apparatus for carrying out steps s5-1, s5-2, and s5-3 is as shown in Fig. 12, and corresponds to the case where q = 3 in the configuration of the same figure. Also, the external appearance of the sealed container GREX 10M-CS is as shown in Fig. 29.
(工程s5-1(iPS細胞から心臓中胚葉への分化誘導))
先ず、容器C1に、iPS細胞株(15M66、1×106cells)と、アテロコラーゲンビーズ溶液(KOKEN:MIC-00)と、液体培地StemFit AK03(味の素)10mL(該液体培地にはY-27632 (富士フィルムWako)10μM濃度が含まれている)を注入した。
次に、該容器C1に、PSC Cardiomyocyte Differentiation Kit(サーモフィッシャーサイエンティフィック社)のA培地を20ml注入した。
さらに、前記A培地を0.5ml/hの流量にて継続的に供給し、容器内の培地を排出させながら、2日間培養した。これにより、iPS細胞の分化誘導が行なわれて、中胚葉を経て、心臓中胚葉が得られた。
(Step s5-1 (induction of differentiation of iPS cells into cardiac mesoderm))
First, an iPS cell line (15M66, 1 x 10 6 cells), an atelocollagen bead solution (KOKEN: MIC-00), and 10 mL of liquid medium StemFit AK03 (Ajinomoto) (containing 10 μM concentration of Y-27632 (Fujifilm Wako)) were poured into container C1.
Next, 20 ml of medium A from a PSC Cardiomyocyte Differentiation Kit (Thermo Fisher Scientific) was poured into the container C1.
The medium A was continuously supplied at a flow rate of 0.5 ml/h and cultured for 2 days while discharging the medium from the vessel, thereby inducing differentiation of the iPS cells into mesoderm and then cardiac mesoderm.
(容器C1の内容物を、容器C2に移動させる操作)
先ず、容器C1に空気を送り込み、容器C1内の上澄み部分を廃液用のシリンジに押し出した(容器内への空気の送り込みによって、上澄み部分が押し出されるように、他の入出用ポートは適宜に閉鎖した)。その結果、容器C1の底部には、沈殿していた心臓中胚葉とアテロコラーゲンビーズとそれらを含んだ少量の液体培地が残った。
次に入出用ポートの接続を切り替え、容器C1にシリンジにて空気を注入し、該底部に残っていた沈殿部分(心臓中胚葉とアテロコラーゲンビーズと少量の液体培地)を容器C2へと押し出して、該沈殿部分の移動を完了した。
(Operation of transferring the contents of container C1 to container C2)
First, air was pumped into the container C1, and the supernatant in the container C1 was pushed out into the waste liquid syringe (other inlet/outlet ports were appropriately closed so that the supernatant was pushed out by pumping air into the container). As a result, the precipitated cardiac mesoderm and atelocollagen beads, as well as a small amount of liquid medium containing them, remained at the bottom of the container C1.
Next, the connection of the input/output ports was switched, air was injected into container C1 using a syringe, and the sediment remaining at the bottom (cardiac mesoderm, atelocollagen beads, and a small amount of liquid medium) was pushed out into container C2, completing the movement of the sediment.
(工程s5-2(心臓中胚葉から心筋前駆細胞への分化誘導))
先ず、前記の容器C2内に、PSC Cardiomyocyte Differentiation Kit(サーモフィッシャーサイエンティフィック社)のB培地を20ml注入した、
次に、前記B培地を0.5ml/hの流量にて継続的に供給し、容器内の培地を排出させながら、2日間培養した。これにより、心臓中胚葉の分化誘導が行なわれて、心筋前駆細胞が得られた。
(Step s5-2 (induction of differentiation from cardiac mesoderm to cardiac progenitor cells))
First, 20 ml of medium B from the PSC Cardiomyocyte Differentiation Kit (Thermo Fisher Scientific) was poured into the container C2.
Next, the B medium was continuously supplied at a flow rate of 0.5 ml/h and cultured for 2 days while discharging the medium from the vessel, thereby inducing differentiation of cardiac mesoderm to obtain cardiac progenitor cells.
(容器C2の内容物を、容器C3に移動させる操作)
上記の容器C1の内容物を容器C2に移動させる操作と同様に、容器C2に空気を送り込み、容器C2内の上澄み部分を廃液用のシリンジに押し出した。その結果、容器C2の底部には、沈殿していた心筋前駆細胞とアテロコラーゲンビーズとそれらを含んだ少量の液体培地が残った。
次に入出用ポートの接続を切り替え、容器C2にシリンジにて空気を注入し、該底部に残っていた沈殿部分(心筋前駆細胞とアテロコラーゲンビーズと少量の液体培地)を容器C3へと押し出して、該沈殿部分の移動を完了した。
(Operation of moving the contents of container C2 to container C3)
In the same manner as in the above operation of transferring the contents of container C1 to container C2, air was pumped into container C2, and the supernatant in container C2 was pushed out into a syringe for waste liquid. As a result, the precipitated myocardial progenitor cells and atelocollagen beads, as well as a small amount of liquid medium containing them, remained at the bottom of container C2.
Next, the connection of the input/output ports was switched, air was injected into container C2 using a syringe, and the precipitate remaining at the bottom (myocaloid progenitor cells, atelocollagen beads, and a small amount of liquid medium) was pushed out into container C3, completing the movement of the precipitate.
(工程s5-3)心筋前駆細胞から心筋細胞への分化誘導
先ず、前記の容器C3内に、PSC Cardiomyocyte Differentiation Kit(サーモフィッシャーサイエンティフィック社)のC培地を20ml注入した。
次に、前記C培地を0.5ml/hの流量にて継続的に供給し、容器内の培地を排出させながら、10日間培養した。これにより、心筋前駆細胞の分化誘導が行なわれて、心筋細胞が得られた。
(Step s5-3) Induction of Differentiation from Cardiac Muscle Progenitor Cells to Cardiac Muscles First, 20 ml of C medium from the PSC Cardiomyocyte Differentiation Kit (Thermo Fisher Scientific) was poured into the container C3.
Next, the C medium was continuously supplied at a flow rate of 0.5 ml/h, and the medium was discharged from the vessel while the vessel was cultured for 10 days, thereby inducing differentiation of the cardiac muscle precursor cells to obtain cardiac muscle cells.
(心筋細胞マーカー陽性細胞)
Day14において容器内容物をサンプリングし、心筋細胞のマーカータンパク質であるトロポニンT(TNNT2)について、定法に従って免疫染色を行った(抗トロポニン抗体:(BD PharmingenAlexa Fluor 647 Anti-CardiacTroponin T(BD 565744))使用)。Hoechst染色(核染色)との二重染色結果を図37に示す。図37に示されるように、アテロコラーゲン上の細胞集塊中に、Hoechstシグナル(核)の周囲にトロポニンTシグナルを有する細胞(すなわち、心筋細胞)が多数観察された。よって、本発明の製造方法により、iPS細胞から心筋細胞が効率よく分化誘導されたことが明らかになった。
(Cardiomyocyte marker positive cells)
On day 14, the contents of the container were sampled and immunostained for troponin T (TNNT2), a marker protein for cardiomyocytes, according to a standard method (anti-troponin antibody: (BD Pharmingen Alexa Fluor 647 Anti-Cardiac Troponin T (BD 565744)) was used). The results of double staining with Hoechst staining (nuclear staining) are shown in FIG. 37. As shown in FIG. 37, many cells (i.e., cardiomyocytes) having troponin T signals around the Hoechst signals (nuclei) were observed in the cell clusters on the atelocollagen. Therefore, it was revealed that the production method of the present invention efficiently induced differentiation of cardiomyocytes from iPS cells.
