Cell expansion

Information

  • Patent Grant
  • 11629332
  • Patent Number
    11,629,332
  • Date Filed
    Monday, April 2, 2018
    6 years ago
  • Date Issued
    Tuesday, April 18, 2023
    a year ago
Abstract
Embodiments described herein generally provide for expanding cells in a cell expansion system. The cells may be grown in a bioreactor, and the cells may be activated by an activator (e.g., a soluble activator complex). Nutrient and gas exchange capabilities of a closed, automated cell expansion system may allow cells to be seeded at reduced cell seeding densities, for example. Parameters of the cell growth environment may be manipulated to load the cells into a particular position in the bioreactor for the efficient exchange of nutrients and gases. System parameters may be adjusted to shear any cell colonies that may form during the expansion phase. Metabolic concentrations may be controlled to improve cell growth and viability. Cell residence in the bioreactor may be controlled. In embodiments, the cells may include T cells. In further embodiments, the cells may include T cell subpopulations, including regulatory T cells (Tregs), for example.
Description
BACKGROUND

Cell Expansion Systems (CESs) are used to expand and differentiate cells. Cell expansion systems may be used to expand, e.g., grow, a variety of adherent and suspension cells. Cells, of both adherent and non-adherent type, may be grown in a bioreactor in a cell expansion system.


SUMMARY

Embodiments of the present disclosure generally relate to expanding cells in a cell expansion system (CES). Such expansion may occur through the use of a bioreactor or cell growth chamber, according to embodiments. In an embodiment, such bioreactor or cell growth chamber may comprise a hollow fiber membrane. Such hollow fiber membrane may include an extracapillary (EC) space and an intracapillary (IC) space. A cell expansion system may expand a variety of cell types. Embodiments may provide for adherent or non-adherent cells to be grown or expanded in the cell expansion system. Non-adherent, or suspension, cells which may be expanded in the system may include T cells, or T lymphocytes, for example. In embodiments, one or more subpopulations or subsets of T cells may be grown. For example, embodiments may provide methods and systems for expanding regulatory T cells (Tregs) or human regulatory T cells (hTregs).


In embodiments, methods and systems may be provided for expanding cells in a closed, automated cell expansion system. In an embodiment, such cell expansion system may include a bioreactor or cell growth chamber. In further embodiments, such bioreactor or cell growth chamber may comprise a hollow fiber membrane.


For example, non-adherent cells, e.g., T cells and/or Treg cells, may be introduced into a hollow fiber bioreactor, in which the hollow fiber bioreactor comprises a plurality of hollow fibers. According to an embodiment, the cells may be exposed to a stimulator or activator to stimulate or activate the expansion of the cells in the hollow fiber bioreactor.


A closed, automated cell expansion system comprising a bioreactor or cell growth chamber may comprise numerous capabilities, such as nutrient and gas exchange capabilities. Such capabilities may allow cells to be seeded at reduced cell seeding densities as compared to cell seeding densities used in culture flasks or other static culture methods. Embodiments provide for parameters of the cell growth environment to be manipulated to load or introduce cells into a position in the bioreactor for the efficient exchange or delivery of nutrients and gases to the growing cells. In embodiments, communication between the cells may also be promoted. For example, in an embodiment, a centralization, or centering, of cells in the bioreactor may increase cell density to promote communication, such as chemical signaling, between cells.


Embodiments also may provide for system parameters to be managed to control cell residence in the bioreactor or cell growth chamber. By controlling cell residence in the bioreactor during the cell growth phase, for example, the system may provide for an efficient gas and nutrient exchange to expanding cells. In embodiments, a bioreactor may be designed to provide gas exchange, and, in some embodiments, nutrient exchange, to growing cells. In an example embodiment, a bioreactor comprising a semi-permeable hollow fiber membrane provides gas and nutrient exchange through the semi-permeable hollow fiber membrane. The semi-permeable hollow fibers of the bioreactor allow essential nutrients (e.g., glucose and/or cell growth formulated media) to reach the cells and metabolic waste products (e.g., lactate) to exit the system via diffusion through the walls of the hollow fibers. However, according to embodiments, cells residing in the headers of the bioreactor or in other areas outside the hollow fibers may not receive proper gas exchange and nutrient exchange, which may result in cell aggregation and death. Embodiments therefore relate to providing methods to retain cell populations, e.g., non-adherent cell populations, in the hollow fibers of the bioreactor when feeding the cells. Such retaining of the cells in the bioreactor may also promote cell-to-cell communications because with more cells in the bioreactor itself, cell densities may increase and cell-to-cell communications may therefore improve.


Additional embodiments may provide for system features to be harnessed to shear any cell colonies, micro-colonies, or cell clusters that may form during the expansion phase of cell growth. For example, an embodiment provides for, after a period of expansion, the shearing of groups of cells, e.g., colonies, micro-colonies, or clusters, through a bioreactor hollow fiber membrane. Such shearing may reduce the number of cells in the micro-colony, colony, or cluster, in which a micro-colony, colony, or cluster may be a group of one or more attached cells. Embodiments may provide a protocol to shear any colonies by circulating the suspension cell culture through, for example, the hollow fiber Intracapillary (IC) loop (e.g., with hollow fibers of 215 μm inner diameter) during the expansion phase of growth. In embodiments, a colony, micro-colony, or cluster of cells may be sheared to reduce a size of the colony, micro-colony, or cluster of cells. In an embodiment, a colony, micro-colony, or cluster of cells may be sheared to provide a single cell uniform suspension and improve cell growth/viability. Such capabilities may contribute to the continuous perfusion growth of the cells, e.g., T cells or Tregs.


Embodiments of the present disclosure further relate to growing cells using a combination of features for cell expansion. For example, in an embodiment, cells may be seeded at a reduced seeding density as compared to a cell seeding density used in culture flasks or other static culture methods. For example, in some embodiments, the volume of fluid used to seed the cells may include from about 1×104 cells/mL to about 1×106 cell/mL, such as on the order of 105 cells/mL. In other embodiments, the volume of fluid used to seed the cells may include less than about 1×106 cells/mL. According to embodiments, the cells may be loaded, or introduced, into a desired position in a bioreactor, e.g., hollow fiber bioreactor, in the cell expansion system. In an embodiment, cells may be exposed to an activator to activate expansion of the cells in the hollow fiber bioreactor, for example. Any colonies or clusters of cells that may form during cell expansion may be sheared by circulating the cells to cause the cells to incur a shear stress and/or shear force, for example, according to an embodiment. Such shear stress and/or shear force may cause one or more cells to break apart from a cell colony, for example. To provide proper gas and nutrient exchange to growing cell populations, further embodiments provide for retaining cell populations, e.g., suspension cell populations, inside the hollow fibers of the bioreactor during the feeding of the cells, for example. By centralizing and/or retaining cells in the hollow fibers of the bioreactor during the cell growth phase, cell densities may increase and cell-to-cell communications may improve. Reduced cell seeding densities, as compared to static cultures, may therefore be used.


Embodiments further provide for a cell expansion system for expanding cells, in which such system may include, for example, a hollow fiber bioreactor comprising an inlet port and an outlet port; a first fluid flow path having at least opposing ends, in which a first opposing end of the first fluid flow path is fluidly associated with an inlet port of the bioreactor, and a second end of the first fluid flow path is fluidly associated with an outlet port of the bioreactor, wherein the first fluid flow path is fluidly associated with an intracapillary portion of the bioreactor; a fluid inlet path fluidly associated with the first fluid flow path; and a first fluid circulation path fluidly associated with the first fluid flow path and the intracapillary portion of the bioreactor.


Embodiments further provide for the cell expansion system to include a processor for executing instructions to perform methods described and/or illustrated herein. For example, embodiments of the present disclosure provide for implementing such expansion of cells through the use of one or more protocols or tasks for use with a cell expansion system. For example, such protocols or tasks may include pre-programmed protocols or tasks. In embodiments, a pre-programmed, default, or otherwise previously saved task may be selected by a user or system operator to perform a specific function by the cell expansion system. In other embodiments, such protocols or tasks may include custom or user-defined protocols or tasks. For example, through a user interface (UI) and one or more graphical user interface (GUI) elements, a custom or user-defined protocol or task may be created. A task may comprise one or more steps.


As used herein, “at least one,” “one or more,” and “and/or” are open-ended expressions that are both conjunctive and disjunctive in operation. For example, each of the expressions “at least one of A, B and C,” “at least one of A, B, or C,” “one or more of A, B, and C,” “one or more of A, B, or C” and “A, B, and/or C” means A alone, B alone, C alone, A and B together, A and C together, B and C together, or A, B and C together.


This Summary is included to provide a selection of concepts in a simplified form, in which such concepts are further described below in the Detailed Description. This Summary is not intended to be used in any way to limit the claimed subject matter's scope. Features, including equivalents and variations thereof, may be included in addition to those provided herein.





BRIEF DESCRIPTION OF THE DRAWINGS

Embodiments of the present disclosure may be described by referencing the accompanying figures. In the figures, like numerals refer to like items. In the figures, dashed lines may be used to indicate that an element(s) may be optional.



FIG. 1A depicts an embodiment of a cell expansion system (CES).



FIG. 1B illustrates a front elevation view of an embodiment of a bioreactor showing circulation paths through the bioreactor.



FIG. 1C depicts a rocking device for moving a cell growth chamber rotationally or laterally during operation of a cell expansion system, according to embodiments of the present disclosure.



FIG. 2 illustrates a perspective view of a cell expansion system with a premounted fluid conveyance device, in accordance with embodiments of the present disclosure.



FIG. 3 depicts a perspective view of a housing of a cell expansion system, in accordance with embodiments of the present disclosure.



FIG. 4 illustrates a perspective view of a premounted fluid conveyance device, in accordance with embodiments of the present disclosure.



FIG. 5A depicts a schematic of a cell expansion system, including an operational configuration showing fluid movement, in accordance with an embodiment of the present disclosure.



FIG. 5B depicts a schematic of a cell expansion system, including another operational configuration showing fluid movement, in accordance with an embodiment of the present disclosure.



FIG. 5C depicts a schematic of a cell expansion system, including another operational configuration showing fluid movement, in accordance with an embodiment of the present disclosure.



FIG. 6 illustrates a schematic of a cell expansion system, in accordance with another embodiment of the present disclosure.



FIG. 7 depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 8A illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 8B depicts a schematic of a portion of a cell expansion system, in accordance with an embodiment of the present disclosure.



FIG. 9A depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 9B illustrates a graph of oxygen consumption in a cell expansion system, in accordance with embodiments of the present disclosure.



FIG. 10A illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 10B illustrates a table with example pump rates that may be used in a cell expansion system, in accordance with embodiments of the present disclosure.



FIG. 11A depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 11B illustrates a graph of the metabolism of expanding cells, in accordance with embodiments of the present disclosure.



FIG. 11C illustrates a graph of the metabolism of expanding cells, in accordance with embodiments of the present disclosure.



FIG. 12 illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 13 depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 14 illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 15A illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 15B illustrates a graph of cell number versus flow rate during cell expansion, in accordance with embodiments of the present disclosure.



FIG. 16 depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 17A depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 17B depicts views of cells expanding in a cell expansion system, in accordance with embodiments of the present disclosure.



FIG. 17C illustrates a graph showing inner diameters of cell disassociation, in accordance with embodiments of the present disclosure.



FIG. 18 illustrates a graph of cell numbers and flow rate versus culture days during cell expansion, in accordance with embodiments of the present disclosure.



FIG. 19A illustrates a flow diagram depicting the operational characteristics of a process for operating pumps to expand cells, in accordance with embodiments of the present disclosure.



FIG. 19B depicts a schematic of a portion of a cell expansion system, in accordance with an embodiment of the present disclosure.



FIG. 19C depicts a schematic of a portion of a cell expansion system, in accordance with an embodiment of the present disclosure.



FIG. 19D depicts a schematic of a portion of a cell expansion system, in accordance with an embodiment of the present disclosure.



FIG. 20 illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 21 depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 22 illustrates a flow diagram depicting the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 23 depicts a flow diagram illustrating the operational characteristics of a process for expanding cells, in accordance with embodiments of the present disclosure.



FIG. 24 depicts an example processing system of a cell expansion system upon which embodiments of the present disclosure may be implemented.



FIG. 25 depicts a flow diagram illustrating an example experimental flow, in accordance with embodiments of the present disclosure.





DETAILED DESCRIPTION

The following Detailed Description provides a discussion of illustrative embodiments with reference to the accompanying drawings. The inclusion of specific embodiments herein should not be construed as limiting or restricting the present disclosure. Further, while language specific to features, acts, and/or structures, for example, may be used in describing embodiments herein, the claims are not limited to the features, acts, and/or structures described. A person of skill in the art will appreciate that other embodiments, including improvements, are within the spirit and scope of the present disclosure. Further, any alternatives or additions, including any listed as separate embodiments, may be used or incorporated with any other embodiments herein described.


Embodiments of the present disclosure are generally directed to systems and methods for expanding cells in a cell expansion system (CES). Such expansion may occur through the use of a bioreactor or cell growth chamber, according to embodiments. In an embodiment, such bioreactor or cell growth chamber may comprise a hollow fiber membrane. Such hollow fiber membrane may include a plurality of hollow fibers and may include an extracapillary (EC) and/or intracapillary (IC) space. Embodiments may provide for adherent or non-adherent cells to be grown or expanded in the cell expansion system. For example, non-adherent or suspension cells, such as T cells, or T lymphocytes, may be expanded in the system. In embodiments, one or more subpopulations or subsets of T cells may be grown. For example, embodiments may provide methods and systems for expanding regulatory T cells (Tregs) and/or human regulatory T cells (hTregs).


In embodiments, methods and systems may be provided for expanding cells in a closed, automated cell expansion system. In an embodiment, such cell expansion system may include a bioreactor or cell growth chamber. In further embodiments, such bioreactor or cell growth chamber may comprise a hollow fiber membrane. The capabilities of such system, such as nutrient and gas exchange capabilities, may allow cells to be seeded at reduced cell seeding densities. Embodiments provide for parameters of the cell growth environment to be manipulated to load or introduce cells into a position in the bioreactor for the efficient exchange of nutrients and gases to the growing cells. For example, in an embodiment, the centralization of cells in the bioreactor may increase cell density.


In embodiments, non-adherent cell populations, e.g., T cells and/or Treg cells, may be introduced or loaded into a hollow fiber bioreactor, in which the hollow fiber bioreactor may comprise a plurality of hollow fibers. In embodiments, the cells may be exposed to an activator to activate the expansion of the cells in the hollow fiber bioreactor. In an embodiment, a plurality of cells may be introduced into a cell expansion system using a “load cells centrally without circulation” task, for example. Such task may be performed on Day 0 and on Days 4-8, according to an example embodiment. Other days may be used in other embodiments. In embodiments, such loading of cells task may result in the centralization of cells in the bioreactor to increase cell density. In other embodiments, the cells may be located in other portions or regions of the bioreactor to increase cell density. In embodiments, by positioning the cells in a first position, e.g., about a central region, of the bioreactor, the cells may receive an efficient exchange of nutrients and gases.


In embodiments, a reduced cell seeding density, as compared to cell seeding densities used with static culture methods, may be used. In an embodiment using a cell expansion system, cells, e.g., Tregs or Treg cells, may be expanded from a cell seeding density of 2.54×105 cells/mL to 3.69×105 cells/mL. In other embodiments, the cell seeding density may be less than about 1×106 cells/mL. In addition, a Treg cell inoculum may be prepared from a cell seeding density of 1.0×105 cells/mL. Other methods, e.g., static Treg cell culture methods, may use a cell seeding density of 1.0×106 Treg cells/mL for in vitro expansion. In an embodiment, lower cell seeding densities may be used due to the system's overall efficiency in delivering nutrients to the culture environment, for example. In other embodiments, one or more of the steps used during expansion in combination with the system's overall efficiency in delivering nutrients to the culture environment may allow lower initial cell seeding densities to be used.


In embodiments, an automated cell, e.g., Treg, expansion may be performed with a soluble activator complex. In other embodiments, other types of activators, such as beads for the stimulation of cells, may be used, for example. In other embodiments, cells, e.g., Treg cells, may be expanded without the use of a bead-based stimulation. In one embodiment, cell expansion may be performed using the Stem Cell Technologies soluble ImmunoCult™ Human CD3/CD28/CD2 T Cell Activator to activate and expand Treg cells in the presence of 200 IU/mL of the cytokine IL-2 with an automated cell expansion system. Using a soluble activator complex may reduce costs for stimulation over the cost of a bead-based protocol, for example. Other types of activators may be used in other embodiments. Further, other types of cytokines or other growth factors may be used in other embodiments.


Embodiments also may provide for system parameters to be adjusted or managed to control cell residence in the bioreactor or cell growth chamber. By controlling cell residence in the bioreactor hollow fibers during the cell growth phase, for example, the system may provide for an efficient gas and nutrient exchange to expanding cells. In embodiments, a bioreactor may be designed to provide gas exchange, and, in some embodiments, nutrient exchange, to growing cells. In an example embodiment, a bioreactor comprising a semi-permeable hollow fiber membrane may provide gas and nutrient exchange through the semi-permeable hollow fiber membrane. In embodiments, methods for providing media constituents, such as various cytokines, proteins, etc., for example, to growing cells which cannot pass through the membrane may use a fluid inlet to the side of the bioreactor, e.g., intracapillary (IC) side, where cells are growing. However, even low, decreased, or reduced, e.g., minimum, inlet flow rates (0.1 mL/min, for example) may result in cells collecting in the outlet header of the bioreactor, according to embodiments. Cells residing in the headers of the bioreactor may not receive proper gas exchange and nutrient exchange, which may result in cell death and aggregation.


Embodiments relate to providing methods to retain cells, e.g., non-adherent cell populations, in the bioreactor when feeding the cells using an inlet, e.g., IC inlet, flow. While embodiments herein may refer to cells being on the IC side of the membrane when feeding, for example, other embodiments may provide for the cells to be on the EC side of the membrane, in which cells may be contained within a first circulation path and/or a second circulation path, according to embodiments. In embodiments, a feeding method may provide for pumping a first volume of fluid, e.g., media or cell growth formulated media, into a first port of the bioreactor at a first volumetric flow rate, volume flow rate, fluid flow rate, flow rate, rate of fluid flow, or volume velocity, for example. Volumetric flow rate, volume flow rate, fluid flow rate, flow rate, rate of fluid flow, or volume velocity, for example, may be used interchangeably. In some embodiments, flow rate may be a vector having both a speed and a direction. A second volume of the fluid may be pumped into a second port of the bioreactor at a second volumetric flow rate, volume flow rate, fluid flow rate, flow rate, rate of fluid flow, or volume velocity. In embodiments, such volumetric flow rate, volume flow rate, fluid flow rate, flow rate, rate of fluid flow, or volume velocity, may be controlled by one or more pump rate(s) and/or pump flow rate(s), for example. A pump rate may produce, cause, or affect a volumetric flow rate or flow rate of a fluid upon which the pump may act. As used herein, a pump rate may be described in embodiments as the volumetric flow rate or fluid flow rate produced, caused, or affected by the pump.


In embodiments, the second flow rate of the fluid into the bioreactor may be opposite the direction of the first flow rate of the fluid into the bioreactor. For example, FIGS. 5B and 5C illustrate example operational configurations showing flow rates and flow directions that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B, & 5C), in accordance with embodiments of the present disclosure. In embodiments, cell expansion system pumps, e.g., IC pumps, may be used to control cell residence in the bioreactor. In embodiments, cells may be lost from the bioreactor into the IC circulation path or IC loop, for example, during the expansion phase of growth. In embodiments, cells in the bioreactor that are closer to IC inlet port, for example, may receive the freshest growth media, whereas, cells in the portion of the IC circulation path outside of the bioreactor, for example, may essentially be receiving expended or conditioned media which may affect their glycolytic metabolism. In addition, cells in the bioreactor may receive mixed gas (O2, CO2, N2) input from the gas transfer module (GTM) by diffusion from the EC loop circulation, whereas cells in the portion of the IC circulation path outside of the bioreactor may not, according to embodiments.