(心筋細胞・心筋前駆細胞マーカーの発現誘導)
Day14において容器内容物(工程s5-3)をサンプリングし、コラゲナーゼ処理を行ってアテロコラーゲンビーズを溶解した。得られた細胞ペレットからSuperPrep II Cell Lysis & RT Kit for qPCR(東洋紡株式会社: SCQ-401)を用いてmRNAを抽出し、cDNAを合成した。StepOnePlus system (Life Technologies, Carlsbad, CA, USA)、および、Luna Universal qPCR Master Mix (New England Biolabs Inc., Ipswich, MA, USA)を用いてリアルタイムPCR解析を行い、T brachyury(中胚葉マーカー)、NKX2.5(心筋細胞マーカー:心筋特異的転写因子)、KDRとISL1(心筋前駆細胞マーカー)の発現量を解析した。コントロールには、分化誘導を行わずにアテロコラーゲン上で培養したiPS細胞株(15M66)を用いた。結果を図38に示す。
(Induction of expression of cardiomyocyte and cardiac progenitor cell markers)
On day 14, the contents of the container (step s5-3) were sampled and treated with collagenase to dissolve the atelocollagen beads. From the obtained cell pellet, mRNA was extracted using SuperPrep II Cell Lysis & RT Kit for qPCR (Toyobo Co., Ltd.: SCQ-401), and cDNA was synthesized. Real-time PCR analysis was performed using the StepOnePlus system (Life Technologies, Carlsbad, CA, USA) and Luna Universal qPCR Master Mix (New England Biolabs Inc., Ipswich, MA, USA), and the expression levels of T brachyury (mesodermal marker), NKX2.5 (cardiomyocyte marker: cardiac muscle-specific transcription factor), KDR and ISL1 (cardiac progenitor cell marker) were analyzed. As a control, an iPS cell line (15M66) cultured on atelocollagen without differentiation induction was used. The results are shown in FIG. 38.
図38に示されるように、中胚葉マーカー(T brachyury)の発現はコントロール、分化誘導後の細胞のいずれにおいても検出されなかった。これに対し、分化誘導後の細胞では心筋細胞マーカー(NKX2.5とTNNT2)の発現が非常に高く(コントロールと比べて約8~10倍)、心筋前駆細胞マーカー(KDRとISL1)の発現も高いこと(コントロールと比べて約4~6倍)が確認された。
よって、本法により、iPSから心筋前駆細胞を経て心筋細胞への分化が順調に誘導されたことが示唆された。これらの結果より、本発明の製造方法および製造装置を用いて、細胞の分化誘導が行えることが示された。
As shown in Figure 38, the expression of mesodermal marker (T brachyury) was not detected in either the control or differentiation-induced cells. In contrast, the expression of cardiomyocyte markers (NKX2.5 and TNNT2) was very high (approximately 8-10 times higher than the control) and the expression of cardiac progenitor cell markers (KDR and ISL1) was also high (approximately 4-6 times higher than the control) in the differentiation-induced cells.
This suggests that the differentiation of iPS cells into cardiomyocytes via myocardial progenitor cells was successfully induced by this method. These results demonstrate that cell differentiation can be induced using the production method and production device of the present invention.
実施例6
(iPS細胞から膵前駆細胞の製造・評価(iPS細胞を分化誘導する工程s5))
本実施例では、本発明による細胞製造装置を用い、本発明による分化細胞の製造方法を実施し、実際にiPS細胞を分化誘導して膵前駆細胞を製造した。本発明による分化細胞の製造方法では、好ましい態様として、本発明の製造方法によって得られたiPS細胞が用いられるが、本実施例では、京都大学iPS細胞研究財団提供のiPS細胞(ヒト臨床用iPS細胞の研究用株(15M66))を用いて、本発明による分化細胞の製造方法(上記工程s5)を実施した。
Example 6
(Production and evaluation of pancreatic progenitor cells from iPS cells (step s5 of inducing differentiation of iPS cells))
In this example, the cell manufacturing device according to the present invention was used to carry out the method for manufacturing differentiated cells according to the present invention, and pancreatic progenitor cells were actually produced by inducing differentiation of iPS cells. In the method for manufacturing differentiated cells according to the present invention, as a preferred embodiment, iPS cells obtained by the manufacturing method of the present invention are used, but in this example, the method for manufacturing differentiated cells according to the present invention (step s5 above) was carried out using iPS cells provided by the Kyoto University iPS Cell Research Foundation (a research strain of human clinical iPS cells (15M66)).
本実施例では、iPS細胞を分化誘導する工程s5が、さらに3つの工程(s5-1、s5-2、s5-3)に分かれており、これらの各工程を、下記のように、3つの密閉容器(C1、C2、C3)に1対1で対応付け、容器から容器へと内容物を送りながら、各容器内で各工程を順次実施した。
工程s5-1:容器C1内において、iPS細胞を分化誘導し、胚体内胚葉を経て、原始腸管を得る工程。
工程s5-2:原始腸管を容器C2へと移動させ、該容器C2内において、前記の原始腸管を分化誘導し、後部前腸内胚葉を得る工程。
工程s5-3:後部前腸内胚葉を容器C3へと移動させ、該容器C3内において、前記の後部前腸内胚葉を分化誘導し、膵前駆細胞を得る工程。
In this example, step s5 of inducing differentiation of iPS cells was further divided into three steps (s5-1, s5-2, s5-3), and each of these steps was assigned one-to-one to three sealed containers (C1, C2, C3) as described below, and each step was carried out sequentially in each container while the contents were transferred from container to container.
Step s5-1: A step of inducing differentiation of iPS cells in the container C1 to obtain a primitive gut tube through definitive endoderm.
Step s5-2: A step of transferring the primitive gut to a container C2, and inducing differentiation of the primitive gut in the container C2 to obtain posterior foregut endoderm.
Step s5-3: a step of transferring the posterior foregut endoderm to a container C3, and inducing differentiation of the posterior foregut endoderm in the container C3 to obtain pancreatic progenitor cells.
本実施例では、前記3つの密閉容器として、上記実施例3および4で用いた容器と同様のGREX 10M-CSを用いた。
工程s5-1、s5-2、s5-3を実施するための細胞製造装置の概要は、図12に示すとおりであり、同図の構成におけるq=4の場合に該当する。また、密閉容器GREX 10M-CSの外観は、図29に示したとおりである。
In this example, the three sealed containers were made of GREX 10M-CS, which was the same as the containers used in Examples 3 and 4 above.
The outline of the cell manufacturing apparatus for carrying out steps s5-1, s5-2, and s5-3 is as shown in Fig. 12, and corresponds to the case where q = 4 in the configuration of the same figure. Also, the external appearance of the sealed container GREX 10M-CS is as shown in Fig. 29.
(工程s5-1(iPS細胞から胚体内胚葉への分化誘導))
先ず、容器C1に、iPS細胞株(15M66、1×106 cells)と、アテロコラーゲンビーズ溶液(KOKEN:MIC-00)と、液体培地StemFit AK03(味の素)10mL(該液体培地にはY-27632 (富士フィルムWako)10μM濃度が含まれている)を注入した。
以降の手法はTEMdiff Pancreatic Progenitor Kit(STEMCELL Technologies社)の推奨プロトコールに準じた(https://cdn.stemcell.com/media/files/pis/DX20464-PIS_1_4_0.pdf)。
次に、該容器C1に、STEMdiff Pancreatic Progenitor Kit(STEMCELL Technologies社)のMedium 1Aを10ml注入した。Day1より、Medium 1Bを0.5ml/hの流量にて継続的に供給し、Day5まで培養した。これにより、iPS細胞の分化誘導が行なわれて、胚体内胚葉が得られた。
(Step s5-1 (Induction of differentiation of iPS cells into definitive endoderm))
First, an iPS cell line (15M66, 1 × 10 6 cells), an atelocollagen bead solution (KOKEN: MIC-00), and 10 mL of liquid medium StemFit AK03 (Ajinomoto) (containing 10 μM concentration of Y-27632 (Fujifilm Wako)) were poured into container C1.
The procedure thereafter followed the recommended protocol for the TEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) (https://cdn.stemcell.com/media/files/pis/DX20464-PIS_1_4_0.pdf).
Next, 10 ml of Medium 1A from the STEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) was poured into the container C1. From
(容器C1の内容物を、容器C2に移動させる操作)
先ず、容器C1に空気を送り込み、容器C1内の上澄み部分を廃液用のシリンジに押し出した(容器内への空気の送り込みによって、上澄み部分が押し出されるように、他の入出用ポートは適宜に閉鎖した)。その結果、容器C1の底部には、沈殿していた胚体内胚葉とアテロコラーゲンビーズとそれらを含んだ少量の液体培地が残った。
次に入出用ポートの接続を切り替え、容器C1にシリンジにて空気を注入し、該底部に残っていた沈殿部分(胚体内胚葉とアテロコラーゲンビーズと少量の液体培地)を容器C2へと押し出して、該沈殿部分の移動を完了した。
(Operation of transferring the contents of container C1 to container C2)
First, air was pumped into the container C1, and the supernatant in the container C1 was pushed out into a syringe for waste liquid (other inlet/outlet ports were appropriately closed so that the supernatant was pushed out by pumping air into the container). As a result, the precipitated definitive endoderm and atelocollagen beads, as well as a small amount of liquid medium containing them, remained at the bottom of the container C1.
Next, the connection of the input/output ports was switched, air was injected into container C1 using a syringe, and the sediment remaining at the bottom (definitive endoderm, atelocollagen beads, and a small amount of liquid medium) was pushed out into container C2, completing the movement of the sediment.
(工程s5-2(胚体内胚葉から原始腸管への分化誘導))
先ず、前記の容器C2内に、STEMdiff Pancreatic Progenitor Kit(STEMCELL Technologies社)のMedium 2Aを10ml注入した。Day7より、Medium 2Bを0.5ml/hの流量にて継続的に供給し、Day9まで培養した。これにより、胚体内胚葉の分化誘導が行なわれて、原始腸管が得られた。
(Step s5-2 (Induction of Differentiation from Definitive Endoderm to Primitive Gut))
First, 10 ml of Medium 2A from the STEMdiff Pancreatic Progenitor Kit (STEMCELL Technologies) was poured into the container C2. From
(容器C2の内容物を、容器C3に移動させる操作)
上記の容器C1の内容物を容器C2に移動させる操作と同様に、容器C2に空気を送り込み、容器C2内の上澄み部分を廃液用のシリンジに押し出した。その結果、容器C2の底部には、沈殿していた原始腸管とアテロコラーゲンビーズとそれらを含んだ少量の液体培地が残った。
次に入出用ポートの接続を切り替え、容器C2にシリンジにて空気を注入し、該底部に残っていた沈殿部分(原始腸管とアテロコラーゲンビーズと少量の液体培地)を容器C3へと押し出して、該沈殿部分の移動を完了した。
(Operation of moving the contents of container C2 to container C3)
In the same manner as in the above operation of transferring the contents of container C1 to container C2, air was pumped into container C2, and the supernatant in container C2 was pushed out into a syringe for waste liquid. As a result, the precipitated primitive intestine and atelocollagen beads, as well as a small amount of liquid medium containing them, remained at the bottom of container C2.
Next, the connection of the input/output ports was switched, air was injected into container C2 using a syringe, and the sediment remaining at the bottom (primitive intestine, atelocollagen beads, and a small amount of liquid medium) was pushed out into container C3, completing the movement of the sediment.
(工程s5-3)原始腸管から後部前腸内胚葉への分化誘導
先ず、前記の容器C3内に、STEMdiff Pancreatic Progenitor Kit(STEMCELL Technologies社)のMedium 3を10ml注入した。Day10より、Medium 3を0.5ml/hの流量にて継続的に供給し、Day13まで培養した。これにより、原始腸管の分化誘導が行なわれて、後部前腸内胚葉が得られた。
(Step s5-3) Differentiation induction from primitive gut to posterior foregut endoderm First, 10 ml of
(容器C3の内容物を、容器C4に移動させる操作)
上記の容器C1の内容物を容器C2に移動させる操作と同様に、容器C3に空気を送り込み、容器C3内の上澄み部分を廃液用のシリンジに押し出した。その結果、容器C3の底部には、沈殿していた後部前腸内胚葉とアテロコラーゲンビーズとそれらを含んだ少量の液体培地が残った。
次に入出用ポートの接続を切り替え、容器C3にシリンジにて空気を注入し、該底部に残っていた沈殿部分(後部前腸内胚葉とアテロコラーゲンビーズと少量の液体培地)を容器C4へと押し出して、該沈殿部分の移動を完了した。
(Operation of moving the contents of container C3 to container C4)
In the same manner as in the above operation of transferring the contents of container C1 to container C2, air was pumped into container C3, and the supernatant in container C3 was pushed out into a syringe for waste liquid. As a result, the precipitated posterior foregut endoderm and atelocollagen beads, as well as a small amount of liquid medium containing them, remained at the bottom of container C3.
Next, the connection of the input/output ports was switched, air was injected into container C3 using a syringe, and the sediment remaining at the bottom (posterior foregut endoderm, atelocollagen beads, and a small amount of liquid medium) was pushed out into container C4, completing the movement of the sediment.
(工程s5-4)後部前腸内胚葉から膵前駆細胞への分化誘導
先ず、前記の容器C3内に、STEMdiff Pancreatic Progenitor Kit(STEMCELL Technologies社)のMedium 4を10ml注入した。Day10より、Medium 4を0.5ml/hの流量にて継続的に供給し、Day19まで培養した。これにより、後部前腸内胚葉の分化誘導が行なわれて、膵前駆細胞が得られた。
(Step s5-4) Differentiation induction from posterior foregut endoderm to pancreatic progenitor cells First, 10 ml of
(膵前駆細胞マーカー陽性細胞)
Day19において容器内容物(工程s5-4)をサンプリングし、膵前駆細胞のマーカータンパク質であるPDX-1 (膵前駆細胞マーカー)、NKX6.1 (膵前駆細胞マーカー)について、定法に従って免疫染色を行った。((抗PDX-1抗体:(Human/Mouse PDX-1/IPF1 Alexa Fluor 647 MAb (Clone 267712) (R&D Systems, Inc. IC2419R-100UG))使用)と、Hoechst染色(核染色)との二重染色結果を図39(膵前駆細胞PDX-1)に示す。抗NKX6.1抗体:(BD PharmingenAlexa Fluor 647 Mouse Anti-NKX6.1 (BD 563338))使用)と、Hoechst染色(核染色)との二重染色結果を図40(膵前駆細胞NKX6.1)に示す。
図39(膵前駆細胞PDX-1)、および図40(膵前駆細胞NKX6.1)に示されるように、アテロコラーゲン上の細胞集塊中に、Hoechstシグナルと共在するPDX-1、NKX6.1シグナルを有する細胞(すなわち、膵前駆細胞)が多数観察された。よって、本発明の製造方法により、iPS細胞から膵前駆細胞が効率よく分化誘導されたことが明らかになった。
(Pancreatic progenitor cell marker positive cells)
On Day 19, the contents of the container (step s5-4) were sampled, and immunostaining was performed for pancreatic progenitor cell marker proteins PDX-1 (pancreatic progenitor cell marker) and NKX6.1 (pancreatic progenitor cell marker) according to a standard method. The results of double staining with anti-PDX-1 antibody (Human/Mouse PDX-1/IPF1 Alexa Fluor 647 MAb (Clone 267712) (R&D Systems, Inc. IC2419R-100UG)) and Hoechst staining (nuclear staining) are shown in Figure 39 (Pancreatic progenitor cells PDX-1). The results of double staining with anti-NKX6.1 antibody (BD PharmingenAlexa Fluor 647 Mouse Anti-NKX6.1 (BD 563338)) and Hoechst staining (nuclear staining) are shown in Figure 40 (Pancreatic progenitor cells NKX6.1).