In embodiments, reducing the loss of cells from a hollow fiber membrane (HFM) bioreactor may be accomplished by matching, or closely or substantially matching, the IC circulation pump rate to the IC inlet pump rate, but in the opposite direction, during feeding. For example, an IC inlet pump rate of +0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min in order to maintain cells in the bioreactor during the growth phase of the cell culture which may be Days 4-7, in embodiments. This pump adjustment may counteract the forces associated with the loss of cells from the IC outlet port, in accordance with embodiments. In other embodiments, other pump rates may be used. For example, in other embodiments, the pump rates may be different. In embodiments, other pumps or additional pumps may be used. In an embodiment, fewer pumps may be used. Further, other time periods may be used in other embodiments.


In embodiments, the metabolic activity of the cell population may affect feeding parameters. For example, cell culture lactate values may be maintained at or below a predefined level. In an embodiment, cell culture values may be maintained at or below about 7 mmol/L, for example. In embodiments, by using a cell expansion system graphical user interface (GUI) to control a rate(s) of media addition, lactate metabolic waste product from glycolysis may be maintained at or below a predefined value, during the expansion of cells, e.g., regulatory T cells. In other embodiments, rate(s) of media addition, for example, and/or other settings may be controlled to maintain, or attempt to maintain, the lactate levels≤about 5 mmol/L, for example, to improve cell growth and viability. Other concentrations may be used in other embodiments.


Additional embodiments may provide for system features to be harnessed to shear any cell colonies, micro-colonies, or cell clusters that may form during the expansion phase. For example, an embodiment provides for the shearing of colonies, e.g., micro-colonies, of cells through a bioreactor hollow fiber membrane to reduce the number of cells in the micro-colony, colony, or cluster, in which a micro-colony, colony, or cluster may be a group of one or more attached cells. In embodiments, a cell expansion system (CES) bioreactor architecture may be used to shear cell, e.g., Treg cell, micro-colonies. In embodiments, as cells, e.g., Treg cells, grow, they tend to form micro-colonies that may limit the diffusion of nutrients to the cell(s) in the center of the colony. This may lead to adverse effects such as necrosis during cell culture. Embodiments may provide a protocol to shear the colonies by circulating the suspension cell culture through, for example, the hollow fiber Intracapillary (IC) loop (e.g., with hollow fibers of 215 μm inner diameter) during the expansion phase of growth. In embodiments, a colony, micro-colony, or cluster of cells may be sheared to reduce a size of the colony, micro-colony, or cluster of cells. In an embodiment, a colony or cluster of cells may be sheared to provide a single cell suspension and create cell growth/viability. In embodiments, such capabilities may contribute to the continuous perfusion growth of the cells, e.g., T cells or Tregs.


In embodiments, a therapeutic dose of cells, e.g., Tregs, may be expanded in, and harvested from, a cell expansion system. In embodiments, the number of cells at harvest may be from about 1×106 cells to about 1×1010 cells, such as on the order of 1×109 cells. In one embodiment, the number of harvested cells may be from about 1×108 and 1×1010 cells, one example being between about 7.0×108 to about 1.4×109 cells. In embodiments, the harvested cells may have viabilities between about 60% and about 100%. For example, the viability of the harvested cells may be above about 65%, above about 70%, above about 75%, above about 80%, above about 85%, above about 90%, or even above about 95%. The harvested cells may express biomarkers consistent with Tregs, in some embodiments. For example, the cells may express CD4+, CD25+, and/or FoxP3+ biomarkers, in some embodiments. In embodiments, the harvested cells may include the CD4+CD25+ phenotype at a frequency of between about 50% and about 100%. The harvested cells may include the CD4+CD25+ phenotype at a frequency of above about 75%, above about 80%, above about 85%, above about 90%, or even above about 95%. In other embodiments, the cells may include the CD4+FoxP3+ phenotype at a frequency of between about 30% to about 100%. In some embodiments, the harvested cells may include the CD4+FoxP3+ phenotype at a frequency of above about 30%, above about 35%, above about 40%, above about 45%, above about 50%, above about 55%, above about 60%, above about 65%, or even above about 70%.


Embodiments are directed to a cell expansion system, as noted above. In embodiments, such cell expansion system is closed, in which a closed cell expansion system comprises contents that are not directly exposed to the atmosphere. Such cell expansion system may be automated. In embodiments, cells, of both adherent and non-adherent or suspension type, may be grown in a bioreactor in the cell expansion system. According to embodiments, the cell expansion system may include base media or other type of media. Methods for replenishment of media are provided for cell growth occurring in a bioreactor of the closed cell expansion system. In embodiments, the bioreactor used with such systems is a hollow fiber bioreactor. Many types of bioreactors may be used in accordance with embodiments of the present disclosure.


The system may include, in embodiments, a bioreactor that is fluidly associated with a first fluid flow path having at least opposing ends, a first opposing end of the first fluid flow path fluidly associated with a first port of a hollow fiber membrane and a second end of the first fluid flow path fluidly associated with a second port of the hollow fiber membrane. In embodiments, a hollow fiber membrane comprises a plurality of hollow fibers. The system may further include a fluid inlet path fluidly associated with the first fluid flow path, in which a plurality of cells may be introduced into the first fluid flow path through the first fluid inlet path. In some embodiments, a pump for transferring intracapillary inlet fluid from an intracapillary media bag to the first fluid flow path and a controller for controlling operation of the pump are included. The controller, in embodiments, controls the pump to transfer cells from a cell inlet bag to the first fluid flow path, for example. Another pump for circulating fluid in a first fluid circulation path may also be included, in which such pump may also include a controller for controlling operation of the pump. In an embodiment, a controller is a computing system, including a processor(s), for example. The one or more controller(s) may be configured, in embodiments, to control the one or more pump(s), such as to circulate a fluid at a flow rate within the first fluid circulation path, for example. A number of controllers may be used, e.g., a first controller, second controller, third controller, fourth controller, fifth controller, sixth controller, etc., in accordance with embodiments. Further, a number of pumps may be used, e.g., a first pump, second pump, third pump, fourth pump, fifth pump, sixth pump, etc., in accordance with embodiments of the present disclosure. In addition, while the present disclosure may refer to a media bag, a cell inlet bag, etc., multiple bags, e.g., a first media bag, a second media bag, a third media bag, a first cell inlet bag, a second cell inlet bag, a third cell inlet bag, etc., and/or other types of containers, may be used in embodiments. In other embodiments, a single media bag, a single cell inlet bag, etc., may be used. Further, additional or other fluid paths, e.g., a second fluid flow path, a second fluid inlet path, a second fluid circulation path, etc., may be included in embodiments.


In embodiments, the system is controlled by, for example: a processor coupled to the cell expansion system; a display device, in communication with the processor, and operable to display data; and a memory, in communication with and readable by the processor, and containing a series of instructions. In embodiments, when the instructions are executed by the processor, the processor receives an instruction to prime the system, for example. In response to the instruction to prime the system, the processor may execute a series of steps to prime the system and may next receive an instruction to perform an IC/EC washout, for example. In response to an instruction to load cells, for example, the processor may execute a series of steps to load the cells from a cell inlet bag, for example, into the bioreactor.


A schematic of an example cell expansion system (CES) is depicted in FIG. 1A, in accordance with embodiments of the present disclosure. CES 10 includes first fluid circulation path 12 and second fluid circulation path 14. First fluid flow path 16 has at least opposing ends 18 and 20 fluidly associated with a hollow fiber cell growth chamber 24 (also referred to herein as a “bioreactor”), according to embodiments. Specifically, opposing end 18 may be fluidly associated with a first inlet 22 of cell growth chamber 24, and opposing end 20 may be fluidly associated with first outlet 28 of cell growth chamber 24. Fluid in first circulation path 12 flows through the interior of hollow fibers 116 (see FIG. 1B) of hollow fiber membrane 117 (see FIG. 1B) disposed in cell growth chamber 24 (cell growth chambers and hollow fiber membranes are described in more detail infra). Further, first fluid flow control device 30 may be operably connected to first fluid flow path 16 and may control the flow of fluid in first circulation path 12.


Second fluid circulation path 14 includes second fluid flow path 34, cell growth chamber 24, and a second fluid flow control device 32. The second fluid flow path 34 has at least opposing ends 36 and 38, according to embodiments. Opposing ends 36 and 38 of second fluid flow path 34 may be fluidly associated with inlet port 40 and outlet port 42 respectively of cell growth chamber 24. Fluid flowing through cell growth chamber 24 may be in contact with the outside of hollow fiber membrane 117 (see FIG. 1B) in the cell growth chamber 24, in which a hollow fiber membrane comprises a plurality of hollow fibers. Second fluid circulation path 14 may be operably connected to second fluid flow control device 32.


First and second fluid circulation paths 12 and 14 may thus be separated in cell growth chamber 24 by a hollow fiber membrane 117 (see FIG. 1B). Fluid in first fluid circulation path 12 flows through the intracapillary (“IC”) space of the hollow fibers in the cell growth chamber 24. First circulation path 12 may be referred to as the “IC loop.” Fluid in second circulation path 14 flows through the extracapillary (“EC”) space in the cell growth chamber 24. Second fluid circulation path 14 may be referred to as the “EC loop.” Fluid in first fluid circulation path 12 may flow in either a co-current or counter-current direction with respect to flow of fluid in second fluid circulation path 14, according to embodiments.


Fluid inlet path 44 may be fluidly associated with first fluid circulation path 12. Fluid inlet path 44 allows fluid into first fluid circulation path 12, while fluid outlet path 46 allows fluid to leave CES 10. Third fluid flow control device 48 may be operably associated with fluid inlet path 44. Alternatively, third fluid flow control device 48 may alternatively be associated with first outlet path 46.


Fluid flow control devices as used herein may comprise a pump, valve, clamp, or combination thereof, according to embodiments. Multiple pumps, valves, and clamps can be arranged in any combination. In various embodiments, the fluid flow control device is or includes a peristaltic pump. In embodiments, fluid circulation paths, inlet ports, and outlet ports may be constructed of tubing of any material.


Various components are referred to herein as “operably associated.” As used herein, “operably associated” refers to components that are linked together in operable fashion and encompasses embodiments in which components are linked directly, as well as embodiments in which additional components are placed between the two linked components. “Operably associated” components can be “fluidly associated.” “Fluidly associated” refers to components that are linked together such that fluid can be transported between them. “Fluidly associated” encompasses embodiments in which additional components are disposed between the two fluidly associated components, as well as components that are directly connected. Fluidly associated components can include components that do not contact fluid, but contact other components to manipulate the system (e.g., a peristaltic pump that pumps fluids through flexible tubing by compressing the exterior of the tube).


Generally, any kind of fluid, including buffers, protein containing fluid, and cell-containing fluid, for example, can flow through the various circulations paths, inlet paths, and outlet paths. As used herein, “fluid,” “media,” and “fluid media” are used interchangeably.


Turning to FIG. 1B, an example of a hollow fiber cell growth chamber 100 which may be used with the present disclosure is shown in front side elevation view. Cell growth chamber 100 has a longitudinal axis LA-LA and includes cell growth chamber housing 104. In at least one embodiment, cell growth chamber housing 104 includes four openings or ports: IC inlet port 108, IC outlet port 120, EC inlet port 128, and EC outlet port 132.


According to embodiments of the present disclosure, fluid in a first circulation path enters cell growth chamber 100 through IC inlet port 108 at a first longitudinal end 112 of the cell growth chamber 100, passes into and through the intracapillary side (referred to in various embodiments as the intracapillary (“IC”) side or “IC space” of a hollow fiber membrane) of a plurality of hollow fibers 116 comprising hollow fiber membrane 117, and out of cell growth chamber 100 through IC outlet port 120 located at a second longitudinal end 124 of the cell growth chamber 100. The fluid path between the IC inlet port 108 and the IC outlet port 120 defines the IC portion 126 of the cell growth chamber 100. Fluid in a second circulation path flows in the cell growth chamber 100 through EC inlet port 128, comes in contact with the extracapillary side or outside (referred to as the “EC side” or “EC space” of the membrane) of the hollow fibers 116, and exits cell growth chamber 100 via EC outlet port 132. The fluid path between the EC inlet port 128 and the EC outlet port 132 comprises the EC portion 136 of the cell growth chamber 100. Fluid entering cell growth chamber 100 via the EC inlet port 128 may be in contact with the outside of the hollow fibers 116. Small molecules (e.g., ions, water, oxygen, lactate, etc.) may diffuse through the hollow fibers 116 from the interior or IC space of the hollow fiber to the exterior or EC space, or from the EC space to the IC space. Large molecular weight molecules, such as growth factors, may be typically too large to pass through the hollow fiber membrane, and may remain in the IC space of the hollow fibers 116. The media may be replaced as needed, in embodiments. Media may also be circulated through an oxygenator or gas transfer module to exchange gasses as needed. Cells may be contained within a first circulation path and/or a second circulation path, as described below, and may be on either the IC side and/or EC side of the membrane, according to embodiments.


The material used to make the hollow fiber membrane 117 may be any biocompatible polymeric material which is capable of being made into hollow fibers. One material which may be used is a synthetic polysulfone-based material, according to an embodiment of the present disclosure.


In embodiments, the CES (such as CES 500 (see FIGS. 5A, 5B, & 5C) and/or CES 600 (see FIG. 6), for example) may include a device configured to move or “rock” the cell growth chamber relative to other components of the cell expansion system by attaching it to a rotational and/or lateral rocking device. FIG. 1C shows one such device, in which a bioreactor 100 may be rotationally connected to two rotational rocking components and to a lateral rocking component, according to an embodiment.


A first rotational rocking component 138 rotates the bioreactor 100 around central axis 142 of the bioreactor 100. Rotational rocking component 138 may be rotationally associated with bioreactor 100. In embodiments, bioreactor 100 may be rotated continuously in a single direction around central axis 142 in a clockwise or counterclockwise direction. Alternatively, bioreactor 100 may rotate in alternating fashion, first clockwise, then counterclockwise, for example, around central axis 142, according to embodiments.


The CES may also include a second rotational rocking component that rotates bioreactor 100 around rotational axis 144. Rotational axis 144 may pass through the center point of bioreactor 100 and may be normal to central axis 142. Bioreactor 100 may be rotated continuously in a single direction around rotational axis 144 in a clockwise or counterclockwise direction, in embodiments. Alternatively, bioreactor 100 may be rotated around rotational axis 144 in an alternating fashion, first clockwise, then counterclockwise, for example. In various embodiments, bioreactor 100 may also be rotated around rotational axis 144 and positioned in a horizontal or vertical orientation relative to gravity.


In embodiments, lateral rocking component 140 may be laterally associated with bioreactor 100. The plane of lateral rocking component 140 moves laterally in the −x and −y directions, in embodiments. The settling of cells in the bioreactor may be reduced by movement of cell-containing media within the hollow fibers, according to embodiments.


The rotational and/or lateral movement of a rocking device may reduce the settling of cells within the device and reduce the likelihood of cells becoming trapped within a portion of the bioreactor. The rate of cells settling in the cell growth chamber is proportional to the density difference between the cells and the suspension media, according to Stokes's Law. In certain embodiments, a 180-degree rotation (fast) with a pause (having a total combined time of 30 seconds, for example) repeated as described above keeps non-adherent red blood cells, for example, suspended. A minimum rotation of about 180 degrees would be preferred in an embodiment; however, one could use rotation of up to 360 degrees or greater. Different rocking components may be used separately, or may be combined in any combination. For example, a rocking component that rotates bioreactor 100 around central axis 142 may be combined with the rocking component that rotates bioreactor 100 around axis 144. Likewise, clockwise and counterclockwise rotation around different axes may be performed independently in any combination.


Turning to FIG. 2, an embodiment of a cell expansion system 200 with a premounted fluid conveyance assembly is shown in accordance with embodiments of the present disclosure. The CES 200 includes a cell expansion machine 202 that comprises a hatch or closable door 204 for engagement with a back portion 206 of the cell expansion machine 202. An interior space 208 within the cell expansion machine 202 includes features adapted for receiving and engaging a premounted fluid conveyance assembly 210. The premounted fluid conveyance assembly 210 is detachably-attachable to the cell expansion machine 202 to facilitate relatively quick exchange of a new or unused premounted fluid conveyance assembly 210 at a cell expansion machine 202 for a used premounted fluid conveyance assembly 210 at the same cell expansion machine 202. A single cell expansion machine 202 may be operated to grow or expand a first set of cells using a first premounted fluid conveyance assembly 210 and, thereafter, may be used to grow or expand a second set of cells using a second premounted fluid conveyance assembly 210 without needing to be sanitized between interchanging the first premounted fluid conveyance assembly 210 for the second premounted fluid conveyance assembly 210. The premounted fluid conveyance assembly 210 includes a bioreactor 100 and an oxygenator or gas transfer module 212 (also see FIG. 4). Tubing guide slots are shown as 214 for receiving various media tubing connected to premounted fluid conveyance assembly 210, according to embodiments.


Next, FIG. 3 illustrates the back portion 206 of cell expansion machine 202 prior to detachably-attaching a premounted fluid conveyance assembly 210 (FIG. 2), in accordance with embodiments of the present disclosure. The closable door 204 (shown in FIG. 2) is omitted from FIG. 3. The back portion 206 of the cell expansion machine 202 includes a number of different structures for working in combination with elements of a premounted fluid conveyance assembly 210. More particularly, the back portion 206 of the cell expansion machine 202 includes a plurality of peristaltic pumps for cooperating with pump loops on the premounted fluid conveyance assembly 210, including the IC circulation pump 218, the EC circulation pump 220, the IC inlet pump 222, and the EC inlet pump 224. In addition, the back portion 206 of the cell expansion machine 202 includes a plurality of valves, including the IC circulation valve 226, the reagent valve 228, the IC media valve 230, the air removal valve 232, the cell inlet valve 234, the wash valve 236, the distribution valve 238, the EC media valve 240, the IC waste or outlet valve 242, the EC waste valve 244, and the harvest valve 246. Several sensors are also associated with the back portion 206 of the cell expansion machine 202, including the IC outlet pressure sensor 248, the combination IC inlet pressure and temperature sensors 250, the combination EC inlet pressure and temperature sensors 252, and the EC outlet pressure sensor 254. Also shown is an optical sensor 256 for an air removal chamber, according to an embodiment.


In accordance with embodiments, a shaft or rocker control 258 for rotating the bioreactor 100 is shown. Shaft fitting 260 associated with the shaft or rocker control 258 allows for proper alignment of a shaft access aperture, see e.g., 424 (FIG. 4) of a tubing-organizer, see e.g., 300 (FIG. 4) of a premounted conveyance assembly 210 or 400 with the back portion 206 of the cell expansion machine 202. Rotation of shaft or rocker control 258 imparts rotational movement to shaft fitting 260 and bioreactor 100. Thus, when an operator or user of the CES 200 attaches a new or unused premounted fluid conveyance assembly 400 (FIG. 4) to the cell expansion machine 202, the alignment is a relatively simple matter of properly orienting the shaft access aperture 424 (FIG. 4) of the premounted fluid conveyance assembly 210 or 400 with the shaft fitting 260.


Turning to FIG. 4, a perspective view of a detachably-attachable premounted fluid conveyance assembly 400 is shown. The premounted fluid conveyance assembly 400 may be detachably-attachable to the cell expansion machine 202 (FIGS. 2 and 3) to facilitate relatively quick exchange of a new or unused premounted fluid conveyance assembly 400 at a cell expansion machine 202 for a used premounted fluid conveyance assembly 400 at the same cell expansion machine 202. As shown in FIG. 4, the bioreactor 100 may be attached to a bioreactor coupling that includes a shaft fitting 402. The shaft fitting 402 includes one or more shaft fastening mechanisms, such as a biased arm or spring member 404 for engaging a shaft, e.g., 258 (shown in FIG. 3), of the cell expansion machine 202.