As shown in Figure 39 (pancreatic progenitor cells PDX-1) and Figure 40 (pancreatic progenitor cells NKX6.1), many cells (i.e., pancreatic progenitor cells) having PDX-1 and NKX6.1 signals coexisting with Hoechst signals were observed in the cell clusters on atelocollagen. Therefore, it was revealed that the method of the present invention efficiently induced differentiation of pancreatic progenitor cells from iPS cells.
(膵前駆細胞マーカーの発現誘導)
Day19において容器内容物(工程s5-4)をサンプリングし、コラゲナーゼ処理を行ってアテロコラーゲンビーズを溶解した。得られた細胞ペレットからSuperPrep II Cell Lysis & RT Kit for qPCR(東洋紡株式会社: SCQ-401)を用いてmRNAを抽出し、cDNAを合成した。StepOnePlus system (Life Technologies, Carlsbad, CA, USA)、および、TaqManTM Fast Advanced Master Mix (Life Technologies, Carlsbad, CA, USA) を用いてリアルタイムPCR解析を行い、SOX17(内胚葉マーカー)、HNF1bとFoxA2(原始腸管マーカー)、SOX9、PDX-1とNKX6.1(膵前駆細胞マーカー)の発現量を解析した。コントロールには、分化誘導を行わずにアテロコラーゲン上で培養したiPS細胞株(15M66)を用いた。結果を図41(膵前駆細胞の遺伝子発現)に示す。
(Induction of pancreatic progenitor cell marker expression)
On day 19, the contents of the container (step s5-4) were sampled and treated with collagenase to dissolve the atelocollagen beads. From the obtained cell pellet, mRNA was extracted using SuperPrep II Cell Lysis & RT Kit for qPCR (Toyobo Co., Ltd.: SCQ-401), and cDNA was synthesized. Real-time PCR analysis was performed using the StepOnePlus system (Life Technologies, Carlsbad, CA, USA) and TaqMan TM Fast Advanced Master Mix (Life Technologies, Carlsbad, CA, USA), and the expression levels of SOX17 (endodermal marker), HNF1b and FoxA2 (primitive gut markers), SOX9, PDX-1 and NKX6.1 (pancreatic progenitor cell markers) were analyzed. As a control, an iPS cell line (15M66) cultured on atelocollagen without differentiation induction was used. The results are shown in Figure 41 (gene expression of pancreatic progenitor cells).
図41(膵前駆細胞の遺伝子発現)に示されるように、分化誘導後の細胞では内胚葉マーカー(SOX17)の発現はコントロールと比べて約1.5倍であり有意に増加した。分化誘導後の細胞では膵前駆細胞マーカー(SOX9、PDX-1とNKX6.1)の発現はコントロールと比べて約1.3~1.4倍であり有意に増加した。原始腸管マーカー(FoxA2)の発現はコントロールと比べて約1.4倍であり有意に増加した。よって、本法により、iPSから原始腸管を経て膵前駆細胞への分化が順調に誘導されたことが示唆された。これらの結果より、本発明の製造方法および製造装置を用いて、細胞の分化誘導が行えることが示された。 As shown in Figure 41 (gene expression in pancreatic progenitor cells), in cells after differentiation induction, the expression of endoderm marker (SOX17) was approximately 1.5 times higher than in the control, showing a significant increase. In cells after differentiation induction, the expression of pancreatic progenitor cell markers (SOX9, PDX-1, and NKX6.1) was approximately 1.3 to 1.4 times higher than in the control, showing a significant increase. The expression of primitive intestinal tract marker (FoxA2) was approximately 1.4 times higher than in the control, showing a significant increase. This suggests that this method smoothly induced differentiation from iPS cells via the primitive intestine to pancreatic progenitor cells. These results demonstrate that cell differentiation can be induced using the production method and production device of the present invention.
本発明のiPS細胞の製造方法、分化細胞の製造方法、および、細胞製造装置によって、iPS細胞や分化細胞を従来よりも安価で簡単に製造することが可能になり、さらに自働化製造も可能になる。特に、臨床的な観点から、本発明は、移植療法を必要とする患者の体細胞からiPS細胞を樹立し、該iPS細胞から種々の分化細胞を製造し、前記患者に移植(自家移植)するといった用途に好ましく利用可能である。 The method for producing iPS cells, the method for producing differentiated cells, and the cell production device of the present invention make it possible to produce iPS cells and differentiated cells more cheaply and easily than before, and also enable automated production. In particular, from a clinical perspective, the present invention is preferably applicable to applications such as establishing iPS cells from the somatic cells of a patient requiring transplantation therapy, producing various differentiated cells from the iPS cells, and transplanting them into the patient (autotransplantation).
本出願は、国際出願(PCT/JP)2023/006753(出願日:2023年2月24日)を基礎としており、その内容は本明細書に全て包含されるものである。 This application is based on International Application (PCT/JP) 2023/006753 (filing date: February 24, 2023), the contents of which are incorporated in their entirety into this specification.
A1~An(n≧3) 密閉容器
J1~J(n-1) 接続管路
F1~F(n-1) 送り機構
G1~Gn 材料供給源
A1 to An (n≧3) Sealed container J1 to J(n-1) Connecting pipe F1 to F(n-1) Feeding mechanism G1 to Gn Material supply source
Claims (17)
接続管路を介して直列に接続されたn(n≧3)個の密閉容器間を、第1番目の密閉容器(A1)から第n番目の密閉容器(An)まで、それぞれ送り機構によって、細胞を一方向に順次移動させ、各密閉容器内において人工多能性幹細胞の製造工程を順次実施することを有し、
前記密閉容器(A1~An)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、各密閉容器からその次の密閉容器へと内容物を移動させる機構であり、
前記人工多能性幹細胞の製造工程が、
第1番目の密閉容器(A1)内において、液体培地中で体細胞に初期化因子を接触させる工程(s1)と、
第2番目の密閉容器(A2)から第(n-1)番目の密閉容器(A(n-1))内において、液体培地中の初期化因子の濃度を低減させる工程(s2)と、
第n番目の密閉容器(An)内において、液体培地中で前記体細胞を培養して、人工多能性幹細胞を樹立する工程(s3)とを有する、
前記人工多能性幹細胞の製造方法。 A method for producing induced pluripotent stem cells, comprising:
The method comprises: sequentially moving cells in one direction from a first sealed container (A1) to an nth sealed container (An) by a feed mechanism among n (n≧3) sealed containers connected in series via a connecting pipeline; and sequentially carrying out a production process of induced pluripotent stem cells in each sealed container;
Each of the sealed containers (A1 to An) has one or more openable/closable inlet/outlet ports,
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
the feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to a communicating state,
The process for producing the induced pluripotent stem cells comprises:
A step (s1) of contacting a reprogramming factor with a somatic cell in a liquid medium in a first sealed container (A1);
A step (s2) of reducing the concentration of the reprogramming factor in the liquid medium in the second sealed container (A2) to the (n-1)th sealed container (A(n-1));
and (s3) culturing the somatic cells in a liquid medium in the n-th closed container (An) to establish induced pluripotent stem cells.
A method for producing the induced pluripotent stem cells.
上記第n番目の密閉容器(An)の後には、前記拡大培養の回数に対応するp個の密閉容器(B1~Bp)が、接続管路を介して直列に接続され、
密閉容器(An)から密閉容器(Bp)まで、それぞれ送り機構によって、人工多能性幹細胞が一方向に順次移動し、密閉容器(B1~Bp)のそれぞれの内部で1回の拡大培養が実施され、それにより、合計p回の拡大培養が実施され、
密閉容器(B1~Bp)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、各密閉容器からその次の密閉容器へと内容物を移動させる機構である、
請求項1記載の人工多能性幹細胞の製造方法。 After the step (s3), the method further includes a step (s4) of expanding the induced pluripotent stem cells p times (p≧1),
p sealed containers (B1 to Bp) corresponding to the number of times of expansion culture are connected in series to the n-th sealed container (An) via a connecting pipeline;
The artificial pluripotent stem cells are sequentially moved in one direction from the sealed container (An) to the sealed container (Bp) by the respective feeding mechanisms, and one expansion culture is carried out in each of the sealed containers (B1 to Bp), thereby carrying out a total of p expansion cultures;
Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports,
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from each sealed container to the next sealed container through the connecting pipe line switched to the communicating state.
A method for producing the induced pluripotent stem cell according to claim 1.
可撓性材料で構成された容器本体を有する密閉容器、
硬質材料で構成された容器本体を有する密閉容器、および、
可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器
からなる群から選択される密閉容器である、
請求項1または2に記載の人工多能性幹細胞の製造方法。 The sealed container is
A sealed container having a container body made of a flexible material;
A sealed container having a container body made of a hard material;
A sealed container having a container body made of a composite material of a flexible material and a hard material.
A method for producing an artificial pluripotent stem cell according to claim 1 or 2.
移動元の密閉容器内に流体を追加することによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
移動元の密閉容器の容積を減少させることによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
上記接続管路に設けたポンプ装置によって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
移動元の密閉容器に対して、その次の密閉容器から吸引力を作用させることによって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
重力を利用して、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、および、
磁性が付与されたマイクロキャリアに細胞を接着させ、該マイクロキャリアに外部から磁力を作用させて、移動元の密閉容器内のマイクロキャリアとそれに接着した該細胞を、その次の密閉容器へと移動させる機構
からなる群から選ばれる機構、または、該群から選ばれる2以上の機構を組み合わせた機構である、
請求項1~3のいずれか1項に記載の人工多能性幹細胞の製造方法。 The feed mechanism is
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container;
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container;
a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline;
a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container;
A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and
a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
A method for producing an artificial pluripotent stem cell according to any one of claims 1 to 3.
請求項1~4のいずれか1項に記載の製造方法における人工多能性幹細胞を製造する工程と、
製造された人工多能性幹細胞を分化誘導する工程(s5)とを有し、
前記人工多能性幹細胞の製造方法で用いられた密閉容器のうち最後尾の密閉容器(X1)の後には、前記工程(s5)を実施するための密閉容器(C1)が、接続管路を介してさらに接続され、
密閉容器(C1)は、1以上の開閉可能な入出用ポートを有し、該入出用ポートを通じて、工程(s5)の分化誘導に必要な材料が密閉容器(C1)の内部に供給され、
送り機構によって、密閉容器(X1)から密閉容器(C1)へと、人工多能性幹細胞が移動し、密閉容器(C1)において前記工程(s5)が実施され、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(X1)から密閉容器(C1)へと内容物を移動させる機構である、
前記分化細胞の製造方法。 A method for producing differentiated cells, comprising:
A step of producing induced pluripotent stem cells by the production method according to any one of claims 1 to 4;
and a step (s5) of inducing differentiation of the produced induced pluripotent stem cells,
a sealed container (C1) for carrying out the step (s5) is further connected to the rear end of the sealed container (X1) among the sealed containers used in the method for producing induced pluripotent stem cells via a connecting pipeline;
The sealed container (C1) has one or more openable/closable inlet/outlet ports, and a material necessary for differentiation induction in step (s5) is supplied to the inside of the sealed container (C1) through the inlet/outlet ports;
The artificial pluripotent stem cells are transferred from the sealed container (X1) to the sealed container (C1) by a transfer mechanism, and the step (s5) is carried out in the sealed container (C1);
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (X1) to the sealed container (C1) through the connecting pipe line switched to a communicating state.
The method for producing the differentiated cells.
上記分化誘導する工程で用いられた密閉容器のうち最後尾の密閉容器(X2)の後には、前記工程(s6)を実施するための密閉容器(D1)が、接続管路を介してさらに接続され、
密閉容器(D1)は、1以上の開閉可能な入出用ポートを有し、
送り機構によって、密閉容器(X2)から密閉容器(D1)へと、分化細胞が移動し、密閉容器(D1)において前記工程(s6)が実施され、
前記接続管路は、連通状態と非連通状態とに切り替え可能に構成され、
前記送り機構は、連通状態に切り替えられた接続管路を通じて、密閉容器(X2)から密閉容器(D1)へと内容物を移動させる機構である、
請求項5に記載の分化細胞の製造方法。 The method further comprises a step (s6) of removing undifferentiated cells after the differentiation induction step,
a sealed container (D1) for carrying out the step (s6) is further connected to the rear end of the sealed container (X2) among the sealed containers used in the differentiation induction step via a connecting pipeline;
The sealed container (D1) has one or more openable/closable inlet/outlet ports,
The differentiated cells are transferred from the sealed container (X2) to the sealed container (D1) by a transfer mechanism, and the step (s6) is carried out in the sealed container (D1);
The connecting pipe is configured to be switchable between a communicating state and a non-communicating state,
The feed mechanism is a mechanism for moving the contents from the sealed container (X2) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
The method for producing differentiated cells according to claim 5 .
液体培地中で体細胞に初期化因子を接触させる工程(s1)を実施するための密閉容器(A1)と、
前記液体培地中の初期化因子の濃度を低減させる工程(s2)を実施するための密閉容器(A2)と、
前記液体培地中で前記体細胞を培養して、人工多能性幹細胞を樹立する工程(s3)を実施するための密閉容器(A3)とを有し、
密閉容器(A1)~(A3)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
密閉容器(A1)~(A3)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程の順に直列に接続されているか、または前記工程の順に直列に接続可能な状態となっており、かつ、
当該細胞製造装置は、
連通状態に切り替えられた前記接続管路を通じて、密閉容器(A1)の内容物を密閉容器(A2)へと移動させる送り機構を有し、かつ、
連通状態に切り替えられた前記接続管路を通じて、密閉容器(A2)の内容物を密閉容器(A3)へと移動させる送り機構を有する、
前記細胞製造装置。 A cell manufacturing device comprising:
A sealed container (A1) for carrying out a step (s1) of contacting a somatic cell with a reprogramming factor in a liquid medium;
A sealed container (A2) for carrying out a step (s2) of reducing the concentration of the reprogramming factor in the liquid medium;
and a sealed container (A3) for carrying out a step (s3) of culturing the somatic cells in the liquid medium to establish induced pluripotent stem cells;
Each of the sealed containers (A1) to (A3) has one or more openable/closable inlet/outlet ports,
The sealed containers (A1) to (A3) are connected in series in the order of the steps via a connecting pipe line that can be switched between a communicating state and a non-communicating state, or are capable of being connected in series in the order of the steps, and
The cell manufacturing device comprises:
a feed mechanism for moving the content of the sealed container (A1) to the sealed container (A2) through the connecting pipe line switched to a communicating state,
a feed mechanism for moving the content of the sealed container (A2) to the sealed container (A3) through the connecting pipe line switched to a communicating state;
The cell manufacturing device.