According to embodiments, the premounted fluid conveyance assembly 400 includes tubing 408A, 408B, 408C, 408D, 408E, etc., and various tubing fittings to provide the fluid paths shown in FIGS. 5A, 5B, 5C, and 6, as discussed below. Pump loops 406A, 406B, and 406C may also be provided for the pump(s). In embodiments, although the various media may be provided at the site where the cell expansion machine 202 is located, the premounted fluid conveyance assembly 400 may include sufficient tubing length to extend to the exterior of the cell expansion machine 202 and to enable welded connections to tubing associated with media bag(s) or container(s), according to embodiments.


Next, FIGS. 5A, 5B, and 5C illustrate schematics of embodiments of a cell expansion system 500, and FIG. 6 illustrates a schematic of another embodiment of a cell expansion system 600. In the embodiments shown in FIGS. 5A, 5B, 5C, and 6, and as described below, the cells are grown in the IC space. However, the disclosure is not limited to such examples and may in other embodiments provide for cells to be grown in the EC space.


As noted, FIGS. 5A, 5B, and 5C illustrate a CES 500. While FIGS. 5A, 5B, and 5C depict substantially similar structural components of CES 500, FIGS. 5A, 5B, and 5C illustrate possible operational configurations of fluid movement in a first fluid circulation path using the structural features of CES 500, in accordance with embodiments of the present disclosure. As shown, CES 500 includes first fluid circulation path 502 (also referred to as the “intracapillary loop” or “IC loop”) and second fluid circulation path 504 (also referred to as the “extracapillary loop” or “EC loop”), according to embodiments. First fluid flow path 506 may be fluidly associated with cell growth chamber 501 to form first fluid circulation path 502. Fluid flows into cell growth chamber 501 through IC inlet port 501A, through hollow fibers in cell growth chamber 501, and exits via IC outlet port 501B. Pressure gauge 510 measures the pressure of media leaving cell growth chamber or bioreactor 501. Media flows through IC circulation pump 512 which may be used to control the rate of media flow. IC circulation pump 512 may pump the fluid in a first direction or second direction opposite the first direction. Exit port 501B may be used as an inlet in the reverse direction. For example, in a first configuration, the IC circulation pump may pump the fluid in a positive direction, in which the fluid enters the IC inlet port 501A. In a second configuration, for example, the IC circulation pump may pump the fluid in a negative direction, in which the fluid enters the IC outlet port 501B, for example.


Media entering the IC loop may enter through valve 514. As those skilled in the art will appreciate, additional valves, pressure gauges, pressure/temperature sensors, ports, and/or other devices may be placed at various locations to isolate and/or measure characteristics of the media along portions of the fluid paths. Accordingly, it is to be understood that the schematic shown represents one possible configuration for various elements of the CES 500, and modifications to the schematic shown are within the scope of the one or more present embodiments.


With regard to the IC loop 502, samples of media may be obtained from sample port 516 or sample coil 518 during operation. Pressure/temperature gauge 520 disposed in first fluid circulation path 502 allows detection of media pressure and temperature during operation. Media then returns to IC inlet port 501A to complete fluid circulation path 502. Cells grown/expanded in cell growth chamber 501 may be flushed out of cell growth chamber 501 into harvest bag 599 through valve 598 or redistributed within the hollow fibers for further growth.


Fluid in second fluid circulation path 504 enters cell growth chamber 501 via EC inlet port 501C, and leaves cell growth chamber 501 via EC outlet port 501D. Media in the EC loop 504 may be in contact with the outside of the hollow fibers in the cell growth chamber 501, thereby allowing diffusion of small molecules into and out of the hollow fibers.


Pressure/temperature gauge 524 disposed in the second fluid circulation path 504 allows the pressure and temperature of media to be measured before the media enters the EC space of the cell growth chamber 501, according to an embodiment. Pressure gauge 526 allows the pressure of media in the second fluid circulation path 504 to be measured after it leaves the cell growth chamber 501. With regard to the EC loop, samples of media may be obtained from sample port 530 or a sample coil during operation.


In embodiments, after leaving EC outlet port 501D of cell growth chamber 501, fluid in second fluid circulation path 504 passes through EC circulation pump 528 to oxygenator or gas transfer module 532. EC circulation pump 528 may also pump the fluid in opposing directions. Second fluid flow path 522 may be fluidly associated with oxygenator or gas transfer module 532 via oxygenator inlet port 534 and oxygenator outlet port 536. In operation, fluid media flows into oxygenator or gas transfer module 532 via oxygenator inlet port 534, and exits oxygenator or gas transfer module 532 via oxygenator outlet port 536. Oxygenator or gas transfer module 532 adds oxygen to, and removes bubbles from, media in the CES 500, for example. In various embodiments, media in second fluid circulation path 504 may be in equilibrium with gas entering oxygenator or gas transfer module 532. The oxygenator or gas transfer module 532 may be any appropriately sized oxygenator or gas transfer device. Air or gas flows into oxygenator or gas transfer module 532 via filter 538 and out of oxygenator or gas transfer device 532 through filter 540. Filters 538 and 540 reduce or prevent contamination of oxygenator or gas transfer module 532 and associated media. Air or gas purged from the CES 500 during portions of a priming sequence may vent to the atmosphere via the oxygenator or gas transfer module 532.


In accordance with at least one embodiment, media, including cells (from bag 562), and fluid media from bag 546 may be introduced to first fluid circulation path 502 via first fluid flow path 506. Fluid container 562 (e.g., Cell Inlet Bag or Saline Priming Fluid for priming air out of the system) may be fluidly associated with the first fluid flow path 506 and the first fluid circulation path 502 via valve 564.


Fluid containers, or media bags, 544 (e.g., Reagent) and 546 (e.g., IC Media) may be fluidly associated with either first fluid inlet path 542 via valves 548 and 550, respectively, or second fluid inlet path 574 via valves 570 and 576. First and second sterile sealable input priming paths 508 and 509 are also provided. An air removal chamber (ARC) 556 may be fluidly associated with first circulation path 502. The air removal chamber 556 may include one or more ultrasonic sensors including an upper sensor and lower sensor to detect air, a lack of fluid, and/or a gas/fluid interface, e.g., an air/fluid interface, at certain measuring positions within the air removal chamber 556. For example, ultrasonic sensors may be used near the bottom and/or near the top of the air removal chamber 556 to detect air, fluid, and/or an air/fluid interface at these locations. Embodiments provide for the use of numerous other types of sensors without departing from the spirit and scope of the present disclosure. For example, optical sensors may be used in accordance with embodiments of the present disclosure. Air or gas purged from the CES 500 during portions of the priming sequence or other protocols may vent to the atmosphere out air valve 560 via line 558 that may be fluidly associated with air removal chamber 556.


EC media (e.g., from bag 568) or wash solution (e.g., from bag 566) may be added to either the first or second fluid flow paths. Fluid container 566 may be fluidly associated with valve 570 that may be fluidly associated with first fluid circulation path 502 via distribution valve 572 and first fluid inlet path 542. Alternatively, fluid container 566 may be fluidly associated with second fluid circulation path 504 via second fluid inlet path 574 and EC inlet path 584 by opening valve 570 and closing distribution valve 572. Likewise, fluid container 568 may be fluidly associated with valve 576 that may be fluidly associated with first fluid circulation path 502 via first fluid inlet path 542 and distribution valve 572. Alternatively, fluid container 568 may be fluidly associated with second fluid inlet path 574 by opening valve 576 and closing distribution valve 572.


An optional heat exchanger 552 may be provided for media reagent or wash solution introduction.


In the IC loop, fluid may be initially advanced by the IC inlet pump 554. In the EC loop, fluid may be initially advanced by the EC inlet pump 578. An air detector 580, such as an ultrasonic sensor, may also be associated with the EC inlet path 584.


In at least one embodiment, first and second fluid circulation paths 502 and 504 are connected to waste line 588. When valve 590 is opened, IC media may flow through waste line 588 and to waste or outlet bag 586. Likewise, when valve 582 is opened, EC media may flow through waste line 588 to waste or outlet bag 586.


In embodiments, cells may be harvested via cell harvest path 596. Here, cells from cell growth chamber 501 may be harvested by pumping the IC media containing the cells through cell harvest path 596 and valve 598 to cell harvest bag 599.


Various components of the CES 500 may be contained or housed within a machine or housing, such as cell expansion machine 202 (FIGS. 2 and 3), wherein the machine maintains cells and media, for example, at a predetermined temperature.


In the configuration depicted for CES 500 in FIG. 5A, fluid media in first fluid circulation path 502 and second fluid circulation path 504 flows through cell growth chamber 501 in the same direction (a co-current configuration), in an embodiment. The CES 500 may also be configured to flow in a counter-current conformation (not shown), in another embodiment. In the configuration shown in FIG. 5A, fluid in first fluid circulation path 502 enters the bioreactor 501 at IC inlet port 501A and exits the bioreactor 501 at IC outlet port 501B. In the configurations depicted in FIGS. 5B and 5C, fluid media in first circulation path 502 may flow in opposite or opposing directions from connection 517 such that fluid may enter IC inlet port, a first port, 501A on one end of the bioreactor, and fluid may enter IC outlet port, a second port, 501B on the opposing end of the bioreactor to retain cells in the bioreactor itself, according to embodiments. The first fluid flow path may be fluidly associated with the first fluid circulation path through connection 517. In embodiments, connection 517 may be a point or location from which the fluid may flow in opposite directions, for example, based on the direction of the IC inlet pump and the direction of the IC circulation pump. In an embodiment, connection 517 may be a T-fitting or T-coupling. In another embodiment, connection 517 may be a Y-fitting or Y-coupling. Connection 517 may be any type of fitting, coupling, fusion, pathway, tubing, etc., allowing the first fluid flow path to be fluidly associated with the first circulation path. It is to be understood that the schematics and operational configurations shown in FIGS. 5A, 5B, and 5C represent possible configurations for various elements of the cell expansion system, and modifications to the schematics and operational configurations shown are within the scope of the one or more present embodiments.


Turning to FIG. 6, a schematic of another embodiment of a cell expansion system 600 is shown. CES 600 includes a first fluid circulation path 602 (also referred to as the “intracapillary loop” or “IC loop”) and second fluid circulation path 604 (also referred to as the “extracapillary loop” or “EC loop”). First fluid flow path 606 may be fluidly associated with cell growth chamber 601 to form first fluid circulation path 602. Fluid flows into cell growth chamber 601 through IC inlet port 601A, through hollow fibers in cell growth chamber 601, and exits via IC outlet port 601B. Pressure sensor 610 measures the pressure of media leaving cell growth chamber 601. In addition to pressure, sensor 610 may, in embodiments, also be a temperature sensor that detects the media pressure and temperature during operation. Media flows through IC circulation pump 612 which may be used to control the rate of media flow. IC circulation pump 612 may pump the fluid in a first direction or second direction opposite the first direction. Exit port 601B may be used as an inlet in the reverse direction. Media entering the IC loop may enter through valve 614. As those skilled in the art will appreciate, additional valves, pressure gauges, pressure/temperature sensors, ports, and/or other devices may be placed at various locations to isolate and/or measure characteristics of the media along portions of the fluid paths. Accordingly, it is to be understood that the schematic shown represents one possible configuration for various elements of the CES 600, and modifications to the schematic shown are within the scope of the one or more present embodiments.


With regard to the IC loop, samples of media may be obtained from sample coil 618 during operation. Media then returns to IC inlet port 601A to complete fluid circulation path 602. Cells grown/expanded in cell growth chamber 601 may be flushed out of cell growth chamber 601 into harvest bag 699 through valve 698 and line 697. Alternatively, when valve 698 is closed, the cells may be redistributed within chamber 601 for further growth.


Fluid in second fluid circulation path 604 enters cell growth chamber 601 via EC inlet port 601C and leaves cell growth chamber 601 via EC outlet port 601D. Media in the EC loop may be in contact with the outside of the hollow fibers in the cell growth chamber 601, thereby allowing diffusion of small molecules into and out of the hollow fibers that may be within chamber 601, according to an embodiment.


Pressure/temperature sensor 624 disposed in the second fluid circulation path 604 allows the pressure and temperature of media to be measured before the media enters the EC space of the cell growth chamber 601. Sensor 626 allows the pressure and/or temperature of media in the second fluid circulation path 604 to be measured after it leaves the cell growth chamber 601. With regard to the EC loop, samples of media may be obtained from sample port 630 or a sample coil during operation.


After leaving EC outlet port 601D of cell growth chamber 601, fluid in second fluid circulation path 604 passes through EC circulation pump 628 to oxygenator or gas transfer module 632. EC circulation pump 628 may also pump the fluid in opposing directions, according to embodiments. Second fluid flow path 622 may be fluidly associated with oxygenator or gas transfer module 632 via an inlet port 632A and an outlet port 632B of oxygenator or gas transfer module 632. In operation, fluid media flows into oxygenator or gas transfer module 632 via inlet port 632A, and exits oxygenator or gas transfer module 632 via outlet port 632B. Oxygenator or gas transfer module 632 adds oxygen to, and removes bubbles from, media in the CES 600, for example. In various embodiments, media in second fluid circulation path 604 may be in equilibrium with gas entering oxygenator or gas transfer module 632. The oxygenator or gas transfer module 632 may be any appropriately sized device useful for oxygenation or gas transfer. Air or gas flows into oxygenator or gas transfer module 632 via filter 638 and out of oxygenator or gas transfer device 632 through filter 640. Filters 638 and 640 reduce or prevent contamination of oxygenator or gas transfer module 632 and associated media. Air or gas purged from the CES 600 during portions of a priming sequence may vent to the atmosphere via the oxygenator or gas transfer module 632.


In the configuration depicted for CES 600, fluid media in first fluid circulation path 602 and second fluid circulation path 604 flows through cell growth chamber 601 in the same direction (a co-current configuration). The CES 600 may also be configured to flow in a counter-current conformation, according to embodiments.


In accordance with at least one embodiment, media, including cells (from a source such as a cell container, e.g., a bag) may be attached at attachment point 662, and fluid media from a media source may be attached at attachment point 646. The cells and media may be introduced into first fluid circulation path 602 via first fluid flow path 606. Attachment point 662 may be fluidly associated with the first fluid flow path 606 via valve 664, and attachment point 646 may be fluidly associated with the first fluid flow path 606 via valve 650. A reagent source may be fluidly connected to point 644 and be associated with fluid inlet path 642 via valve 648, or second fluid inlet path 674 via valves 648 and 672.


Air removal chamber (ARC) 656 may be fluidly associated with first circulation path 602. The air removal chamber 656 may include one or more sensors including an upper sensor and lower sensor to detect air, a lack of fluid, and/or a gas/fluid interface, e.g., an air/fluid interface, at certain measuring positions within the air removal chamber 656. For example, ultrasonic sensors may be used near the bottom and/or near the top of the air removal chamber 656 to detect air, fluid, and/or an air/fluid interface at these locations. Embodiments provide for the use of numerous other types of sensors without departing from the spirit and scope of the present disclosure. For example, optical sensors may be used in accordance with embodiments of the present disclosure. Air or gas purged from the CES 600 during portions of a priming sequence or other protocol(s) may vent to the atmosphere out air valve 660 via line 658 that may be fluidly associated with air removal chamber 656.


An EC media source may be attached to EC media attachment point 668, and a wash solution source may be attached to wash solution attachment point 666, to add EC media and/or wash solution to either the first or second fluid flow path. Attachment point 666 may be fluidly associated with valve 670 that may be fluidly associated with first fluid circulation path 602 via valve 672 and first fluid inlet path 642. Alternatively, attachment point 666 may be fluidly associated with second fluid circulation path 604 via second fluid inlet path 674 and second fluid flow path 684 by opening valve 670 and closing valve 672. Likewise, attachment point 668 may be fluidly associated with valve 676 that may be fluidly associated with first fluid circulation path 602 via first fluid inlet path 642 and valve 672. Alternatively, fluid container 668 may be fluidly associated with second fluid inlet path 674 by opening valve 676 and closing distribution valve 672.


In the IC loop, fluid may be initially advanced by the IC inlet pump 654. In the EC loop, fluid may be initially advanced by the EC inlet pump 678. An air detector 680, such as an ultrasonic sensor, may also be associated with the EC inlet path 684.


In at least one embodiment, first and second fluid circulation paths 602 and 604 are connected to waste line 688. When valve 690 is opened, IC media may flow through waste line 688 and to waste or outlet bag 686. Likewise, when valve 692 is opened, EC media may flow to waste or outlet bag 686.


After cells have been grown in cell growth chamber 601, they may be harvested via cell harvest path 697. Here, cells from cell growth chamber 601 may be harvested by pumping the IC media containing the cells through cell harvest path 697, with valve 698 open, into cell harvest bag 699.


Various components of the CES 600 may be contained or housed within a machine or housing, such as cell expansion machine 202 (FIGS. 2 and 3), wherein the machine maintains cells and media, for example, at a predetermined temperature. It is further noted that, in embodiments, components of CES 600 and CES 500 may be combined. In other embodiments, a CES may include fewer or additional components than those shown in CES 500 and/or CES 600 and still be within the scope of the present disclosure. An example of a cell expansion system that may incorporate features of the present disclosure is the Quantum® Cell Expansion System, manufactured by Terumo BCT, Inc. in Lakewood, Colo.


It is to be understood that the schematic shown in FIG. 6 represents a possible configuration for various elements of the cell expansion system, and modifications to the schematic shown are within the scope of the one or more present embodiments.


Examples and further description of cell expansion systems are provided in U.S. Pat. No. 8,309,347 (“Cell Expansion System and Methods of Use,” issued on Nov. 13, 2012) and U.S. Pat. No. 9,057,045, filed on Dec. 15, 2010, (“Method of Loading and Distributing Cells in a Bioreactor of a Cell Expansion System,” issued on Jun. 16, 2015), which are hereby incorporated by reference herein in their entireties for all that they teach and for all purposes.


While various example embodiments of a cell expansion system and methods associated therewith have been described, FIG. 7 illustrates example operational steps 700 of a process for expanding non-adherent, or suspension, cells in a cell expansion system, such as CES 500 or CES 600, in accordance with embodiments of the present disclosure.


START operation 702 is initiated, and process 700 proceeds to preparation of cells 704. In embodiments, the preparation of cells 704 may involve a number of different and optional steps. For example, the cells may be collected 706. The collection of cells 706 may involve separating and collecting the cells from a donor. In some embodiments, an apheresis procedure may be performed to collect a volume of lymphocytes from the peripheral blood of a donor, e.g., leukapheresis. The volume of lymphocytes may include the target cell population to be expanded by process 700. In other embodiments, the cells may be collected from cord blood.


After collection 706, optionally, the cells may be isolated 708 as part of the preparation 704. The volume of cells collected at step 706 may include a number of different cell types including the cells that are targeted for expansion. Optional step 708 may be performed to isolate the target cells. As one example, the target cells may be T cells, e.g., regulated T cells. In one embodiment, the regulated T cells may be CD4+CD25+ T cells. The cells may be isolated using any suitable isolation technique. For example, the cells may be isolated using immunomagnetic separation where magnetic beads functionalized with antibodies are contacted with the cells collected at 706. The functionalized beads may preferentially attach to the target cell population. A magnetic field may then be used to retain the beads with the attached target cell population, while the other cells may be removed.


The cells may be optionally resuspended 710 after isolation 708. In embodiments, the cells may be resuspended in a media that includes a number of nutrients and/or reagents that aid in maintaining the viability of the cells. In embodiments, the media may include at least serum albumin and a reagent, such as a cytokine. The cytokine may in embodiments be a recombinant human IL-2 cytokine. The media may include the cytokine at a concentrate of 200 IU/ml, in one embodiment.


Following the preparation of the cells 704, process 700 proceeds to expose cells 712 in order to activate the cells to expand. The cells may optionally be exposed to an activator 714 that is soluble. The activator, which may include antibody complexes, may be added to the media in which the cells are resuspended. In embodiments, the activator may be a human antibody CD3/CD28/CD2 cell activator complex. In some embodiments, the activator, may be included in the media used in the resuspension of the cells 710. Optionally, the cells may be exposed to beads 716, which may have an activator on their surface. In embodiments, exposing the cells to the beads may involve adding a predetermined amount of beads to the resuspended cells. The beads may be added at different ratios with respect to the number of cells. For example, the beads may be added in a 1 bead:2 cell ratio. Other embodiments may provide for adding beads at different ratios, e.g., 1 bead:1 cell, 1 bead:3 cells, etc. The beads may have antibodies on their surface to activate the cells to expand. In embodiments, the beads may include antibodies CD3/CD28 on their surface.