密閉容器(B1~Bp)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
(i)前記拡大培養の回数であるp回が1回である場合には、
前記p個の密閉容器は、1個の密閉容器(B1)であって、
密閉容器(B1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、上記密閉容器(A3)に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、上記密閉容器(A3)の内容物を密閉容器(B1)へと移動させる送り機構を有し、
(ii)前記拡大培養の回数であるp回が2回以上である場合には、
前記p個の密閉容器は、2個以上の密閉容器(B1~Bp)であって、
密閉容器(B1)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、上記密閉容器(A3)に接続されているか、または接続可能な状態となっており、
密閉容器(B1)~(Bp)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程(s4)の順に直列に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、上記密閉容器(A3)の内容物を密閉容器(B1)~(Bp)へと順に移動させる送り機構を有する、
請求項7に記載の細胞製造装置。 The method further includes p sealed containers (B1 to Bp) for carrying out a step (s4) of expanding the induced pluripotent stem cells in a liquid medium p times (p≧1),
Each of the sealed containers (B1 to Bp) has one or more openable/closable inlet/outlet ports,
(i) When the number of times of expansion culture, p, is 1,
The p number of sealed containers is one sealed container (B1),
the sealed container (B1) is connected to or can be connected to the sealed container (A3) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the cell manufacturing apparatus has a transfer mechanism that transfers the content of the sealed container (A3) to the sealed container (B1) through the connecting pipeline that has been switched to a communicating state;
(ii) When the number of times of the expansion culture, p, is 2 or more,
The p number of sealed containers is two or more sealed containers (B1 to Bp),
the sealed container (B1) is connected to or can be connected to the sealed container (A3) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the sealed containers (B1) to (Bp) are connected in series or are connectable in the order of the step (s4) via connecting pipes that can be switched between a communicating state and a non-communicating state;
The cell manufacturing apparatus has a feed mechanism that moves the contents of the sealed container (A3) to the sealed containers (B1) to (Bp) in order through the connecting pipeline that is switched to a communicating state.
The cell manufacturing device according to claim 7.
密閉容器(C1)~(Cq)のそれぞれは、1以上の開閉可能な入出用ポートを有し、
(i)前記q個の密閉容器が1個の密閉容器(C1)の場合には、
密閉容器(C1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、上記密閉容器(A3)に、もしくは密閉容器(B1)~(Bp)のうちの最後尾の密閉容器(Bp)に、接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、前記密閉容器(A3)または密閉容器(Bp)の内容物を密閉容器(C1)へと移動させる送り機構を有し、
(ii)前記q個の密閉容器が2個以上の密閉容器(C1)~(Cq)である場合には、
密閉容器(C1)~(Cq)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、前記工程(s5)の順に直列に接続されているか、または接続可能な状態となっており、かつ、密閉容器(C1)は、連通状態と非連通状態とに切り替え可能な接続管路を介して、上記密閉容器(A3)に、もしくは密閉容器(B1)~(Bp)のうちの最後尾の密閉容器(Bp)に、接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、連通状態に切り替えられた前記接続管路を通じて、前記密閉容器(A3)または最後尾の密閉容器(Bp)の内容物を、密閉容器(C1)~(Cq)へと順に移動させる送り機構を有する、
請求項7または8に記載の細胞製造装置。 The method further includes q (q≧1) sealed containers (C1) to (Cq) for carrying out the step (s5) of inducing differentiation of the induced pluripotent stem cells,
Each of the sealed containers (C1) to (Cq) has one or more openable/closable inlet/outlet ports,
(i) When the q number of sealed containers is one sealed container (C1),
the sealed container (C1) is connected or connectable to the sealed container (A3) or to the last sealed container (Bp) among the sealed containers (B1) to (Bp) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
the cell manufacturing apparatus has a transfer mechanism that transfers the contents of the sealed container (A3) or the sealed container (Bp) to the sealed container (C1) through the connecting pipeline that has been switched to a communicating state;
(ii) When the q number of sealed containers is two or more sealed containers (C1) to (Cq),
the sealed containers (C1) to (Cq) are connected in series or are connectable in the order of the step (s5) via a connecting pipe that can be switched between a communicating state and a non-communicating state, and the sealed container (C1) is connected or is connectable to the sealed container (A3) or to the last sealed container (Bp) among the sealed containers (B1) to (Bp) via a connecting pipe that can be switched between a communicating state and a non-communicating state,
The cell manufacturing apparatus has a feed mechanism that moves the contents of the sealed container (A3) or the last sealed container (Bp) to the sealed containers (C1) to (Cq) in sequence through the connecting pipeline that has been switched to a communicating state.
The cell manufacturing apparatus according to claim 7 or 8.
密閉容器(D1)は、1以上の開閉可能な入出用ポートを有し、
密閉容器(D1)は、連通状態と非連通状態に切り替え可能な接続管路を介して、前記最後尾の密閉容器(Cq)に接続されているか、または接続可能な状態となっており、
当該細胞製造装置は、
連通状態に切り替えられた前記接続管路を通じて、上記最後尾の密閉容器(Cq)の内容物を密閉容器(D1)へと移動させる送り機構を有する、
請求項9に記載の細胞製造装置。 The method further includes a sealed container (D1) for carrying out a step (s6) of removing undifferentiated cells from the content of the last sealed container (Cq) among the sealed containers (C1) to (Cq),
The sealed container (D1) has one or more openable/closable inlet/outlet ports,
the sealed container (D1) is connected to or can be connected to the rearmost sealed container (Cq) via a connecting pipe that can be switched between a communicating state and a non-communicating state;
The cell manufacturing device comprises:
A feed mechanism is provided for moving the contents of the rearmost sealed container (Cq) to the sealed container (D1) through the connecting pipe line switched to the communicating state.
The cell manufacturing device according to claim 9.
可撓性材料で構成された容器本体を有する密閉容器、
硬質材料で構成された容器本体を有する密閉容器、および、
可撓性材料と硬質材料との複合材料で構成された容器本体を有する密閉容器
からなる群から選択される密閉容器である、
請求項7~10のいずれか1項に記載の細胞製造装置。 The sealed container is
A sealed container having a container body made of a flexible material;
A sealed container having a container body made of a hard material;
A sealed container having a container body made of a composite material of a flexible material and a hard material.
The cell manufacturing device according to any one of claims 7 to 10.
移動元の密閉容器内に流体を追加することによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
移動元の密閉容器の容積を減少させることによって、移動元の密閉容器の内容物をその次の密閉容器へと押し出して移動させる機構、
上記接続管路に設けたポンプ装置によって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
移動元の密閉容器に対して、その次の密閉容器から吸引力を作用させることによって、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、
重力を利用して、移動元の密閉容器の内容物をその次の密閉容器へと移動させる機構、および、
磁性が付与されたマイクロキャリアに細胞を接着させ、該マイクロキャリアに外部から磁力を作用させて、移動元の密閉容器内のマイクロキャリアとそれに接着した該細胞を、その次の密閉容器へと移動させる機構
からなる群から選ばれる機構、または、該群から選ばれる2以上の機構を組み合わせた機構である、
請求項7~11のいずれか1項に記載の細胞製造装置。 The feed mechanism is
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by adding fluid to the source sealed container;
A mechanism for pushing and transferring the contents of the source sealed container to the next sealed container by reducing the volume of the source sealed container;
a mechanism for transferring the contents of the source sealed container to the next sealed container by a pump device provided in the connecting pipeline;
a mechanism for transferring the contents of the source sealed container to the next sealed container by applying suction force from the next sealed container to the source sealed container;
A mechanism for transferring the contents of the source sealed container to the next sealed container by utilizing gravity; and
a mechanism selected from the group consisting of mechanisms for adhering cells to a magnetic microcarrier and applying an external magnetic force to the microcarrier to move the microcarrier and the cells attached thereto in a source sealed container to a next sealed container, or a mechanism combining two or more mechanisms selected from the group.
The cell manufacturing device according to any one of claims 7 to 11.
該基板上には上記密閉容器が配置され、各密閉容器は該基板に固定されており、
各密閉容器は、連通状態と非連通状態に切り替え可能な上記接続管路を介して、上記工程の順に接続されているか、または接続可能な状態となっている、
請求項7~12のいずれか1項に記載の細胞製造装置。 Further comprising a substrate for arranging all of the sealed containers;
The sealed containers are disposed on the substrate, and each sealed container is fixed to the substrate;
The sealed containers are connected or connectable in the order of the steps via the connecting pipes that can be switched between a communicating state and a non-communicating state.
The cell manufacturing device according to any one of claims 7 to 12.
(i)前記折り曲げ中心線によって分けられた基板面の2つの領域(e1)、(e2)のうちの一方の領域(e1)には、上記密閉容器のうちの所定数の密閉容器が、該折り曲げ中心線に沿って、一つの向き(d1)に、順に並ぶように配置され、
(ii)前記折り曲げ中心線によって分けられた基板面の2つの領域(e1)、(e2)のうちの他方の領域(e2)には、上記密閉容器のうちの残りの密閉容器が、該折り曲げ中心線に沿って、前記向き(d1)とは逆の向き(d2)に、順に並ぶように配置され、
(iii)前記一方の領域(e1)の密閉容器のうちの最後尾の密閉容器と、前記他方の領域(e2)の密閉容器のうちの先頭の密閉容器とが、連通状態と非連通状態に切り替え可能な上記接続管路によって接続されているか、または接続可能な状態となっている、
請求項13に記載の細胞製造装置。 The substrate can be folded in two around a folding center line,
(i) in one region (e1) of two regions (e1) and (e2) on the substrate surface separated by the folding center line, a predetermined number of the sealed containers are arranged in order in one direction (d1) along the folding center line;
(ii) in the other region (e2) of the two regions (e1) and (e2) of the substrate surface separated by the folding center line, the remaining sealed containers among the sealed containers are arranged in order along the folding center line in a direction (d2) opposite to the direction (d1);
(iii) the rearmost sealed container among the sealed containers in the one region (e1) and the frontmost sealed container among the sealed containers in the other region (e2) are connected or in a connectable state by the connecting pipe line that can be switched between a communicating state and a non-communicating state;
The cell manufacturing device according to claim 13.
上記の全ての密閉容器となる領域と、1以上の入出用ポートとなる領域と、接続管路となる領域とが、前記2枚の可撓性シートの間の所定の位置に形成されるように、これらの領域を非接合領域として残して、前記2枚の可撓性シートは互いに接合されており、
該2枚の可撓性シートの外面には、該接続管路を開閉するための押圧用アクチュエーターが設けられており、
該押圧用アクチュエーターは、前記接続管路となる領域を該可撓性シートの外側から押圧して非連通状態とする押圧ポジションと、前記接続管路となる領域を押圧せず連通状態とする非押圧ポジションとを取るように作動し、該押圧用アクチュエーターの作動により、前記接続管路となる領域は、連通状態と非連通状態とに切り替え可能な接続管路として機能し、
1以上の入出用ポートとなる領域は、各密閉容器となる領域から前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、該開口端部には、開閉可能な入出用ポートのための構成が付与されている、
請求項7~14いずれか1項に記載の細胞製造装置。 The device further comprises two overlapping flexible sheets,
the two flexible sheets are bonded to each other while leaving the regions that become all of the above-mentioned sealed containers, the region that becomes one or more inlet/outlet ports, and the region that becomes the connecting pipeline as non-bonded regions so that these regions are formed at predetermined positions between the two flexible sheets;
a pressing actuator for opening and closing the connection pipe line is provided on the outer surfaces of the two flexible sheets;
the pressing actuator operates to take a pressing position in which the region that will become the connecting pipeline is pressed from the outside of the flexible sheet to bring it into a non-communicating state, and a non-pressing position in which the region that will become the connecting pipeline is not pressed to bring it into a communicating state, and by operation of the pressing actuator, the region that will become the connecting pipeline functions as a connecting pipeline that can be switched between a communicating state and a non-communicating state;
The region that will become one or more inlet/outlet ports extends from the region that will become each sealed container to the outer periphery of the two flexible sheets to form an open end, and the open end is provided with a structure for an openable/closable inlet/outlet port.
A cell manufacturing device according to any one of claims 7 to 14.
(i)前記折り曲げ中心線によって分けられた2つの領域(e3)、(e4)のうちの一方の領域(e3)には、
上記密閉容器のうちの所定数の密閉容器となる領域が、該折り曲げ中心線に沿って、一つの向き(d3)に、順に並ぶように形成され、
前記所定数の密閉容器となる領域の各外周のうちの該折り曲げ中心線から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域が、該折り曲げ中心線から離れる方向に延び、かつ、前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、
前記所定数の密閉容器となる領域同士の間には、該密閉容器となる領域同士を接続する上記接続管路となる領域が形成されており、
(ii)前記折り曲げ中心線によって分けられた2つの領域(e3)、(e4)のうちの他方の領域(e4)には、
上記密閉容器のうちの残りの密閉容器となる領域が、該折り曲げ中心線に沿って、前記一つの向き(d3)とは逆の向き(d4)に、順に並ぶように形成され、
前記残りの密閉容器となる領域の各外周のうちの該折り曲げ中心線から遠い側に位置する外周部分からは、上記1以上の入出用ポートとなる領域が、該折り曲げ中心線から離れる方向に延び、かつ、前記2枚の可撓性シートの外周縁まで延びて開口端部となっており、
前記残りの密閉容器となる領域同士の間には、該密閉容器となる領域同士を接続する接続管路となる領域が形成されており、
(iii)前記一方の領域(e3)の密閉容器のうちの最後尾の密閉容器と、前記他方の領域(e4)の密閉容器のうちの先頭の密閉容器とが、前記折り曲げ中心線を横切る接続管路となる領域によって接続されている、
請求項15に記載の細胞製造装置。 the outer peripheral shape of the two flexible sheets that are overlapped and joined to each other is a shape that can be folded in two around a folding center line;
(i) In one of the two regions (e3) and (e4) separated by the folding center line,
A predetermined number of regions that become sealed containers among the sealed containers are formed so as to be arranged in sequence in one direction (d3) along the folding center line,
from an outer periphery of each of the regions that will become the predetermined number of sealed containers that is located farther from the folding center line, a region that will become the one or more inlet/outlet ports extends in a direction away from the folding center line and extends to an outer periphery of the two flexible sheets to form an open end,
Between the regions that will become the predetermined number of sealed containers, a region that will become the connecting pipeline that connects the regions that will become the sealed containers is formed,
(ii) In the other region (e4) of the two regions (e3) and (e4) separated by the folding center line,
The remaining sealed containers are arranged in order along the folding center line in a direction (d4) opposite to the one direction (d3),
a region that becomes the one or more inlet/outlet ports extends from an outer periphery of each of the outer peripheries of the remaining region that becomes the sealed container, the outer periphery being located farther from the folding center line, in a direction away from the folding center line, and extends to an outer periphery of the two flexible sheets to form an open end;
Between the remaining regions that will become the sealed containers, a region that will become a connecting pipe that connects the regions that will become the sealed containers is formed,
(iii) the rearmost sealed container among the sealed containers in the one region (e3) and the frontmost sealed container among the sealed containers in the other region (e4) are connected by a region that serves as a connecting pipeline that crosses the folding center line;
The cell manufacturing device according to claim 15.