Process 700 proceeds to expand cells 718. As part of cell expansion 718, the cells may be loaded into a cell growth chamber, e.g., a hollow fiber membrane bioreactor, where the cells are expanded. The cells may be fed 720 nutrients to promote their expansion. For example, media may be delivered into the cell growth chamber to provide the nutrients for expansion. The expansion of the cells 718 may also include adding reagents periodically to the cell growth chamber to continue to promote their expansion. For example, in some embodiments, reagents (e.g., cytokines) may be added to the cell growth chamber to promote the expansion of the cells. In one embodiment, the reagent may be additional IL-2 cytokine, e.g., recombinant human IL-2 cytokine.


Also as part of expanding the cells 718, the environment inside the cell growth chamber may be controlled. For example, gasses may be delivered and exchanged continuously to provide a balance of for example, carbon dioxide and oxygen to the cells expanding in the cell growth chamber. Additionally, the temperature may be controlled to be within a range optimized for cell expansion. Expansion of cells 718 may also include monitoring metabolites 726. For example, the lactate and glucose levels may be periodically monitored. A rise or fall in the metabolites may prompt changes (e.g., additional feeding, additional reagent additions, additional gas exchange, etc.) to control the environment 724 in the cell growth chamber.


Process 700 next proceeds to harvest the cells 728. Further processing of the removed cells or other analysis may optionally be performed at step 730. For example, the cells may be characterized to determine cell phenotype(s). The further processing or other analysis 730 may include performing flow cytometry, for example, to characterize cell phenotypes. Process 700 may then terminate at END operation 732. If it is not desired to perform further processing/analysis, process 700 terminates at END operation 732.



FIG. 8A illustrates operational steps 800 of a process, which may be used to position cells or other material (e.g., proteins, nutrients, growth factors) into a cell growth chamber according to embodiments of the present disclosure. In embodiments, the process 800 may be implemented as part of a “load cells centrally without circulation” task. Start operation 802 is initiated and process 800 proceeds to step 804, where a first volume of fluid with cells may be loaded into a cell growth chamber of a cell expansion system. In embodiments, the cells may comprise non-adherent cells, such as one or more types of T cells. In one embodiment, the plurality of cells comprises Tregs. As may be appreciated, loading of the first volume of fluid with cells may be performed by components of a cell expansion system such as systems CES 500 (e.g., FIG. 5A) and CES 600 (FIG. 6), described above. FIG. 8B illustrates a portion of a cell expansion system that includes a first inlet pump 840, a first fluid flow path 860, a first fluid circulation pump 848, a first fluid circulation path 852, a cell growth chamber 844, and a second fluid circulation path 854. The first fluid flow path 860 is fluidly associated with fluid circulation path 852 through connection 860B. Embodiments may provide for the first volume of fluid with the cells to be loaded 804 through the first fluid flow path 860 utilizing the first fluid inlet pump 840 and into the first fluid circulation path 852. In embodiments, the first volume of fluid is loaded without activating the first fluid circulation pump 848.


As illustrated in FIG. 8B, a volume of the first fluid circulation path 852 may be comprised of a number of volumes of its portions. For example, a first portion of the volume may be an intracapillary space (when the cell growth chamber is a hollow fiber membrane bioreactor) of the cell growth chamber 844. A second portion of the volume may be from connection 860B to an inlet port 844A of the cell growth chamber 844. A third portion may be from the connection 860B to an outlet port 844A of the cell growth chamber 844.


Process 800 proceeds to loading of a second volume of fluid 806. The second volume of fluid may comprise media and may be introduced into a portion of the first fluid flow path 860. In embodiments, the second volume may be a predetermined amount selected in order to position 808 the first volume into a first portion of the cell growth chamber 844. In embodiments, the first volume of fluid and the second volume of fluid may be the same. In other embodiments, the first volume of fluid and the second volume of fluid may be different. In yet other embodiments, a sum of the first volume of fluid and the second volume of fluid may be equal to a percentage of a volume of the first fluid circulation path, e.g., path 852 (FIG. 8B).


In order to position the first volume of fluid, the second volume of fluid has to be enough to push the first volume into the desired position in the cell growth chamber 844. The second volume of fluid may therefore, in embodiments, be about as large as the volume of the first fluid circulation path 852 between connection 860B and the inlet port 844A. As may be appreciated, this would push the first volume of fluid with the cells into a position within the cell growth chamber 844.


In other embodiments, the first volume of fluid (with cells) may be positioned 808 about a central region 866 of the cell growth chamber 844. In these embodiments, the second volume may be about as large a sum of the volume of the first fluid circulation path 852, between connection 860B and the inlet port 844A and the volume of the first fluid circulation path made up by the cell growth chamber 844 (e.g., the volume of the intracapillary space) that would not be occupied by the first volume of fluid when positioned in the cell growth chamber. For example, in one embodiment, the first volume of fluid (with the cells) may be 50 ml. The cell growth chamber may have a volume of, for example, 124 ml. When the first volume is positioned around the central region 866, it will occupy the 50 ml around the central region 866, leaving 74 ml, which is on either side of the central region 866. Accordingly, 50% of 74 ml (or 37 ml) may be added to the volume between connection 860 and inlet port 844A to position the 50 ml of the first volume around the central region 866.


Positioning 808 the first volume may in embodiments, involve adding additional volumes of fluid to position the first volume with cells in the cell growth chamber. For example, if the desired position of the first volume is not achieved with the second volume, additional fluid may be added to position the first volume 808.


Process 800 proceeds to the query 810 to determine whether the first volume should be repositioned. For example, in embodiments, the first volume may be positioned closer to inlet port 844A. If it is desired to move the first volume closer to central region 866 of cell growth chamber 844, process 800 may loop back to step 808 where additional fluid may be added to the first fluid circulation path to position the first volume of fluid.


If it is determined at query 810 that the first volume does not need to be repositioned, process 800 proceeds to feeding of the cells 812. In embodiments, the cells may be fed using a media that includes a number of compounds such as glucose, proteins, growth factors, reagents, or other nutrients. In embodiments, feeding of the cells 812 may involve activating inlet pumps and circulation pumps (e.g., pumps 840 and 848) to deliver media with nutrients to the cells in the cell growth chamber 844. Some embodiments provide for the cells to be maintained in the cell growth chamber 844 during step 812, as described below. These embodiments may involve activating pumps, e.g., inlet pumps and circulation pumps (e.g., pumps 840 and 848, as described below) so that flow of fluid into the cell growth chamber 844 may be from both directions such as from the inlet port 844A and the outlet port 844B into the cell growth chamber 844.


Process 800 then proceeds to the expansion of the cells 814 where the cells may be expanded or grown. While step 814 is shown after step 812, step 814 may occur before, or simultaneous with, step 812, according to embodiments. Cells may then be removed 816 from the cell growth chamber and collected in a storage container. In embodiments, step 816 may involve a number of sub-steps. For example, the cells may be circulated by the circulation pump (e.g., pump 848) before they are collected and stored in a container. Process 800 terminates at END operation 830.


Next, FIG. 9A illustrates example operational steps 900 of a process for retaining cells in a bioreactor of a cell expansion system, such as CES 500 (e.g., FIGS. 5B & 5C), in accordance with embodiments of the present disclosure. As discussed above, cells residing in the headers of the bioreactor or in the IC loop outside of the bioreactor may not receive proper gas exchange and nutrient exchange, which may result in cell aggregation and death. In an embodiment, the bioreactor provides gas exchange and nutrient exchange through the semi-permeable hollow fiber membrane. It can be important that such exchange is efficient as the surface area to volume ratio in a cell expansion system comprising a hollow fiber membrane may be significantly larger than that of other cell culturing methods (e.g., about 15 times that of cell culture flasks, for example). Such efficiency may be accomplished by minimizing the diffusion distance for media constituents able to pass the membrane surface where exchange takes place.


For example, FIG. 9B illustrates a graph 936 of the oxygen consumption demands placed on a cell expansion system, such as the Quantum® Cell Expansion System during cell proliferation (approximately 3E+09 T-cells present in the bioreactor). FIG. 9B depicts the percentage (%) of oxygen (O2) at the bioreactor outlet 938. For example, a sensor to measure oxygen levels may be placed at the EC outlet port of the bioreactor, according to an embodiment. In another embodiment, a sensor to measure oxygenation may be placed at the IC outlet port of the bioreactor. The percentage of O2 is measured against the Run Time (e.g., in minutes) 940. To maximize the oxygen supply to the cells, the EC circulation flow rate, QEC Circ may be set to 300 mL/min (942), according to an embodiment. FIG. 9B shows a change in oxygenation when the EC circulation rate is dropped from 300 mL/min (942) to 50 mL/min (944). For example, the cells may consume oxygen (O2) in the media as the media travels across the bioreactor. Fluid at 50 mL/min (944) moves more slowly across the bioreactor than fluid at 300 mL/min (942), so there may be a longer time period or greater opportunity for the cells to strip the media of oxygen when the EC circulation rate is at 50 mL/min. The oxygenation then recovers when the EC circulation rate is taken back up to 300 mL/min (946). FIG. 9B shows a possible benefit of keeping the cells in the fibers themselves where gas transfer takes place, as opposed to in the portion of the IC circulation path outside of the bioreactor, for example, where the cells may be deprived of oxygen. It may therefore be beneficial to retain cell, e.g., non-adherent cell, populations inside the hollow fibers of the bioreactor during feeding by directing the flow of media to enter both sides of the bioreactor, e.g., the IC inlet port and the IC outlet port. In an embodiment, an equal distribution of flow to the IC inlet port and the IC outlet port may be used. In another embodiment, a larger flow to the IC inlet port as compared to the IC outlet port may be used and vice versa, depending on where it is desired to locate the cells in the bioreactor, for example.


Returning to FIG. 9A, START operation 902 is initiated and process 900 proceeds to load a disposable set or premounted fluid conveyance assembly (e.g., 210 or 400) onto a cell expansion system 904. The disposable set may then be primed 906, in which the set may be primed 906 with Lonza Ca2+/Mg2+-free PBS, for example. In preparation for the loading and seeding of cells, the priming fluid may be exchanged using an IC/EC washout 908. In an embodiment, the PBS in the system may be exchanged for TexMACS GMP Base Medium, for example. Next, process 900 proceeds to close the IC outlet valve 910. In embodiments, the EC outlet valve may be open to allow for ultrafiltration of fluid added to the hollow fibers of a bioreactor comprising a hollow fiber membrane. The media may next be conditioned 912. Next process 900 proceeds to load cells 914, e.g., suspension or non-adherent cells, such as T cells or Tregs. In an embodiment, such cells may be loaded 914 by a “load cells centrally without circulation” task. In another embodiment, such cells may be loaded 914 by a “load cells with uniform suspension” task. In other embodiments, other loading tasks and/or loading procedures may be used.


In an embodiment, the cells being loaded 914 may be suspended in a solution comprising media to feed the cells during and after such loading, for example. In another embodiment, such solution may comprise both media for feeding the cells and a soluble activator complex to stimulate the cells, e.g., T cells. Such loading 914 may occur on Day 0, for example.


Following the loading of cells 914, the cells may be further fed 916. During such feeding 916, it may be desired to control the cell residence in the bioreactor itself. Through the adjustment of flow control parameters 918, the cells may be retained in the bioreactor itself instead of losing cells from the bioreactor into the portion(s) of the IC loop outside of the bioreactor, for example, during the expansion phase of growth. By retaining the cells in the bioreactor, cells in the bioreactor may be closer to the IC inlet port, in which such cells may receive the freshest growth media, according to embodiments. On the other hand, cells in the IC loop may be receiving expended or conditioned media which may affect their glycolytic metabolism, for example. In addition, cells in the bioreactor may receive mixed gas (e.g., oxygen, carbon dioxide, and nitrogen) input from a gas transfer module (GTM) by diffusion from the EC loop circulation, whereas cells in other portions of the IC loop may not receive such mixed gas, according to an embodiment. It should be noted that while embodiments may provide for retaining the cells in the bioreactor itself, other embodiments may provide for maintaining the cells in any location allowing for improved nutrient delivery and/or gas exchange. Embodiments thus provide for the use of other locations for retaining cells, or controlling the residence of cells, without departing from the spirit and scope of the present disclosure.


Returning to FIG. 9A and process 900, the loss of cells from the hollow fiber membrane bioreactor may be reduced by matching, or closely or substantially matching, the IC circulation pump rate to the IC inlet pump rate, but in the opposite direction, in accordance with embodiments. The IC inlet pump 920 may be adjusted to produce a first flow rate or volumetric flow rate, and the IC circulation pump may be adjusted 922 to produce a second counter-flow rate or second counter-volumetric flow rate, in which a volumetric flow rate or fluid flow rate or rate of fluid flow or flow rate may be considered as the volume of fluid which passes per unit time (may be represented by the symbol “Q”). For example, an IC inlet pump rate of 0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture, which may be Days 4-7, for example, in embodiments. Such pump adjustment 918 may allow for counteracting any forces associated with a loss of cells from the IC outlet port of the bioreactor, for example.


Next, the cells may be allowed to grow or expand 924. The cells are not limited to growing or expanding at step 924, but, instead, the cells may also expand during step(s) 914, 916, 918, 920, 922, for example. Process 900 may next proceed to harvest operation 926, in which the cells may be transferred to a harvest bag(s) or container(s). The disposable set may then be unloaded 932 from the cell expansion system, and process 900 then terminates at END operation 934.


Alternatively, from harvest operation 926, process 900 may optionally proceed to allow for further processing/analysis 928. Such further processing 928 may include characterization of the phenotype(s), for example, of the harvested cells. From optional further processing/analysis step 928, process 900 may proceed to optionally reload any remaining cells 930. Process 900 may then proceed to unload the disposable set 932, and process 900 may then terminate at END operation 934. Alternatively, process 900 may proceed from further processing/analysis step 928 to unload disposable set 932. Process 900 may then terminate at END operation 934.


Next, FIG. 10A illustrates example operational steps 1000 of a process for feeding cells that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B &5C), in accordance with embodiments of the present disclosure. START operation 1002 is initiated, and process 1000 proceeds to load a disposable set onto the cell expansion system, prime the set, perform an IC/EC washout, condition media, and load cells, e.g., suspension or non-adherent cells, for example. Next, process 1000 proceeds to feed the cells during a first time period 1004. In embodiments, a first inlet flow rate and a first circulation flow rate may be used. As an example, a first IC inlet flow rate and a first IC circulation flow rate may be used, in which the first IC inlet flow rate may be controlled by the IC inlet pump, e.g., first pump, and the first IC circulation flow rate may be controlled by the IC circulation pump, e.g., second pump. In an example embodiment, the IC inlet pump (554) may cause a volumetric flow rate of 0.1 mL/min to enter the IC inlet port (501A) of the bioreactor (501) with the IC circulation pump (512) causing a complementary IC circulation volumetric flow rate or fluid flow rate of −0.1 mL/min to enter the IC outlet port (501B) of the bioreactor (501), in which the negative symbol (“−”) used in −0.1 mL/min, for example, indicates a direction of the IC circulation pump (512) to cause or produce a counter-flow rate to maintain cells in the bioreactor during the growth phase of the cell culture.


During the feeding of the cells and the use of the IC pumps during feeding to control cell residence in the bioreactor through flow and counter-flow properties, the cells continue to grow and expand. As a result, the cells may demand additional media, e.g., glucose and/or cell growth formulated media, to support the expanding population. Efforts may also be made to control lactate values of the expanding cell population. In embodiments, cell culture lactate values may be maintained at or below about 20 mmol/L, at or below about 15 mmol/L, at or below about 10 mmol/L, or even at or below about 7 mmol/L. In other embodiments, rate(s) of media addition, for example, and/or other settings may be controlled to attempt to maintain the lactate levels≤about 5 mmol/L, for example, to improve cell growth and viability. Other concentrations may be used in other embodiments.


In an example embodiment, an effort may be made to control lactate values at about 7 mmol/L by concurrently increasing both the IC inlet (+) pump rate and IC circulation (−) pump rate from ±0.1 to ±0.4 mL/min within the lumen of the hollow fiber membrane over multiple time periods, e.g., days (Days 4-8), according to embodiments. For example, FIG. 10B provides a table 1018 of example IC pump rates for feeding using a “feed cells” task, for example, with a cell expansion system (e.g., CES 500). Table 1018 provides example time periods 1020, e.g., Days, versus example IC pump rates 1022 to produce volumetric flow rates to both sides of the bioreactor to maintain cells in the bioreactor. For example, Days 0-4 (1024) may use an IC inlet or input pump rate of 0.1 mL/min (1026) and an IC circulation pump rate of −0.1 mL/min (1028); Day 5 (1030) may use an IC inlet pump rate of 0.2 mL/min (1032) and an IC circulation pump rate of −0.2 mL/min (1034); Day 6 (1036) may use an IC inlet pump rate of 0.3 mL/min (1038) and an IC circulation pump rate of −0.3 mL/min (1040); and Day 7 (1042) may use an IC inlet pump rate of 0.4 mL/min (1044) and an IC circulation pump rate of −0.4 mL/min (1046). While table 1018 of FIG. 10B provides example pump rates of ±0.1 to ±0.4 mL/min for feeding the cells while retaining the cells in the bioreactor during the growth phase of the cell culture, other pump rates and resulting flow rates may be used according to embodiments without departing from the spirit and scope of the present disclosure. For example, while increments of ±0.1 mL/min for increasing the feed flow rate are shown in this example, other increments, e.g., ±0.005 mL/min, ±0.05 mL/min, etc., may be used to increase the feed flow rate in embodiments. The time periods, e.g., Days, and pump rates in table 1018 of FIG. 10B are offered merely for illustrative purposes and are not intended to be limiting.


Returning to FIG. 10A, process 1000 proceeds from feeding the cells during a first time period 1004 to increasing the first inlet flow rate by a first amount to achieve a second inlet flow rate 1006. For example, as in the embodiment depicted in FIG. 10B as discussed above, the IC inlet pump rate (+) may increase from 0.1 mL/min to 0.2 mL/min to produce an IC inlet flow rate of 0.2 mL/min. Further, the IC circulation pump rate (−) may concurrently increase from −0.1 mL/min to −0.2 mL/min to produce an IC circulation flow rate of −0.2 mL/min. The first circulation flow rate is thus increased by the first amount to achieve the second circulation flow rate 1008. The cells may then be fed during a second time period at the second inlet flow rate and at the second circulation flow rate 1010 to maintain the cells in the bioreactor and outside of the headers and outside of the portion of the IC circulation path outside of the bioreactor, for example. Following the second time period of feeding 1010, process 1000 may terminate at END operation 1016 if it is not desired to continue feeding and/or expanding the cells, for example. Alternatively, process 1000 may optionally continue to increase, or otherwise change, the feeding flow rates 1012. There may be any number of feeding time periods, as represented by ellipsis 1014. Following the desired number of feeding time periods 1014, process 1000 then terminates at END operation 1016. While FIGS. 10A and 10B and process 1000 show “increases” in the flow rates, other adjustments to the flow rates may be made. For example, the flow rates may decrease or remain substantially the same from one feeding period to the next. Considerations, such as metabolic activity, for example, may determine how flow rates are adjusted. The “increases” in flow rates in FIGS. 10A and 10B are offered merely for illustrative purposes and are not intended to be limiting.


Turning to FIG. 11A, example operational steps 1100 of a process for feeding cells that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B & 5C), are provided in accordance with embodiments of the present disclosure. START operation is initiated 1102, and process 1100 proceeds to load a disposable set onto the cell expansion system, prime the set, perform an IC/EC washout, condition media, and load cells, e.g., suspension or non-adherent cells, for example. Next, process 1100 proceeds to feed the cells during a first time period 1104. In embodiments, a first inlet rate or flow rate and a first circulation rate or flow rate may be used. As an example, a first IC inlet flow rate and a first IC circulation flow rate may be used, in which the first IC inlet flow rate may be controlled by the IC inlet pump, e.g., first pump, and the first IC circulation flow rate may be controlled by the IC circulation pump, e.g., second pump. In an example embodiment, the IC inlet pump (554) may cause a volumetric flow rate or fluid flow rate of 0.1 mL/min to enter the IC inlet port (501A) of the bioreactor (501) with the IC circulation pump (512) causing a complementary IC circulation flow rate of −0.1 mL/min to enter the IC outlet port (501B) of the bioreactor (501), in which the negative symbol (“−”) used in −0.1 mL/min, for example, indicates a direction of the IC circulation pump (512) to cause or produce a counter-flow rate to maintain cells in the bioreactor during the growth phase of the cell culture.


During the feeding of the cells and the use of the IC pumps to control cell residence in the bioreactor through flow and counter-flow properties, the cells continue to grow and expand. As a result, the cells may demand additional media, e.g., glucose and/or cell growth formulated media, to support the expanding population. Efforts may also be made to control lactate values of the expanding cell population. In an example embodiment, an effort may be made to control lactate values at about 7 mmol/L, for example, by concurrently increasing both the IC inlet (+) pump rate and IC circulation (−) pump rate from ±0.1 to ±0.4 mL/min within the lumen of the hollow fiber membrane over multiple days, e.g., Days 4-8, according to embodiments. For example, see FIG. 10B, table 1018, and the discussion above, for example pump rates for feeding. As noted, while table 1018 of FIG. 10B provides pump rates of ±0.1 to ±0.4 mL/min for feeding the cells while retaining the cells in the bioreactor during the growth phase of cell culturing, other pump rates and resulting flow rates may be used according to embodiments without departing from the spirit and scope of the present disclosure. The time periods, e.g., Days, and pump rates in table 1018 of FIG. 10B are offered merely for illustrative purposes and are not intended to be limiting.


Returning to FIG. 11A, process 1100 proceeds from feeding the cells during a first time period 1104 to increasing the first inlet rate or flow rate by a first amount to achieve a second inlet rate or flow rate 1106. For example, as in the embodiment depicted in FIG. 10B as discussed above, the IC inlet pump rate (+) may increase from 0.1 mL/min to 0.2 mL/min to produce an IC inlet flow rate of 0.2 mL/min. Further, the IC circulation pump rate (−) may concurrently increase from −0.1 mL/min to −0.2 mL/min to produce an IC circulation flow rate of −0.2 mL/min. The first circulation rate or flow rate is thus increased by the first amount to achieve the second circulation rate or flow rate 1108. The cells may then be fed during a second time period at the second inlet rate or flow rate and at the second circulation rate or flow rate 1110 to maintain the cells in the bioreactor and outside of the headers or the portion of the IC circulation path outside of the bioreactor, for example. Following the second period of feeding 1110, process 1100 may terminate at END operation 1116 if it is not desired to continue feeding and/or expanding the cells, for example.


Alternatively, process 1100 may optionally determine whether to adjust the feeding rates or flow rates based on metabolic activity, in which process 1100 proceeds to optional query 1112 to determine whether to adjust feeding based on metabolic levels. Monitoring the glucose and/or lactate levels can facilitate the adjustment of cell expansion system media flow rates, e.g., IC media flow rate, to support cell, e.g., Treg, expansion in a bioreactor, such as a hollow fiber bioreactor, for example.


As shown in FIGS. 11B and 11C, embodiments provide for controlling the lactate values of cell expansion runs or procedures involving the expansion of hTregs, for example. Graphs 1118 and 1132 indicate that measurements of glucose and lactate levels may be used to adjust cell expansion system media flow rates, e.g., IC media flow rates, to support the expansion of Tregs, for example. For example, FIG. 11B provides a graph 1118 showing the metabolisms of expanding hTregs, in which such cell expansion may occur in a cell expansion system, such as the Quantum® Cell Expansion System. The glucose concentration (mg/dL) 1120 and lactate concentration (mmol/L) 1122 are shown for various cell expansion runs 1124 and 1125 across periods of time, e.g., Days, 1126. Throughout these Treg runs 1124 and 1125, an effort may be made to control the lactate values of the expanding cell population to values about 7 mmol/L by concurrently increasing both the IC inlet (+) pump rate and IC circulation pump rate from ±0.1 to ±0.4 mL/min, for example, within the lumen of the hollow fiber membrane over Days 4-8, for example. In other embodiments, other pump rates may be used. As shown, the lowest glucose levels during the Treg cell expansions may range from a concentration 1128 of 264 mg/dL on Day 7 (Q1584) to a concentration 1130 of 279 mg/dL on Day 8 (Q1558), according to the embodiments shown. As depicted, the base glucose concentration, in the cell growth formulated medium for runs 1124 may range from 325 mg/dL to 335 mg/dL, for example. In other embodiments, it may be desired to maintain the lactate levels≤about 5 mmol/L to improve cell growth and viability. In embodiments, graphical user interface (GUI) elements may be used to control the rate of media addition and to maintain the lactate metabolic waste product from glycolysis below a defined level during the expansion of cells.


Turning to FIG. 11C, graph 1132 shows the metabolisms of expanding hTregs, in which such cell expansion may occur in a cell expansion system, such as the Quantum® Cell Expansion System. The glucose consumption (mmol/day) 1134 and lactate generation (mmol/day) 1136 are shown for various cell expansion runs 1138 and 1139 across periods of time 1140, e.g., Days. To control lactate values at about 7 mmol/L, for example, concurrent increases may be made to the IC inlet (+) pump rate and IC circulation (−) pump rates from ±0.1 to ±0.4 mL/min, in an embodiment. For example, graph 1132 shows IC circulation and IC feed rates of ±0.1 mL/min (1142); ±0.2 mL/min (1144); ±0.3 mL/min (1146); and ±0.4 mL/min (1148). Other embodiments may use other flow rates. The flow rates used and shown in FIGS. 11B and 11C are offered for purposes of illustration and are not intended to be limiting. It is noted that while positive (+) may be shown for the direction of the IC inlet pump and negative (−) may be shown for the direction of the IC circulation pump, such directions are offered for illustrative purposes only, in which such directions depend on the configurations of the pumps used.


Returning to FIG. 11A and optional query 1112, if it is not desired to measure metabolic activity and/or adjust feeding levels based on such measurement(s), process 1100 proceeds “no” to END operation 1116, and process 1100 terminates. Alternatively, where it is desired to adjust feeding levels based on metabolic activity, process 1100 proceeds “yes” to optional step 1114 to continue to increase the rate of media addition and feed the expanding cell population. While step 1114 is shown as one step, this step may involve numerous adjustments to the media addition rate, such as increasing the IC inlet flow rate and increasing the IC circulation flow rate, for example. Step 1114 is shown as one step only for illustrative purposes and is not intended to be limiting. Following any adjustments to the rate of media addition, process 1100 proceeds to optional query 1112 to determine whether to continue measuring metabolic levels and/or adjust feeding. If it is not desired to continue measuring metabolic activity and/or adjusting feeding levels based on metabolic activity, process 1100 proceeds “no” to END operation 1116, and process 1100 terminates. While FIG. 11A and process 1100 show “increases” in the rates or flow rates, other adjustments to the flow rates may be made. For example, the rates or flow rates may decrease or remain substantially the same from one feeding period to the next. The type of adjustments which may be made may depend on the metabolic activity assessment of the growing cell population. The “increases” in flow rate in FIG. 11A, for example, are offered merely for illustrative purposes and are not intended to be limiting.


Next, FIG. 12 illustrates example operational steps 1200 of a process for retaining cells in a location during feeding using a cell expansion system, such as CES 500 (e.g., FIGS. 5B &5C), in accordance with embodiments of the present disclosure. START operation 1202 is initiated, and process 1200 proceeds to load a disposable set onto the cell expansion system, prime the set, perform IC/EC washout, condition media, and load cells, e.g., suspension or non-adherent cells, for example. Next, process 1200 proceeds to feed the cells during a first time period 1204. In embodiments, a first inlet flow rate and a first circulation flow rate may be used. As an example, a first IC inlet flow rate and a first IC circulation flow rate may be used, in which the first IC inlet flow rate may be produced and controlled by the IC inlet pump, e.g., first pump, and the first IC circulation flow rate may be produced and controlled by the IC circulation pump, e.g., second pump. In an example embodiment, the first IC inlet pump (554) may cause a volumetric flow rate of 0.1 mL/min to enter the IC inlet port (501A) of the bioreactor with the IC circulation pump (512) causing a complementary IC circulation volumetric flow rate or fluid flow rate of −0.1 mL/min to enter the IC outlet port (501B) of the bioreactor (501), in which the negative symbol (“−”) used in −0.1 mL/min, for example, indicates a direction of the IC circulation pump to cause or produce a counter-flow rate to maintain cells in the bioreactor during the growth phase of the cell culture. In another embodiment, the first IC circulation flow rate may be a percentage of the first IC inlet flow rate. For example, the first IC circulation flow rate may be about fifty percent (50%) or about one-half (%), or another percentage or portion according to embodiments, of the first IC inlet flow rate.


During the feeding (and expansion) of the cells and the use of the IC pumps, for example, to control cell residence in the bioreactor through flow and counter-flow properties, the cells continue to grow and expand. As a result, the cells may demand additional media, e.g., glucose and/or cell growth formulated media, to support the expanding population. In embodiments, efforts may be made to increase the rate of media addition to feed the expanding cell population. In an example embodiment, an increase in the IC inlet pump rate (+) may cause the IC inlet flow rate to increase by a first amount to achieve a second IC inlet flow rate 1206. For example, the IC inlet flow rate may increase by a first amount of 0.1 mL/min to achieve a second IC inlet flow rate of 0.2 mL/min, according to an embodiment. Other adjustments may be made to the IC inlet flow rate according to other embodiments.


Next, the second IC circulation flow rate may be set, or adjusted or configured, to equal a percentage, or portion, or fraction of the second IC inlet flow rate 1208, according to embodiments. For example, the second IC circulation flow rate may be set, or adjusted or configured, to equal about fifty percent (50%) or about one-half (½), or another percentage or portion according to embodiments, of the value of the IC inlet flow rate, according to an embodiment. Depending on the value of the first IC circulation flow rate, an adjustment to the IC circulation pump rate (−) may cause the second IC circulation flow rate to increase, according to an embodiment. In another embodiment, an adjustment may be made to the IC circulation pump rate to produce or cause the second IC circulation flow rate to decrease such that the IC circulation flow rate may be substantially equal to a pre-defined percentage or pre-defined fraction of the second IC inlet flow rate. In yet another embodiment, no adjustment may be made to the IC circulation pump rate. For example, where the first IC inlet flow rate equals 0.1 mL/min, and the first IC circulation flow rate equals −0.1 mL/min, if the second IC inlet flow rate is increased to 0.2 mL/min, the second IC circulation flow rate may be set to (−½)*(second QIC Inlet) (where QIC Inlet is the IC inlet flow rate) or (−½)*(0.2 mL/min), which provides for a second QIC Circ (where QIC Circ is the IC circulation flow rate) of −0.1 mL/min, and no adjustment to the IC circulation pump rate may be made to achieve such second QIC Circ.


Turning to FIGS. 5B and 5C, such figures depict operational configurations of cell expansion system 500 showing fluid movement in the first circulation path, in accordance with embodiments of the present disclosure. The configurations of FIGS. 5B and 5C show a split, for example, in the flow rate to the IC inlet port, e.g., first port, and to the IC outlet port, e.g., second port, to keep the cells in the cell growth chamber or bioreactor. As discussed with respect to CES 500 above, in the IC loop or first circulation path 502, fluid may be initially advanced by the IC inlet pump 554. Such fluid may be advanced in a first direction, such as a positive direction, for example. Fluid may flow into cell growth chamber or bioreactor 501 through IC inlet port 501A, through hollow fibers in cell growth chamber or bioreactor 501, and may exit via IC outlet port 501B. Media may flow through IC circulation pump 512 which may be used to control the rate of media flow. IC circulation pump may pump the fluid in a first direction or second direction opposite the first direction. Exit port 501B may be used as an inlet in the reverse direction, for example. In an embodiment, the IC circulation pump 512 may pump the fluid in a direction opposite the direction of the IC inlet pump, for example. As an example, the direction of the IC inlet pump may be positive (+), and the direction of the IC circulation pump may be negative (−) to cause a counter-flow such that fluid may enter both sides of the bioreactor to keep the cells in the bioreactor.


In an embodiment, a first portion of fluid may branch at connection 517 to flow into the IC inlet port (501A) of the bioreactor 501. In an embodiment, the IC circulation pump 512 may operate at a pump rate that may be matched, or closely or substantially be matched, to the IC inlet pump 554 rate, but in the opposite direction, such that a second portion of fluid may branch at connection 517 to flow into the IC outlet port (501B) of the bioreactor 501. For example, an IC inlet pump rate of +0.1 mL/min may be matched, or closely or substantially be matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture. Such pump adjustment tactic during the feeding may counteract forces associated with the loss of cells from the IC outlet port. Such type of feeding using pump adjustments to cause flow and counter-flow into the IC inlet port (501A) and IC outlet port (501B), respectively, of the bioreactor may be referred to as a modified feeding method, according to an embodiment. In another example embodiment, the IC circulation pump rate may be adjusted to cause a flow rate into the IC outlet port (501B) that may be equal to about fifty percent (50%) or about one-half (½), or another percentage or fraction in other embodiments, of the IC inlet flow rate, but in the opposite direction. For example, with an IC inlet pump rate of 0.4 mL/min, an IC circulation pump rate may be set, or configured or adjusted, to about −0.2 mL/min. Other percentages or portions may be used in other embodiments.



FIGS. 5B and 5C illustrate operational configurations showing fluid movement in CES 500, in which the flow and counter-flow rates to keep the cells in the cell growth chamber or bioreactor 501 are shown, according to embodiments. For example, such flow rates are illustrated in FIG. 5B as “X” flow rate 503; “(−½) X” flow rate 505 (where the negative symbol (“−”) is an indication of direction, in which a direction of flow rate 505 is shown by the directional arrow in FIG. 5B); and (½) X″ flow rate 507, in which about ½, or a first portion, of the flow rate branches at connection 517 to enter IC inlet port (501A) of bioreactor 501, and about ½, or a second portion, of the flow rate branches at connection 517 to enter the IC outlet port (501B) of bioreactor 501, according to embodiments. As shown, the sum of the first portion and the second portion may be substantially equal to the total flow rate 503, in which the total flow rate 503 is pumped by IC inlet pump 554. Depending on the flow and counter-flow rates which may be used to keep the cells in the bioreactor or cell growth chamber 501, other types of fractions or percentages of the IC inlet pump rate may be used to set, or configure or adjust, the IC circulation pump. As such, FIG. 5C illustrates such flow rates as “X” flow rate 511; “−(y %)*X” flow rate 513 (where the negative symbol (“−”) is an indication of direction, in which a direction of flow rate 513 is shown by the directional arrow in FIG. 5C); and “(100%−y %)*X” flow rate 515, where “y” equals a numeric percentage, according to embodiments. In embodiments, the sum of flow rate 513 and flow rate 515 is substantially equal to flow rate 511.


In an embodiment, all, or substantially all, of the flow from the first fluid flow path 506 may flow to IC inlet port (501A) of bioreactor 501 from connection 517, for example. In another embodiment, all, or substantially all, of the flow from the first fluid flow path 506 may flow to IC outlet port (501B) of bioreactor 501 from connection 517. In yet another embodiment, a first portion of the flow from first fluid flow path 506 may flow from connection 517 to IC inlet port (501A), and a second portion of the flow from the first fluid flow path 506 may flow from connection 517 to IC outlet port (501B). In embodiments, the percentage of the IC inlet flow rate at which the IC circulation flow rate may be set may range from about 0 percent to about 100 percent. In other embodiments, the percentage may be between about 25 percent and about 75 percent. In other embodiments, the percentage may be between about 40 percent and about 60 percent. In other embodiments, the percentage may be between about 45 percent and about 55 percent. In embodiments, the percentage may be about 50 percent. It is to be understood that the operational configurations shown in FIGS. 5B and 5C represent possible configurations for various operations of the cell expansion system, and modifications to the configurations shown are within the scope of the one or more present embodiments.


Returning to FIG. 12, the cells may be fed (and continue expanding) during a second time period at the second inlet flow rate and at the second circulation flow rate 1210 to maintain the cells in the bioreactor and outside of the headers or portion of the IC circulation path outside of the bioreactor, for example. Following the second period of feeding 1210, process 1200 may terminate at END operation 1216 if it is not desired to continue feeding and/or expanding the cells, for example. Alternatively, process 1200 may optionally continue to increase, or otherwise change, the feeding flow rates 1212. There may be any number of feeding time periods, as represented by ellipsis 1214. Following the desired number of feeding time periods 1214, process 1200 may then terminate at END operation 1216. While FIG. 12 and process 1200 show “increases” in the flow rates, other adjustments to the flow rates may be made. For example, the flow rates may decrease or remain substantially the same from one feeding period to the next. Considerations, such as metabolic activity, for example, may determine how flow rates are adjusted. The “increases” in flow rates in FIG. 12 are offered merely for illustrative purposes and are not intended to be limiting.


Turning to FIG. 13, example operational steps 1300 of a process for feeding cells while retaining the cells in a first location, e.g., in the bioreactor, that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B & 5C), are provided in accordance with embodiments of the present disclosure. START operation 1302 is initiated, and process 1300 proceeds to load a disposable set onto the cell expansion system, prime the set, perform IC/EC washout, condition media, and load cells, e.g., suspension or non-adherent cells, for example. Next, process 1300 proceeds to feed the cells during a first time period 1304. In embodiments, a first inlet flow rate and a first circulation flow rate may be used. As an example, a first IC inlet flow rate and a first IC circulation flow rate may be used, in which the first IC inlet flow rate may be produced and controlled by the IC inlet pump, e.g., first pump, and the first IC circulation flow rate may be produced and controlled by the IC circulation pump, e.g., second pump. In an example embodiment, the first IC inlet pump rate may cause a volumetric flow rate of 0.1 mL/min to enter the IC inlet port of the bioreactor with a complementary IC circulation pump rate of −0.1 mL/min causing a volumetric flow rate of −0.1 mL/min to enter the IC outlet port of the bioreactor, in which the negative symbol (“−”) used in −0.1 mL/min, for example, indicates a direction of the IC circulation pump (512) to cause or produce a counter-flow rate to maintain cells in the bioreactor during the growth phase of the cell culture. In another embodiment, the first IC circulation flow rate may be a percentage or fraction or portion of the first IC inlet flow rate. For example, the first IC circulation flow rate may be about fifty percent (50%) or about one-half (½), or another percentage or portion according to embodiments, of the IC inlet flow rate.


During the feeding (and expansion) of the cells and the use of the IC pumps to control cell residence in the bioreactor through flow and counter-flow properties, the cells continue to grow and expand. As a result, the cells may demand additional media, e.g., glucose and/or cell growth formulated media, to support the expanding population. In embodiments, efforts may be made to increase the rate of media addition to feed the expanding cell population. In an example embodiment, an increase in the IC inlet pump rate (+) may cause the IC inlet flow rate to increase by a first amount to achieve a second IC inlet flow rate 1306. For example, the IC inlet flow rate may be increased by a first amount of 0.1 mL/min to achieve a second IC inlet flow rate of 0.2 mL/min, according to an embodiment. Other adjustments may be made to the IC inlet flow rate according to other embodiments.


Next, the second IC circulation flow rate may be set, or configured or adjusted, to equal a percentage or portion or fraction of the second IC inlet flow rate 1308, according to embodiments. For example, the second IC circulation flow rate may be set, or configured or adjusted, to equal about fifty percent (50%) or about one-half (½), or another percentage or portion according to embodiments, of the value of the IC inlet flow rate, according to an embodiment. In an embodiment, all, or substantially all, of the flow from the first fluid flow path 506 may flow to IC inlet port (501A) of bioreactor 501 from connection 517, for example. In another embodiment, all, or substantially all, of the flow from the first fluid flow path 506 may flow to IC outlet port (501B) of bioreactor 501 from connection 517. In yet another embodiment, a first portion of the flow from first fluid flow path 506 may flow from connection 517 to IC inlet port (501A), and a second portion of the flow from the first fluid flow path 506 may flow from connection 517 to IC outlet port (501B). In embodiments, the percentage of the IC inlet flow rate at which the IC circulation flow rate may be set may range from about 0 percent to about 100 percent. In other embodiments, the percentage may be between about 25 percent and about 75 percent. In other embodiments, the percentage may be between about 40 percent and about 60 percent. In other embodiments, the percentage may be between about 45 percent and about 55 percent. In embodiments, the percentage may be about 50 percent.


Depending on the value of the first IC circulation flow rate, an adjustment to the IC circulation pump rate (−) may cause the second IC circulation flow rate to increase, according to an embodiment. In another embodiment, an adjustment may be made to the IC circulation pump rate to cause the second IC circulation flow rate to decrease such that the second IC circulation flow rate may be substantially equals to a pre-defined percentage or pre-defined fraction of the second IC inlet flow rate. In yet another embodiment, no adjustment may be made to the IC circulation pump rate. For example, where the first IC inlet flow rate equals 0.1 mL/min, and the first IC circulation flow rate equals −0.1 mL/min, if the second IC inlet flow rate is increased to 0.2 mL/min, the second IC circulation flow rate may be set to (−½)*(second QIC Inlet) or (−½)*(0.2 mL/min), which provides for a second QIC Circ of −0.1 mL/min, and no adjustment to the IC circulation pump rate may be made to achieve such second QIC Circ, according to an embodiment.


The cells may then be fed (and continue expanding) during a second time period at the second IC inlet flow rate and at the second IC circulation flow rate 1310 to maintain the cells in the bioreactor and outside of the headers or portion of the IC circulation path outside of the bioreactor, for example. Following the second time period of feeding 1310, process 1300 may terminate at END operation 1316 if it is not desired to continue feeding and/or expanding the cells, for example.


In embodiments, a first time period, a second time period, a third time period, a fourth time period, a fifth time period, etc. may each comprise one or more days (and/or hours and/or minutes). For example, a time period may be one (1) to fourteen (14) days, according to embodiments. However, a time period may be less than one (1) day or greater than fourteen (14) days, in other embodiments. In an embodiment, a first time period for feeding may comprise Day 0, Day 1, Day 2, Day 3, Day 4; a second time period for feeding may comprise Day 5; a third time period for feeding may comprise Day 6; and a fourth time period for feeding may comprise Day 7, for example. In another embodiment, a first time period may comprise Day 0, Day 1, and Day 2 (e.g., duration of about 3 days); a second time period may comprise Day 3, Day 4, and Day 5 (e.g., duration of about 3 days); a third time period may comprise Day 6, Day 7, and Day 8 (e.g., duration of about 3 days); a fourth time period may comprise Day 9 and Day 10 (e.g., duration of about 2 days); and a fifth time period may comprise Day 11, Day 12, and Day 13 (e.g., duration of about 3 days). Time periods may be different durations according to embodiments. Each time period may be measured in days, hours, minutes, and/or portions thereof.


Returning to FIG. 13, process 1300 may optionally continue to optional query 1312 to determine whether to adjust feeding rates or flow rates based on metabolic activity or metabolic levels. For the expansion of Tregs, for example, it may be desired to control the lactate values of the expanding cell population to values about 7 mmol/L. Where it is desired to maintain the cell culture lactate values at or below about 7 mmol/L, for example, the rate of media addition may be controlled during the expansion of the cells, e.g., regulatory T cells. In other embodiments, it may be desired to maintain the lactate levels≤about 5 mmol/L to improve cell growth and viability. In embodiments, graphical user interface (GUI) elements may be used to control the rate of media addition and to maintain the lactate metabolic waste product from glycolysis below a pre-defined level during the expansion of cells.


At optional query 1312, if it is not desired to measure metabolic activity and/or adjust feeding levels based on such measurement(s), process 1300 proceeds “no” to END operation 1316, and process 1300 terminates. Alternatively, where it is desired to adjust feeding levels based on metabolic activity, process 1300 proceeds “yes” to optional step 1314 to continue to increase the rate of media addition and feed the expanding cell population. While step 1314 is shown as one step, this step may involve numerous adjustments to the media addition rate, such as increasing the IC inlet flow rate and increasing the IC circulation flow rate, for example. Step 1314 is shown as one step only for illustrative purposes and is not intended to be limiting. Following any adjustments to the rate of media addition, process 1300 returns to optional query 1312 to determine whether to continue to measure metabolic levels and/or adjust feeding. If it is not desired to adjust feeding levels based on metabolic activity, process 1300 proceeds “no” to END operation 1316, and process 1300 terminates. While FIG. 13 and process 1300 show “increases” in the rates or flow rates, other adjustments to the flow rates may be made. For example, the rates or flow rates may decrease or remain substantially the same from one feeding period to the next. The type of adjustments which may be made may depend on the metabolic activity assessment of the growing cell population. The “increases” in flow rate in FIG. 13 are offered merely for illustrative purposes and are not intended to be limiting.


Next, FIG. 14 illustrates example operational steps 1400 of a process for retaining cells during feeding that may be used with a cell expansion system, such as CES 500 (FIGS. 5B & 5C), in accordance with embodiments of the present disclosure. START operation 1402 is initiated, and process 1400 proceeds to load a disposable tubing set or premounted fluid conveyance assembly (e.g., 210 or 400) 1404 onto the cell expansion system. Next, the system may be primed 1406. In an embodiment, a user or operator, for example, may provide an instruction to the system to prime by selecting a task for priming, for example. In an embodiment, such task for priming may be a pre-programmed task, for example. Next, the IC/EC washout task may be performed 1408, in which fluid on the IC circulation loop and on the EC circulation loop may be replaced, for example. The replacement volume may be determined by the number of IC volumes and EC volumes exchanged, according to an embodiment.


Next, to maintain the proper or desired gas concentration across the fibers in the bioreactor membrane, the condition media task 1410 may be executed to allow the media to reach equilibrium with the provided gas supply before cells are loaded into the bioreactor. For example, contact between the media and the gas supply provided by the gas transfer module (GTM) or oxygenator may be provided by adjusting the EC circulation rate. The system may then be maintained in a proper or desired state until a user or operator, for example, is ready to load cells into the bioreactor. In an embodiment, the system may be conditioned with media, such as complete media, for example. Complete media may be any media source used for cell growth. In an embodiment, the system may be conditioned with serum-free media, for example. In an embodiment, the system may be conditioned with base media. Any type of media understood by those of skill in the art may be used.


Process 1400 next proceeds to loading cells 1412 into the bioreactor from a cell inlet bag, for example. In an embodiment, the cells in the cell inlet bag may be in solution with media to feed 1414 the cells, for example. In another embodiment, the cells in the cell inlet bag may be in solution both with media to feed 1414 the cells and with a soluble activator complex to stimulate the cells, e.g., T cells or Tregs. In an embodiment, the cells (and feed solution, in embodiments) may be loaded into the bioreactor from the cell inlet bag until the bag is empty. Cells (and feed solution, in embodiments) may be chased from the air removal chamber to the bioreactor. In an embodiment, a “load cells with uniform suspension” task may be executed to load the cells (and feed solution, in embodiments). In another embodiment, a “load cells centrally without circulation” task may be executed to load the cells (and feed solution, in embodiments) into a specific, e.g., central, region of the bioreactor. Other loading methods and/or loading tasks may be used in accordance with embodiments.


Next, process 1400 proceeds to query 1416 to determine whether to use a modified feeding method to retain the cells, e.g., non-adherent or suspension cells, such as T cells or Tregs, in the bioreactor, e.g., hollow fiber bioreactor. For example, it may be desired to locate the cells in the bioreactor itself and out of the headers of the bioreactor or the rest of the IC loop. If it is not desired to use a modified feeding method to retain the cells in the bioreactor itself, process 1400 proceeds “no” to expand the cells 1426, in which the cells may continue to grow/expand using the media that they were initially fed with in step 1414, for example.


On the other hand, if it is desired to retain the cells in the bioreactor itself, process 1400 proceeds “yes” to modified feed 1418, in which the cells may be fed by using a flow rate into the IC inlet port (501A) of the bioreactor (501) and a flow rate into the IC outlet port (501B) of the bioreactor (501) to keep the cells in the bioreactor. In so doing, an inlet volumetric flow rate or inlet flow rate may be introduced into the first fluid flow path (506) 1420. For example, an IC inlet flow rate may be introduced into the fluid flow path (506) 1420. The IC inlet pump (554), e.g., a first peristaltic pump (in an embodiment), may operate at a pre-defined number of revolutions per minute (RPMs) to cause a pre-defined IC inlet volumetric flow rate, or IC inlet flow rate, of fluid in the first fluid flow path (506) 1420. A processor(s) and/or controller(s) may direct or control the first pump and/or second pump, for example, to operate at a pre-defined number of RPMs, according to an embodiment. Depending on the speed and direction of the IC circulation pump (512), a modified first flow rate, or first portion of the IC inlet flow rate, may enter the IC inlet port (501A), or first port, of the bioreactor (501) 1422. A pump rate of a pump may depend on the diameter of the pump or configuration of the pump, e.g., peristaltic pump. Other types of pumps may also be used, in which the pump rate may depend on the configuration of the pump used. The IC circulation pump (512), e.g., a second peristaltic pump (in an embodiment), may operate at a pre-defined number of RPMs, and in a direction opposite a direction of the first pump, to cause or produce a pre-defined IC circulation flow rate, or second flow rate, or second portion of the IC inlet flow rate, to enter the IC outlet port (501B), or second port, of the bioreactor (501) 1424. For example, embodiments may provide for about one-half (about ½), or a first portion, of the IC inlet flow rate to branch at connection 517 to enter IC inlet port (501A) of bioreactor 501, and about one-half (or ½), or a second portion, of the IC inlet flow rate to branch at connection 517 to enter the IC outlet port (501B) of bioreactor 501. As shown, the sum of the first portion and the second portion may substantially equal the IC inlet flow rate 503 (e.g., FIG. 5B), in which the IC inlet flow rate 503 may be pumped by IC inlet pump 554. Depending on the flow and counter-flow rates which may be used to keep the cells in the bioreactor or cell growth chamber 501, other types of fractions or percentages of the IC inlet pump rate may be used to set, or configure or adjust, the IC circulation pump, according to embodiments.


After feeding the cells with such flow and counter-flow properties to keep the cells in the bioreactor, process 1400 proceeds to grow/expand the cells 1426. While the expansion of cells is shown at step 1426, the cells may also grow/expand during one or more other step(s), such as 1412, 1414, 1416, 1418, 1420, 1422, 1424, for example. From expand step 1426, process 1400 proceeds to harvest or remove the cells 1430. Process 1400 may then terminate at END operation 1432. If any other steps are desired before harvest, such as continuing with a second modified feed method or other type of feeding method, process 1400 proceeds to optional “Other” step 1428, according to embodiments. From optional step 1428, process 1400 proceeds to harvest or remove the cells from the bioreactor 1430, and process 1400 may then terminate at END operation 1432.


Turning to FIG. 15A, example operational steps 1500 of a process for feeding cells to retain the cells in a first location, e.g., in the bioreactor itself, that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B & 5C), are provided in accordance with embodiments of the present disclosure. START operation 1502 is initiated, and process 1500 proceeds to load the disposable tubing set or premounted fluid conveyance assembly (e.g., 210 or 400) 1504 onto the cell expansion system. Next, the system may be primed 1506. In an embodiment, a user or operator, for example, may provide an instruction to the system to prime by selecting a task for priming, for example. In an embodiment, such task for priming may be a pre-programmed task. Next, an IC/EC washout task may be performed 1508, in which fluid on the IC circulation loop and on the EC circulation loop may be replaced, for example. The replacement volume may be determined by the number of IC volumes and EC volumes exchanged, according to an embodiment.


Next, to maintain the proper or desired gas concentration across the fibers in the bioreactor membrane, the condition media task 1510 may be executed to allow the media to reach equilibrium with the provided gas supply before cells are loaded into the bioreactor. For example, contact between the media and the gas supply provided by the gas transfer module (GTM) or oxygenator may be provided by adjusting the EC circulation rate. The system may then be maintained in a proper or desired state until a user or operator, for example, is ready to load cells into the bioreactor. In an embodiment, the system may be conditioned with media, such as complete media, for example. Complete media may be any media source used for cell growth. In an embodiment, the system may be conditioned with serum-free media, for example. In an embodiment, the system may be conditioned with base media. Any type of media understood by those of skill in the art may be used.


Process 1500 next proceeds to loading cells 1512 into the bioreactor from a cell inlet bag, for example. In an embodiment, the cells in the cell inlet bag may be in solution with media to feed the cells, for example. In another embodiment, the cells in the cell inlet bag may be in solution both with media and with a soluble activator complex to stimulate the cells, e.g., T cells or Tregs. In an embodiment, the cells may be loaded into the bioreactor from the cell inlet bag until the bag is empty. Cells may be chased from the air removal chamber to the bioreactor. In an embodiment, a “load cells with uniform suspension” task may be executed to load the cells. In another embodiment, a “load cells centrally without circulation” task may be executed to load the cells into a specific, e.g., central, region of the bioreactor. Other loading methods and/or loading tasks may be used in accordance with embodiments.


Next, process 1500 proceeds to feed the cells per a first process during a first time period 1514. In an embodiment, the cells may be fed at a minimum or low feed rate, for example, where the cell population is beginning to grow/expand, and a minimum or low feed rate is able to meet the feeding demands of such population. For example, an IC inlet pump rate of +0.1 mL/min to cause or produce a first fluid flow rate of 0.1 mL/min may be used during such first time period. While this example provides for a low or minimum feed rate of 0.1 mL/min, a low or minimum feed rate may be greater than or equal to about 0.01 mL/min and less than or equal to about 0.1 mL/min, according to embodiments. In embodiments, the low or minimum feed rate may be greater than 0.1 mL/min. If it is desired to reduce the loss of cells from the hollow fiber membrane bioreactor during such first time period, the IC inlet pump rate of +0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture. Other pump rates and resulting fluid flow rates may be used in other embodiments.


Next, process 1500 proceeds to query 1516 to determine whether to adjust the feeding rate to retain the cells in the bioreactor itself while also accounting for a growing cell population and increasing feeding demands, according to embodiments. For example, FIG. 15B shows an increasing feed rate in response to an increasing cell population. As shown in FIG. 15B, graph 1528 shows the cell number versus the IC flow rate for a run or procedure on a cell expansion system, e.g., Quantum® Cell Expansion System. In an embodiment, the IC flow rate comprises media for feeding the cells, and may thus also be referred to as the IC media flow rate. The number of cells 1530 is shown versus the IC flow rate (mL/min) 1532. As shown, the IC flow rate increases from 0.1 mL/min to 0.2 mL/min to 0.3 mL/min with an increase in the cells and the growing feeding demands of an expanding cell population, according to an embodiment. In the embodiment shown, there may be a substantially linear relationship, as shown by line 1534, between the number of cells and the IC flow rate.


Returning to FIG. 15A and query 1516, if it is not desired to adjust the feeding rate, process 1500 proceeds “no” to expand the cells 1520, in which the cells may continue to grow/expand using the media with which they were fed during the first time period 1514, for example. While the expansion of cells is shown at step 1520, the cells may also grow/expand during one or more other step(s), such as 1512, 1514, 1516, 1518, for example. On the other hand, if it is desired to adjust the feeding rate while keeping the cells in the bioreactor, process 1500 proceeds “yes” to feed the cells per a second process during a second time period 1518. In an embodiment, such second process may involve feeding the cells at substantially the same feed rates as during the first time period, for example. In another embodiment, the second process may involve feeding the cells at different feed rates as compared to the feed rates used during the first time period. In an embodiment, the IC inlet flow rate may be increased, and the IC circulation flow rate may be set, or configured or adjusted, to equal a percentage or portion or fraction of the IC inlet flow rate. For example, the IC circulation flow rate may be set to equal about fifty percent (50%) or about one-half (½), or another percentage or portion according to embodiments, of the value of the IC inlet flow rate, according to an embodiment. Determining whether to set, configure or adjust, the IC circulation flow rate to a percentage or fraction or portion of the IC inlet flow rate may be based on the value of the IC inlet flow rate, according to an embodiment. For example, an embodiment provides for the following method to retain cells in a bioreactor when feeding the cells using IC inlet flow (where QIC Circ=IC circulation flow rate (mL/min); QIC Inlet=IC Inlet flow rate (mL/min)):

QIC Circ=(−)½*QIC Inlet when QIC Inlet≥0.2 mL/min
and
QIC Circ=(−)QIC Inlet when QIC Inlet=0.1 mL/min.


While the above equations provide for different calculations of the IC circulation flow rate based on the values of the IC inlet flow rates (e.g., 0.2 mL/min or 0.1 mL/min), other values of such IC inlet flow rate for making such determination may be used, according to other embodiments. Further, while about fifty percent (50%) or about one-half (½) is used in this example, other percentages, ratios, fractions, and/or portions may be used in accordance with embodiments. Returning to process 1500, after feeding the cells with such flow and counter-flow properties as a part of second process 1518 to keep the cells in the bioreactor, process 1500 proceeds to grow/expand the cells 1520. While the expansion of cells is shown at step 1520, the cells may also grow/expand during one or more other step(s), such as 1512, 1514, 1516, 1518, for example. From expand step 1520, process 1500 proceeds to harvest or remove the cells 1524. Process 1500 may then terminate at END operation 1526. If any other steps are desired before harvesting the cells, process 1500 proceeds to optional “Other” step 1522, according to embodiments. From optional step 1522, process 1500 proceeds to harvest or remove the cells from the bioreactor 1524, and process 1500 may then terminate at END operation 1526.


Next, FIG. 16 illustrates example operational steps 1600 of a process for feeding cells that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5B & 5C), in accordance with embodiments of the present disclosure. START operation 1602 is initiated, in which a disposable set may be loaded onto a cell expansion system, the system may be primed, IC/EC washout may be performed, media may be conditioned, and cells may be loaded, for example. Process 1600 next proceeds to feed the cells according to a first process during a first time period 1604. In an embodiment, the cells may be fed at a minimum or low feed rate, for example, where the cell population is beginning to grow/expand, and a minimum or low feed rate is able to meet the feeding demands of such population. For example, an IC inlet pump rate of +0.1 mL/min may be used during such first time period. While this example provides for a low or minimum feed rate of 0.1 mL/min, a low or minimum feed rate may be greater than or equal to about 0.01 mL/min and less than or equal to about 0.1 mL/min, according to embodiments. In embodiments, the low or minimum feed rate may be greater than 0.1 mL/min. If it is desired to reduce the loss of cells from the hollow fiber membrane bioreactor during such first time period, the IC inlet pump rate of +0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture.


Next, process 1600 proceeds to query 1606 to determine whether the feeding rate may be adjusted to account for a growing cell population and/or to continue efforts to keep the cells in the bioreactor. If it is desired to adjust the feeding rate to retain the cells in the bioreactor, process 1600 proceeds “yes” to feed the cells according to a second process during a second time period 1608. In an embodiment, such second process may involve feeding the cells at substantially the same feed rates as during the first time period, for example. In another embodiment, the second process may involve feeding the cells at different feed rates as compared to the feed rates used during the first time period. In an embodiment, the IC inlet flow rate may be increased, and the IC circulation flow rate may be set, or configured or adjusted, to equal a percentage or fraction or portion of the IC inlet flow rate. For example, the IC circulation flow rate may be set to equal about fifty percent (50%) or about one-half (½), or another percentage or portion according to embodiments, of the value of the IC inlet flow rate, according to an embodiment. Determining whether to set the IC circulation flow rate to a percentage of the IC inlet flow rate may be based on the value of the IC inlet flow rate, according to an embodiment. For example, an embodiment provides for the following (where QIC Circ=IC circulation flow rate (mL/min), QIC Inlet=IC Inlet flow rate (mL/min)):

QIC Circ=(−)½*QIC Inlet when QIC Inlet≥0.2 mL/min
and
QIC Circ=(−)QIC Inlet when QIC Inlet=0.1 mL/min.


While the above equations provide for different calculations of the IC circulation flow rate based on the values of the IC inlet flow rates (e.g., 0.2 mL/min or 0.1 mL/min), other values of such IC inlet flow rate for making such determination may be used, according to other embodiments. Further, while about fifty percent (50%) or about one-half (½) is used in this example, other percentages, ratios, fractions, and/or portions may be used in accordance with embodiments. Returning to process 1600, after feeding the cells with such flow and counter-flow properties as a part of second process 1608 to keep the cells in the bioreactor, process 1600 proceeds to query 1610 to determine whether to monitor or measure the metabolic activity, e.g., glucose consumption and/or lactate generation, of the growing cell population. If it is not desired to monitor the metabolic activity, process 1600 proceeds “no” to expand the cells 1616, in which the cells may continue to grow/expand using the media with which they were fed during the first time period 1604 and/or second time period 1608, for example. While the expansion of cells is shown at step 1616, the cells may also grow/expand during one or more other step(s), such as 1604, 1606, 1608, 1610, 1612, 1614, for example.


Returning to query 1610, if it is desired to monitor or measure the metabolic activity of the growing cell population, process 1600 proceeds “yes” to either continue to feed the cells per the second process or adjust the feed rate, based on the metabolic activity and/or measurements thereof. In an embodiment, monitoring the glucose and/or lactate levels may facilitate the adjustment of media flow rates, e.g., IC flow rate, to support cell, e.g., T cell or Treg, expansion in a bioreactor, e.g., hollow fiber bioreactor. In embodiments, cell culture lactate values may be maintained below about 7 mmol/L, for example. In embodiments, by using a cell expansion system graphical user interface (GUI), for example, to control a rate(s) of media addition, lactate metabolic waste product from glycolysis may be maintained below about 7 mmol/L, for example, during the expansion of cells, e.g., regulatory T cells. In other embodiments, rate(s) of media addition, for example, and/or other settings may be controlled to attempt to maintain the lactate levels≤about 5 mmol/L, for example, to improve cell growth and viability. Other concentrations may be used in other embodiments.


Depending on the metabolic measurements and the desired levels of lactate, for example, process 1600 proceeds to either continue feeding the cells according to the second process 1612 or adjusting the feed rate 1614. For example, the cells may be continued to be fed according to the second process 1612 where measurements of the metabolic activity show lactate levels≤about 5 mmol/L, according to an embodiment. In another embodiment, the cells may be continued to be fed according to the second process 1612 where the measurements of the metabolic activity show lactate levels≤about 7 mmol/L, for example. From continuing to feed the cells per the second process 1612, process 1600 returns to query 1610 to continue monitoring the metabolic activity of the growing cell population.


Depending on the metabolic measurements and the desired levels thereof, process 1600 proceeds to adjust the feed rate 1614, in which the cells may be fed according to an additional process during an additional time period. Such additional process(es) and additional time period(s) may include, for example, a third process during a third time period, a fourth process during a fourth time period, a fifth process during a fifth time period, etc., according to embodiments. In an embodiment, such additional process may involve feeding the cells at substantially the same feed rates as during the first and/or second time periods, for example. In another embodiment, the additional process may involve feeding the cells at different feed rates as compared to the feed rates used during the first and/or second time periods. For example, the IC inlet flow rate may be increased, and the IC circulation flow rate may be matched, or closely or substantially matched, to the IC inlet flow rate, but in the opposite direction, in an embodiment. In another embodiment, the IC inlet flow rate may be increased, and the IC circulation flow rate may be set, or configured or adjusted, to equal a percentage or fraction or portion of the IC inlet flow rate, and in the opposite direction. While adjusting the feed rate 1614 shows an “additional” process and an “additional” time period in step 1614, any number of processes and time periods may be used to adjust the feed rate based on metabolic activity.


From adjusting the feed rate 1614, process 1600 returns to query 1610. If it is not desired to adjust, or further adjust, the feeding rate, process 1600 proceeds “no” to expand the cells 1616, in which the cells may continue to grow/expand using the media with which they were fed during the first time period 1604, second time period 1608, and/or additional time period 1614. While the expansion of cells is shown at step 1616, the cells may also grow/expand during one or more other step(s), such as step(s) 1604, 1606, 1608, 1610, 1612, 1614, for example. From expand step 1616, process 1600 proceeds to harvest or remove the cells 1618 from the bioreactor and into a harvest bag(s) or container(s), for example. Process 1600 may then terminate at END operation 1622. Alternatively, from harvest operation 1618, process 1600 may optionally proceed to allow for further processing/analysis 1620. Such optional further processing/analysis 1620 may include characterizing the phenotype(s), for example, of the harvested cells, e.g., T cells or Tregs. From optional further processing/analysis step 1620, process 1600 may then terminate at END operation 1622.



FIG. 17A illustrates operational steps 1700 of a process for expanding cells that may be used with a cell expansion system in embodiments of the present disclosure. As described below, process 1700 may include steps to shear cells that have been expanded in the cell growth chamber according to embodiments of the present disclosure. In embodiments, these steps may be implemented as part of a “modified circulation” task. START operation 1702 is initiated and process 1700 proceeds to loading fluid 1704 with cells into a cell growth chamber in a cell expansion system. In embodiments, the cells may comprise non-adherent cells, such as one or more types of T cells. In one embodiment, the cells include Tregs.


Process 1700 proceeds to exposing the cells to an activator 1706. The activator, which may include antibody complexes, may be added to the fluid loaded at step 1704. In embodiments, the activator may be a soluble human antibody CD3/CD28/CD2 cell activator complex. Process 1700 proceeds to expanding 1708 the cells for a first time period. Step 1708 may include feeding 1710 the cells. The cells may be fed nutrients to promote their expansion. For example, media with glucose, proteins, and reagents may be delivered into the cell growth chamber to provide nutrients for cell expansion.


The first time period for expanding 1708 the cells may be based on the time it may take for cell colonies, micro-colonies, or clusters to form. A cell colony, micro-colony, or cluster may be a group of one or more attached cells. In embodiments, the cells, e.g., Tregs, may benefit from cell contact. The cell contact may stimulate signaling that promotes expansion and growth. However, after a period of expansion, the cells may attach to each other to form cell colonies, micro-colonies, or clusters. Without being bound by theory, it is believed that after a time period of the cells expanding 1708 the cells may form relatively large cell colonies, micro-colonies, or clusters that continue to grow. The cell colonies, micro-colonies, or clusters may create necrotic centers where nutrients (e.g., glucose), gasses (e.g., oxygen), and reagents (e.g., activator) do not reach cells in the center of the cell colonies, micro-colonies, or clusters. As a result, the conditions for cell expansion in the center of these cell colonies, micro-colonies, or clusters may be such that the expansion rate may slow (e.g., increase doubling time) or the conditions may lead to cell necrosis.


In embodiments, to allow the expansion 1708 of the cells, the first time period may be between about 5 hours and about 48 hours. In some embodiments, the first time period may be greater than about 6 hours, greater than about 12 hours, greater than about 24 hours, or even greater than about 48 hours. In other embodiments, the first time period may be less than about 72 hours, less than about 60 hours, less than about 48 hours, less than about 36 hours, less than about 24 hours, or even less than about 12 hours. After the first time period, process 1700 proceeds to circulate 1712 to shear cell colonies, micro-colonies, or clusters during a second time period. Step 1712 may be performed to reduce the size of the cell colonies, micro-colonies, or clusters. The second time period may in embodiments be less than about 120 minutes such as between about 60 minutes and about 0.5 minutes. In other embodiments, the second time period may be based on a volume of fluid introduced into the first circulation path.



FIG. 17B illustrates a number of views 1750, 1760, and 1770 of cells in a volume of fluid (1752) that may be expanded in a cell growth chamber as part of process 1700. For example, in some embodiments, the cell growth chamber may be a hollow fiber bioreactor. In these embodiments, views 1750, 1760, and 1770 may illustrate cells in a fiber of a hollow fiber bioreactor, for example. 1750A, 1760A, and 1770A are zoomed in portions of views 1750, 1760, and 1770, respectively. Referring to view 1750, the cells may be shown after cells have been loaded 1704, exposed to an activator, 1706 and expanded 1710 for a time period, e.g., the first time period. As illustrated in view 1750, a number of cell colonies 1754A-E have formed.


In order to reduce the number of cells in, and the size of, the cell colonies, micro-colonies, or clusters 1754A-E, step 1712 may circulate fluid and the cells through a first fluid circulation path. Without being bound by theory, it is believed that the circulation may create some force acting on the cell colonies including shear stress, as illustrated by arrow 1756 as shown in zoomed in portion 1760A. The shear stress 1756 may provide enough force to separate cells in the cell colonies. As the circulation continues, the cell colonies may begin to break up into smaller sizes, as shown in view 1760. View 1770 illustrates the cells after the circulation has been performed for the second time period. As illustrated in view 1770, the size of the cell colonies are reduced with some colonies being completely separated into individual cells. In some embodiments, the circulation to shear step 1712 may be performed until the cells and fluid comprise a single cell suspension.


In other embodiments, cell colonies, micro-colonies, or clusters of cells may remain after circulate to shear 1712. For example, colony 1754F in zoomed in portion 1770A illustrates that some colonies of a reduced size may remain after step 1712. In embodiments the cell colonies, micro-colonies, or clusters (e.g., 1754F) that remain may be between about 25 microns and about 300 microns. In other embodiments, circulate to shear 1812 may reduce the size of cell colonies, micro-colonies, or clusters (e.g., 1754F) so the cell colonies, micro-colonies, or clusters may be between about 50 microns and about 250 microns. In yet other embodiments, step 1712 may reduce the size of cell colonies, micro-colonies, or clusters to between about 75 microns and about 200 microns. In some embodiments, the size of the cell colonies, micro-colonies, or clusters may be less than about 200 microns e.g., about 100 microns after step 1712.


In embodiments, the size of the remaining cell colonies, micro-colonies, or clusters may be somewhat a function of some structural features of the cell growth chamber. As mentioned above, the cell growth chamber may be a hollow fiber bioreactor with hollow fibers in some embodiments. As may be appreciated, cell colonies, micro-colonies, or clusters, as they circulate, may be affected by shear stress each time they contact the side walls of the hollow fiber. This contact may more efficiently reduce the size of cell colonies. When the inner diameter is larger, such as in a conventional process that may utilize a pipet to induce shear stress to reduce colony sizes, contact with a side wall may not occur as often. FIG. 17C illustrates differences in inner diameter size between one embodiment of a hollow fiber (e.g., 215 microns) 1772 and a pipet tip 1774 (762 microns), which may be used to disassociate attached cells in cell colonies, micro-colonies, or clusters. In embodiments, the smaller inner diameter of a hollow fiber is believed to more efficiently and effectively reduce a size of cell colonies during the circulating to shear step 1712.


The second time period for the circulate to shear 1712 may in embodiments be less than about 120 minutes, less than about 90 minutes, less than about 60 minutes, less than about 30 minutes, or even less than about 15 minutes. In some embodiments, the second time period may be between about 15 minutes and about 1 minute, such as about 4 minutes.


After the second time period, process 1700 proceeds to move cells into cell growth chamber during a third time period 1714. At step 1714 cells that are not positioned in the cell growth chamber, as a result of the circulation to shear step 1712, are moved back into the cell growth chamber during a third time period. In embodiments, this may involve activating one or more pumps to introduce fluid into a fluid circulation path. For example, fluid may be introduced from a fluid inlet path to a first fluid flow path and then into the cell growth chamber, from both an inlet port and an outlet port of the cell growth chamber. The movement of the fluid into the cell growth chamber from the inlet port and the outlet port may move cells back into the cell growth chamber.


In some embodiments, the fluid used in the step to move the cells back into the cell growth chamber 1714 may include reagents that promote cell growth. For example, in embodiments, the fluid may be media that includes glucose, proteins, or other reagents. In one embodiment, the fluid may include one or more supplements. In one embodiment, the fluid is complete media and includes a cytokine, e.g., human IL-2 cytokine supplement. The addition of the fluid may be referred to as a bolus addition. The combination of steps 1712 and 1714 may be referred to in embodiments as circulate and bolus addition.


In other embodiments, the third time period may be based on a volume of fluid introduced into the cell growth chamber during the circulate to shear step 1712. For example, in embodiments, step 1712 may be performed until about 300 ml, about 250 ml, about 200 ml, or about 150 ml, have been introduced into the fluid circulation path.


After the third time period, process 1700 proceeds to expand during a fourth time period 1716. Similar to step 1708, step 1716 may include feeding 1718 the cells. The cells may be fed nutrients to promote their expansion. For example, media with glucose, proteins, and reagents may be delivered into the cell growth chamber to provide nutrients for cell expansion.


Similar to the first time period, the fourth time period may be based on the time it may take for cell colonies to form. In embodiments, the fourth time period may be between about 5 hours and about 48 hours. In some embodiments, the fourth time period may be greater than about 6 hours, greater than about 12 hours, greater than about 24 hours, or even greater than about 48 hours. In other embodiments, the fourth time period may be less than about 72 hours, less than about 60 hours, less than about 48 hours, less than about 36 hours, less than about 24 hours, or even less than about 12 hours. Because more cells are likely in the cell growth chamber, the fourth time period may be shorter than the first time period in some embodiments.


After the fourth time period, process 1700 proceeds to step 1720 to circulate to shear for a fifth time period to reduce second cell colonies. In embodiments, step 1720 may use the first circulation rate. However, in other embodiments, the circulation rate used at step 1720 may be different, either greater or less than the first circulation rate.


After the fifth time period, process 1700 proceeds to step 1722 where the cells that are not positioned in the cell growth chamber, may be moved back into the cell growth chamber during a sixth time period. Fluid may be introduced from a fluid inlet path to a first fluid flow path and into the cell growth chamber from an inlet port and an outlet port of the cell growth chamber. The movement of the fluid into the cell growth chamber from the inlet port and the outlet port may move cells back into the cell growth chamber. In some embodiments, the fluid used to move the cells back into the cell growth chamber may include reagents that promote cell growth. For example, in embodiments, the fluid may be media that includes glucose, proteins, or other reagents. In one embodiment, the fluid may include one or more supplements. In one embodiment, the fluid is complete media and includes a cytokine, e.g., human IL-2 cytokine.


The process 1700 may optionally perform the steps of expand, circulate, and move cells for an additional number of times as illustrated by optional step 1724 and ellipsis 1726. The steps of expand, circulate, and move may be performed sequentially for a period of time. For example, in some embodiments, the steps may be performed once every four days, once every three days, once every two days, daily, twice daily, or three times daily, for a period of from about two days to about twenty days (such as about 10 days). In some embodiments, the steps may be performed at varying periods of time. For example, in one embodiment, the steps may be performed after three days, and then every other day. As another example, the steps may be performed after two days and then twice daily. These are merely examples and other embodiments may utilize other periods of time.


For example, FIG. 18 shows a graph 1800 of performing circulation and bolus additions at different periods of time during a process of expanding cells. Curve 1808 shows the cell number 1802 versus days of cell culture 1804 on a cell expansion system, e.g., Quantum® Cell Expansion System. Curve 1806 shows IC Flow rate 1818 versus days of cell culture 1804 on a cell expansion system, e.g., Quantum® Cell Expansion System. As shown by curve 1806, the IC flow rate remains the same at 0.1 mL/min for the first three days. There is an increase in the flow rate to 0.2 mL/min at day six and an increase to 0.3 mL/min after day seven. The increase in flow rate may be in response to the increase in cell numbers as the culture days 1804 increase. Also, shown in FIG. 18, are several circulation and bolus addition steps (e.g., 1810, 1812, 1814, and 1816). There is a circulation and bolus addition 1810 performed 3.5 days after cell culture. There is another circulation and bolus addition 1812 performed after 4.5 days of cell culture. Another circulation and bolus addition is performed after 6 days of cell culture 1814, and another circulation and bolus addition after 6.5 days of cell culture 1816. As may be appreciated by looking at curves 1806 and 1808, the multiple circulation and bolus additions (1810-1816), in combination with the increasing IC flow rates, may have a positive effect on the rate of cell expansion, e.g., number of cells.


Referring back to 17A, process 1700 proceeds to remove cells 1728 from the cell growth chamber. In embodiments, this may involve harvesting the cells. Step 1728 may include additional steps such as circulation steps (e.g., 1712 and 1720) prior, or during, removal of the cells from the cell growth chamber. Process 1700 terminates at END operation 1730.



FIG. 19A illustrates operational steps 1900 of a process for operating pumps that may be used in a cell expansion system in embodiments of the present disclosure. As described below, process 1900 may include steps to activate pumps in a process of reducing cells in cell clusters that have been expanded in the cell growth chamber according to embodiments of the present disclosure. In embodiments, these steps may be implemented as part of a “modified circulation” task. In embodiments, the steps of process 1900 may be performed by a computer processor. START operation 1902 is initiated and process 1900 proceeds to step 1904, where a first pump is activated at a first flow rate to introduce a first volume of fluid including cells into the intracapillary portion of a bioreactor of a cell expansion system. In embodiments, the first pump may be an inlet pump.


After activating the first pump at the first flow rate, process 1900 proceeds to the first pump activated at a second flow rate 1906 to introduce media with nutrients into the intracapillary portion of the bioreactor for a first time period. The nutrients may include for example proteins, glucose, and other compounds that are used to feed and promote the expansion of the cells. For example, referring to FIG. 19B, a first pump 1960 may be activated at the second flow rate to introduce media into an inlet port 1962A of bioreactor 1962.


In embodiments, the first time period may be based on the time it may take for cell colonies to form. In embodiments, the first time period may be between about 5 hours and about 48 hours. In some embodiments, the first time period may be greater than about 6 hours, greater than about 12 hours, greater than about 24 hours, or even greater than about 48 hours. In other embodiments, the first time period may be less than about 72 hours, less than about 60 hours, less than about 48 hours, less than about 36 hours, less than about 24 hours, or even less than about 12 hours.


Process 1900 then proceeds to activating a second pump at a third flow rate to direct fluid into the bioreactor 1908. Optional step 1908 may be performed to activate pump 1964 to direct a portion of the fluid, introduced at step 1906, into an outlet port 1962B of the bioreactor 1962 to feed cells, for example.


Process 1900 then proceeds to activating the second pump at a fourth flow rate 1910 to circulate the cells during a second time period and reduce a number of cells in a cell cluster in the bioreactor. In embodiments, the cells are circulated throughout a first fluid circulation path. Referring to FIG. 19C, second pump 1964 may be activated at step 1910 to circulate fluid through first fluid circulation path 1966, s illustrated by arrows 1968A-D.


Without being bound by theory, it is believed that after a time period the expanding cells may form cell colonies, micro-colonies, or clusters. The cell colonies may create necrotic centers where nutrients and proteins (e.g., activator) do not reach cells in the center of the colonies. As a result, the conditions for cell expansion in the center of these cell colonies, micro-colonies, or clusters may be such that the expansion rate may slow (e.g., increase doubling time) and may result in cell necrosis. Step 1910 may be performed to reduce the size of the cell colonies, micro-colonies, or clusters.


In embodiments, the fourth flow rate may be high enough to induce shearing. For example, in embodiments, the fourth flow rate may be as high as about 1000 ml/min. In other embodiments, the fourth flow rate may be between about 100 mL/min and about 600 ml/min, such as for example, 300 mL/min.


After the second time period, process 1900 proceeds to step 1912, where the first pump is activated at a fifth flow rate to introduce fluid through a first fluid flow path for a third time period. A first portion of the fluid introduced in the first fluid flow path may in embodiments move first cells in the first fluid flow path (that may be in the first fluid flow path because of step 1910) back into the cell growth chamber through the inlet port 1962A. Referring to FIG. 19D, pump 1960 may be activated to introduce fluid into the first fluid flow path 1970. As illustrated by arrow 1968A and 1968B, fluid flows from the first fluid flow path 1970 into inlet port 1962A. This moves first cells that are outside bioreactor 1962 back into bioreactor 1962. It is believed that moving cells that are in the first fluid flow path back into the cell growth chamber improves the overall expansion of cells, since the conditions for cell growth are optimized in the cell growth chamber.


In some embodiments, the fluid introduced into the first fluid flow path at step 1912, may include one or more materials (e.g., reagents) that promote cell expansion. For example, in embodiments, the fluid may be media that includes glucose or other nutrients for feeding the cells. In one embodiment, the fluid may include a reagent, which may comprise additional activator for continuing to activate the expansion of cells. The use of the fluid with particular reagent(s) or other materials to move the cells back into the cell growth chamber, and also expose the cells to additional reagents, e.g., growth factors, proteins, etc., that promote expansion may provide improved cell expansion. In embodiments, the additional fluid used in step 1912 may be referred to as a bolus addition.


Also, after the third time period, at step 1914, the second pump may be activated at a sixth flow rate to move a second portion of the fluid introduced through the first fluid flow path, and second cells, into the cell growth chamber through the outlet port 1962B. The second portion of the fluid may in embodiments move second cells in the first fluid flow path (that may be in the first fluid flow path because of step 1910) back into the cell growth chamber through the outlet port. Referring to FIG. 19D, pump 1964 may be activated to move fluid introduced into the first fluid flow path 1970 to the outlet port 1962B. As illustrated by arrow 1968C, pump 1964 moves fluid and cells in first fluid circulation path into bioreactor 1962 through outlet port 1962B. As illustrated, by FIG. 19D, the sixth flow rate may be in a direction opposite the fourth flow rate (FIG. 19C). This fluid movement moves cells that are outside bioreactor 1962 back into bioreactor 1962.


In embodiments, the sixth flow rate may be less than the fifth flow rate, which as noted above moves fluid into the first fluid flow path. As may be appreciated, the fifth flow rate may result in introduction of a volume of fluid into a portion of the circulation path 1966 based on the fifth flow rate. The sixth rate may be set so that a percentage of that volume moves toward the outlet port 1962B.


In embodiments, the sixth flow rate may be set as a percentage of the fifth flow rate. For example, the sixth flow rate may be less than or equal to about 90% of the fifth flow rate. In some embodiments, the sixth flow rate may be set to less than or equal to about 80% of the fifth flow rate. In other embodiments, the sixth flow rate may be less than or equal to about 70% of the fifth flow rate. In yet other embodiments, the sixth flow rate may be less than or equal to about 60% of the fifth flow rate. In some embodiments, the sixth flow rate may be less than or equal to about 50% of the fifth flow rate.


In embodiments, the sixth flow rate is based, at least in part, on the difference between a first volume, between the second pump and the inlet port, and a second volume, between the second pump and the outlet port. Referring to FIG. 19D, portions of the first circulation path may have different volumes. For example, in one embodiment, a first volume between the second pump 1964 and the inlet port 1962A may have a first volume, and a second volume between the second pump 1964 and the outlet port 1962B may have a second volume that is different from the first, e.g., larger. In these embodiments, in order to move the cells from the circulation path back into the bioreactor, so that the cells generally reach the bioreactor at approximately the same time, the sixth flow rate may be set at least in part based on the differences in these volumes.


As may be appreciated, as fluid enters fluid circulation path 1966 from first fluid flow path 1970, the fluid moves toward inlet port 1962A at the flow rate that first pump 1960 is set. When pump 1964 is activated, it will redirect at least a portion of the fluid toward the outlet port 1962B. In embodiments, the second volume (the volume between 1964 and outlet port 1962B) may be larger than the first volume. Therefore, in order to move more fluid into the second volume, the second pump 1964 may be set at a percentage of the rate of pump 1960.


In one embodiment, at step 1912, pump 1960 may be set to 100 ml/min. In this embodiment, the second volume (from pump 1964 to outlet port 1962B) may be larger than the first volume (from pump 1964 to inlet port 1962A). In order to account for the additional volume, embodiments may provide for pump 1964 to be set at 70 ml/min during step 1914. This embodiment may provide for cells to reach bioreactor 1962 during the third time period at about the same time.


In embodiments, process 1900 may optionally perform the steps of 1906-1914 a number of additional times as illustrated by ellipsis 1916 and optional step 1918. The steps of Activate: First Pump (at Second Rate) 1906, Second Pump (at Fourth Rate) 1910, First Pump (at Fifth Rate) 1912, and Second Pump (at Sixth Rate) 1914 may be continuously performed for a period of time. As noted above, the steps may be performed to feed cells, circulate cells to break up cell colonies, micro-colonies, or clusters, and move cells back into a cell growth chamber. For example, in some embodiments, the steps may be performed every three days, every two days, daily, twice daily, or three times a day, for a period of from about two days to about twenty days (such as 10 days). In some embodiments, the steps may be performed at varying periods of time. For example, in one embodiment, the steps may be performed after three days, and then every other day. As another example, the steps may be performed after two days and then twice daily. This is merely one example and other embodiments may utilize other periods of time. Process 1900 terminates at END operation 1920.


It is noted that in some embodiments, process 1900 may include additional steps. For example, a rocking device may be connected to the bioreactor and after the first time period (and during the second time period), when the first pump is activated at step 1910, the rocking device may be activated to rotate the bioreactor as part of circulating the cells to reduce a number of cells in a cell cluster. This is merely one example and other embodiments of process 1900 are not limited thereto.



FIG. 20 illustrates example operational steps 2000 of a process for expanding cells that may be used with a cell expansion system, such as CES 500 (e.g., FIG. 5A) or CES 600 (FIG. 6), in accordance with embodiments of the present disclosure. START operation 2002 is initiated, and process 2000 proceeds to load the disposable tubing set 2004 onto the cell expansion system. Next, the system may be primed 2006. In an embodiment, a user or an operator, for example, may provide an instruction to the system to prime by selecting a task for priming, for example. In an embodiment, such task for priming may be a pre-programmed task. Process 2000 then proceeds to the IC/EC Washout task 2008, in which fluid on the IC circulation loop and on the EC circulation loop is replaced. The replacement volume is determined by the number of IC Volumes and EC Volumes exchanged.


Next, to maintain the proper or desired gas concentration across the fibers in the bioreactor membrane, the condition media task 2010 may be executed to allow the media to reach equilibrium with the provided gas supply before cells are loaded into the bioreactor. For example, rapid contact between the media and the gas supply provided by the gas transfer module or oxygenator is provided by using a high EC circulation rate. The system may then be maintained in a proper or desired state until a user or operator, for example, is ready to load cells into the bioreactor. In an embodiment, the system may be conditioned with complete media, for example. Complete media may be any media source used for cell growth. In an embodiment, complete media may comprise alpha-MEM (α-MEM) and fetal bovine serum (FBS), for example. Any type of media understood by those of skill in the art may be used.


Process 2000 next proceeds to loading cells centrally without circulation 2012 into the bioreactor from a cell inlet bag, for example. In embodiments, a “load cells centrally without circulation” task may be used, in which a first volume of fluid at a first flow rate comprising a plurality of cells may be loaded into the cell expansion system, in which the cell expansion system comprises a cell growth chamber. A second volume of fluid at a second flow rate comprising media may then be loaded into a portion of a first fluid circulation path, for example, to position the first volume of fluid in a first portion of the cell growth chamber. In an embodiment, the first portion of the cell growth chamber or bioreactor may comprise about a central region of the bioreactor. In an embodiment, the first volume is the same as the second volume. In an embodiment, the first flow rate is the same as the second flow rate. In another embodiment, the first volume is different from the second volume. In another embodiment, the first flow rate is different from the second flow rate. In an embodiment, the sum of the first volume and the second volume may equal a percentage or proportion of the volume, e.g., total volume, of the first fluid circulation path, for example. For example, the sum of the first volume and the second volume may be about 50% of the volume, e.g., total volume, of the first fluid circulation path, for example. In an embodiment, fluid in the first fluid circulation path flows through an intracapillary (IC) space of a bioreactor or cell growth chamber. In an embodiment, fluid in a second fluid circulation path flows through an extracapillary (EC) space, for example, of a cell growth chamber or bioreactor. In an embodiment, the sum of the first volume and the second volume may be about 50%, or another percentage or proportion according to embodiments, of the volume of the intracapillary (IC) loop, for example. In an embodiment, the sum of the first volume and the second volume may be about 50%, or another percentage or proportion according to embodiments, of the volume of another fluid path, loop, etc., as applicable. Other percentages or proportions may be used, including, for example, any percentage between and including about 1% and about 100%, in accordance with embodiments.


Following the loading of the cells 2012, process 2000 next proceeds to feed the cells 2014. The cells may be grown/expanded 2016. While step 2016 is shown after step 2014, step 2016 may occur before, or simultaneous with, step 2014, according to embodiments. Next, process 2000 proceeds to query 2018 to determine whether any cell colonies, micro-colonies, or clusters have formed. A cell colony, micro-colony, or cluster may be a group of one or more attached cells. If a cell colony, micro-colony, or cluster has formed, process 2000 proceeds “yes” to shear 2020 any cell colonies, micro-colonies, or clusters. For example, after expanding a plurality of cells for a first time period, the cells may be circulated at a first circulation rate during a second time period to reduce a number of cells in a cell colony, micro-colony, or cluster. In embodiments, the circulating the cells at the first circulation rate may cause the cell colony to incur a shear stress, in which one or more cells in the cell colony may break apart from the cell colony. In an embodiment, reducing the number of cells in the cell colony, micro-colony, or cluster may provide a single cell suspension, for example. In embodiments, circulating the cells to shear any colony, micro-colony, or cluster 2020 may be used every two (2) days, for example, during cell culture to maintain uniform cell density and nutrient diffusion. Other time periods may also be used according to embodiments. In an embodiment, such shearing of any micro-colonies, colonies, or clusters may begin on or after Day 4, for example. Other days or time periods on which to begin such shearing may be used according to embodiments. Following shearing 2020, process 2000 may next return to feed cells 2014.


If it is determined at query 2018 not to shear any cell colonies or clusters, or if none exist, for example, process 2000 proceeds “no” to resuspend cells 2022. In embodiments, circulating the cells may be used to uniformly resuspend those cells that may be loosely adhered during culture. In embodiments, step 2022 may include circulating the cells to uniformly resuspend those cells that may be loosely adhered prior to initiating a harvest task, or other task to remove cells from the bioreactor. Following the resuspension of the cells 2022, process 2000 next proceeds to harvest the cells 2024. Further processing of the removed cells or other analysis may optionally be performed at step 2026, and process 2000 may then terminate at END operation 2028. If it is not desired to perform further processing/analysis, process 2000 terminates at END operation 2030.


Turning to FIG. 21 and process 2100, START operation is initiated 2102, and process 2100 proceeds to load a disposable set 1204 onto a cell expansion system, according to embodiments. The disposable set may then be primed 2106, and an IC/EC washout step 2108 may occur. The media may next be conditioned 2110. Next process 2100 proceeds to load cells 2112, e.g., suspension or non-adherent cells, such as T cells or Tregs. In an embodiment, such cells may be loaded 2112 by a “load cells centrally without circulation” task. In another embodiment, such cells may be loaded 2112 by a “load cells with uniform suspension” task.


Next, process 2100 proceeds with beginning to feed the cells 2114, which may begin on Day 0, according to embodiments. The cells may grow and expand 2116, and by Day 3, for example, it may be desired to add a bolus of a fluid to the IC loop and re-distribute the cells 2118. In an embodiment, such bolus of fluid may comprise a reagent, such as cytokines or other growth factor(s). In another embodiment, such bolus of fluid may comprise a reagent and base media, for example.


Following such bolus addition and re-distribution of cells, process 2100 next proceeds to feeding 2120 the cells again, in which such feeding may occur on Day 3, for example. With such feeding 2120, the parameters of the system, such as one or more pumps controlling flow rate, may be controlled 2122 to achieve complementary flow and counter-flow settings for fluid moving into the bioreactor from both the IC inlet port and the IC outlet port of the bioreactor. For example, the IC inlet pump 2124 may be adjusted or directed to produce a flow, and the IC circulation pump may be adjusted or directed 2126 to produce a counter-flow. For example, an IC inlet pump rate of 0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture, which may be Days 4-7, for example, in embodiments. Such control of settings 2122 may allow for counteracting any forces associated with a loss of cells from the IC outlet port of the bioreactor.


Process 2100 next proceeds to query 2128, in which it is determined whether to continue to add reagent or other bolus addition on other days or other time intervals, for example. If it is desired to add additional reagent or other bolus and re-distribute cells, process 2100 branches “yes” to add reagent and re-distribute cells 2118. For example, such bolus addition and re-distribution of cells may next occur on Days 6 and 9, according to embodiments.


If, or once, it is not desired to continue adding a bolus, e.g., reagent, and re-distributing the cells, process 2100 proceeds “no” to harvest the cells 2130, in which the cells may be transferred to a harvest bag(s) or container(s). Process 2100 then terminates at END operation 2136.


Alternatively, from harvest operation 2130, process 2100 may optionally proceed to allow for further processing/analysis 2132. Such further processing 2132 may include characterization of the phenotype(s), for example, of the harvested cells, e.g., T cells or Tregs. From optional further processing/analysis step 2132, process 2100 may proceed to optionally reload any remaining cells 2134. Process 2100 may then terminate at END operation 2136.


Process 2200 illustrates operational steps for a process of expanding cells in cell expansion system according to embodiments of the present disclosure. Process 2200 may be used in some embodiments to expand T cells. As illustrated, various steps may be performed over the course of a 14-day protocol to expand the cells. START operation 2202 is initiated and process 2200 proceeds to Day 0, where a disposable set is loaded onto a cell expansion system 2206. The disposable set may then be primed 2208, in which the set may be primed 2208 with PBS (e.g., Lonza Ca2+/Mg2+−free), for example. In preparation for the loading of cells, the priming fluid may be exchanged using an IC/EC washout 2210. For example, the PBS in the system may be exchanged for TexMACS GMP Base Medium, according to one embodiment. The media may next be conditioned 2212. The condition media 2212 may be performed to allow the media to reach equilibrium with provided gas supply before cells are loaded into a bioreactor.


Next on Day 0, process 2200 proceeds to load cells 2214, e.g., suspension or non-adherent cells, such as T cells or Tregs. In an embodiment, such cells may be loaded 2214 by a “load cells centrally without circulation” task. In another embodiment, such cells may be loaded 2214 by a “load cells with uniform suspension” task.


At Day 3 2216, a bolus of cytokines may be added while the cells are redistributed 2218. In embodiments, the redistribution of cells may be performed in combination with the bolus addition to mix the cells and more thoroughly expose the cells to the cytokines (e.g., IL-2) that may be in the bolus addition. In embodiments, the redistribution may also break up colonies or clusters of cells that may have formed. In embodiments, the redistribution may occur first by circulating the cells in a fluid circulation path. The bolus addition may then be added in the process of pushing the cells back into the bioreactor, such as by introducing fluid into a fluid circulation path to push cells back into the bioreactor. After the redistribution and bolus addition 2218, process 2000 proceeds to feed cells 2220.


The cells may again be redistributed 2224 with another bolus addition at Day 6 2222. The redistribution may break up colonies or clusters of cells that may have formed during Days 3-5. The bolus addition may expose the cells to additional reagents that promote expansion. Process 2000 proceeds to feed cells 2226 at Day 6. At Day 9 2228, the cells may once again be redistributed 2230 with a bolus addition. The redistribution may break up colonies or clusters of cells that may have formed during Days 6-8. The bolus addition may expose the cells to additional supplements that promotes expansion. Process 2000 proceeds to feed cells 2232 at Day 9.


At Day 11-13 2234, the cells may again be redistributed 2236 with a bolus addition. The redistribution may break up colonies or clusters of cells that may have formed during Days 9-10. The bolus addition may expose the cells to additional reagents that promote expansion. Process 2000 then proceeds to feed cells 2238. In embodiments, the steps 2236 and 2238 may be performed on each of Day 11, Day 12, and Day 13. This may occur as a result of the cells expanding during Days 0-10 and there being larger numbers of cells in the bioreactor. Performing the redistribution and bolus addition of cells may promote expansion of the cells by breaking up colonies and clusters of cells more often and mixing them with reagents in the bolus addition to promote cell expansion. Process 2200 terminates at END operation 2240.


Process 2300 illustrates operational steps for a process of expanding cells, e.g., suspension or non-adherent cells, in a cell expansion system according to embodiments of the present disclosure. Process 2300 may be used in some embodiments to expand T cells, such as Tregs. The combination of steps of process 2300 may allow the expansion of the cells to useful clinical amounts using initial low seeding densities.


START operation 2302 is initiated and process 2300 proceeds to load disposable set 2304 onto a cell expansion system. The disposable set may then be primed 2206, in which the set may be primed 2206 with PBS (e.g., Lonza Ca2+/Mg2+−free), for example. In preparation for the loading of cells, the priming fluid may be exchanged using an IC/EC washout 2308. For example, the PBS in the system may be exchanged for TexMACS GMP Base Medium, according to one embodiment. The media may next be conditioned 2310. The condition media 2310 may be performed to allow the media to reach equilibrium with a provided gas supply before cells are loaded into a bioreactor.


Process 2300 proceeds to 2312 load inlet volume of fluid with cells. In embodiments, the cells may comprise non-adherent cells, such as one or more types of T cells, e.g., Tregs. In one embodiment, the cells comprise Tregs. Embodiments may provide for the inlet volume of fluid with the cells to be loaded through an IC inlet path utilizing an IC inlet pump and into an IC circulation path. In embodiments, the load inlet volume 2312 is loaded without activating an IC circulation pump.


Process 2300 proceeds to positioning the inlet volume in a first portion of a bioreactor 2314. In embodiments, the positioning may be performed by introducing a second volume of fluid, which may comprise media and may be introduced into a portion of the IC circulation path to push the inlet volume with the cells into the first position in the bioreactor. In embodiments, the inlet volume of fluid and the second volume of fluid may be the same. In other embodiments, the inlet volume of fluid and the second volume of fluid may be different. In yet other embodiments, a sum of the inlet volume of fluid and the second volume of fluid may be equal to a percentage of a volume of the IC circulation path.


Following the position inlet volume 2314, process 2300 proceeds to expose cells to activator 2316 in order to activate the cells to expand. The cells may be exposed to an activator 2316 that is soluble in some embodiments. The activator, which may include antibody complexes in some embodiments, may be added to the media and may be included in the inlet volume or added later, such as with the second volume. In embodiments, the activator may be a human antibody CD3/CD28/CD2 cell activator complex, for example.


Process 2300 proceeds to feed the cells per a first process during a first time period 2318. In an embodiment, the cells may be fed at a minimum or low feed rate, for example, where the cell population is beginning to grow/expand for a first time period 2320, and a minimum or low feed rate is able to meet the demands of such population. For example, an IC inlet pump rate of +0.1 mL/min may be used during such first time period. If it is desired to reduce the loss of cells from the hollow fiber membrane bioreactor during such first time period, the IC inlet pump rate of +0.1 mL/min may be matched, or closely or substantially matched, to a complementary IC circulation pump rate of −0.1 mL/min to maintain cells in the bioreactor during the growth phase of the cell culture.


From 2320, process 2300 proceeds to expand cells during a second time period 2322. Expanding during the second period of time 2322 may also involve feeding the cells per a second process during the second time period 2324. In an embodiment, such second process may involve feeding the cells at substantially the same feed rates as during the first time period, for example. In another embodiment, the second process may involve feeding the cells at different feed rates as compared to the feed rates used during the first time period. For example, the feed rates may increase as a result of the expansion of the cells during the first time period.


While expanding the cells during the second time period 2322, the cells may also be circulated to shear cells colonies or cell clusters 2326. Step 2326 may involve circulating the cells in the IC circulation path to shear any colonies or clusters that may have formed during the first time period. The shear colonies or clusters 2326 step may reduce a number of cells in a