上記密閉容器(A1)内において、液体培地中で体細胞に初期化因子を接触させる工程(s1)と
前記工程(s1)の完了後に、連通状態に切り替えられた前記接続管路を通じて、該密閉容器(A1)の内容物を上記密閉容器(A2)内に移動させ、該密閉容器(A2)内において、前記液体培地中の初期化因子の濃度を低減させる工程(s2)と、
前記工程(s2)の完了後に、連通状態に切り替えられた前記接続管路を通じて、該密閉容器(A2)の内容物を上記密閉容器(A3)内に移動させ、該密閉容器(A3)内において、前記液体培地中で人工多能性幹細胞を樹立する工程(s3)と
を少なくとも有する、前記細胞製造方法。 A cell manufacturing method using the cell manufacturing device according to any one of claims 7 to 16,
A step (s1) of contacting a reprogramming factor with a somatic cell in a liquid medium in the sealed container (A1); and a step (s2) of transferring the content of the sealed container (A1) into the sealed container (A2) through the connecting pipe switched to a communicating state after the completion of the step (s1), and reducing the concentration of the reprogramming factor in the liquid medium in the sealed container (A2).
The cell production method comprises at least a step (s3) of transferring the contents of the sealed container (A2) into the sealed container (A3) through the connecting pipeline that has been switched to a communicating state after completion of the step (s2), and establishing artificial pluripotent stem cells in the liquid medium within the sealed container (A3).
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2025502714A JPWO2024177018A1 (en) | 2023-02-24 | 2024-02-19 |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| PCT/JP2023/006753 WO2024176440A1 (en) | 2023-02-24 | 2023-02-24 | Method for producing induced pluripotent stem cells |
| JPPCT/JP2023/006753 | 2023-02-24 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024177018A1 true WO2024177018A1 (en) | 2024-08-29 |
Family
ID=92500471
Family Applications (2)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2023/006753 Ceased WO2024176440A1 (en) | 2023-02-24 | 2023-02-24 | Method for producing induced pluripotent stem cells |
| PCT/JP2024/005834 Ceased WO2024177018A1 (en) | 2023-02-24 | 2024-02-19 | Method for producing induced pluripotent stem cells |
Family Applications Before (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2023/006753 Ceased WO2024176440A1 (en) | 2023-02-24 | 2023-02-24 | Method for producing induced pluripotent stem cells |
Country Status (2)
| Country | Link |
|---|---|
| JP (1) | JPWO2024177018A1 (en) |
| WO (2) | WO2024176440A1 (en) |
Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2018061519A (en) * | 2012-02-01 | 2018-04-19 | 東洋製罐グループホールディングス株式会社 | Cell culture kit |
| JP2018126195A (en) * | 2017-02-06 | 2018-08-16 | 大日本印刷株式会社 | Liquid feeding method, liquid feeding device, and content filled container |
| JP2018526992A (en) * | 2015-08-31 | 2018-09-20 | アイ ピース,インコーポレイテッド | Pluripotent stem cell production system, stem cell induction method, stem cell suspension culture method, stem cell suspension incubator, method for producing induced pluripotent stem cells, and method for producing specific somatic cells from animal cells |
| JP2020130052A (en) * | 2019-02-20 | 2020-08-31 | 東ソー株式会社 | Cell culture apparatus |
| JP2022529740A (en) * | 2019-04-24 | 2022-06-23 | ダブリュ.エル.ゴア アンド アソシエイツ,インコーポレイティド | Highly durable permeable fluoropolymer cell culture bag |
Family Cites Families (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| SG10201912931RA (en) * | 2015-08-31 | 2020-02-27 | I Peace Inc | Pluripotent stem cell manufacturing system, method for inducing stem cells, floating culture method for stem cells, floating culture vessel for stem cells, method for producing induced pluripotent stem cells, and method for producing particular somatic cells from animal cells |
| CA3054119A1 (en) * | 2017-02-24 | 2018-08-30 | I Peace, Inc. | Cell treatment device, suspension culture vessel, and stem cell induction method |
| EP4108757A4 (en) * | 2020-02-21 | 2023-11-22 | I Peace, Inc. | Solution conveyance apparatus |
-
2023
- 2023-02-24 WO PCT/JP2023/006753 patent/WO2024176440A1/en not_active Ceased
-
2024
- 2024-02-19 JP JP2025502714A patent/JPWO2024177018A1/ja active Pending
- 2024-02-19 WO PCT/JP2024/005834 patent/WO2024177018A1/en not_active Ceased
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JP2018061519A (en) * | 2012-02-01 | 2018-04-19 | 東洋製罐グループホールディングス株式会社 | Cell culture kit |
| JP2018526992A (en) * | 2015-08-31 | 2018-09-20 | アイ ピース,インコーポレイテッド | Pluripotent stem cell production system, stem cell induction method, stem cell suspension culture method, stem cell suspension incubator, method for producing induced pluripotent stem cells, and method for producing specific somatic cells from animal cells |
| JP2018126195A (en) * | 2017-02-06 | 2018-08-16 | 大日本印刷株式会社 | Liquid feeding method, liquid feeding device, and content filled container |
| JP2020130052A (en) * | 2019-02-20 | 2020-08-31 | 東ソー株式会社 | Cell culture apparatus |
| JP2022529740A (en) * | 2019-04-24 | 2022-06-23 | ダブリュ.エル.ゴア アンド アソシエイツ,インコーポレイティド | Highly durable permeable fluoropolymer cell culture bag |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2024177018A1 (en) | 2024-08-29 |
| WO2024176440A1 (en) | 2024-08-29 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20240158812A1 (en) | Systems and Methods for Point/Center-Of-Care Immunotherapy | |
| CN114717183B (en) | Pluripotent stem cell production system and method for producing induced pluripotent stem cells | |
| CN114807035B (en) | Reproducible differentiation method for clinical-grade retinal pigment epithelial cells | |
| JP2023162277A (en) | Macs-based purification of stem cell-derived retinal pigment epithelium | |
| US20030211603A1 (en) | Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells | |
| CN107267442A (en) | Multipotential stem cell culture on microcarrier | |
| US20230002717A1 (en) | Somatic cell production system | |
| WO2015143342A1 (en) | Production of midbrain dopaminergic neurons and methods for the use thereof | |
| CN108410797A (en) | The expanding stem cells in hollow-fiber bioreactor | |
| WO2017117333A1 (en) | Microtissue formation using stem cell-derived human hepatocytes | |
| US12486485B2 (en) | Cell treatment device, suspension culture vessel, and stem cell induction method | |
| Lin et al. | Integrated generation of induced pluripotent stem cells in a low-cost device | |
| US20220306981A1 (en) | Erythrocyte removal device, mononuclear cell collector, cell culture device, cell culture system, cell culture method, and method for collecting mononuclear cells | |
| JP2014060991A (en) | Method of culturing stem cells using inner lumen of porous hollow fiber | |
| CN120041395B (en) | Preparation method and kit of PBMC (PBMC) reprogrammed Cheng Youdao pluripotent stem cells | |
| JP2019080575A (en) | Somatic cell production system | |
| WO2024177018A1 (en) | Method for producing induced pluripotent stem cells | |
| WO2002014469A2 (en) | Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells | |
| HK1252090A1 (en) | Pluripotent stem cell manufacturing system and method for producing induced pluripotent stem cells | |
| WO2023037544A1 (en) | Method for producing pluripotent stem cells | |
| JP7721836B1 (en) | Method for producing induced pluripotent stem cells | |
| WO2025206056A1 (en) | Production method for induced pluripotent stem cells | |
| JP2025179174A (en) | An end-to-end platform for human pluripotent stem cell manufacturing | |
| Li | Scalable and Cell-friendly Technologies for Cell Manufacturing | |
| WO2025089333A1 (en) | Package of cell production device, cell production method, and management system 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: 24760316 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025502714 Country of ref document: JP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |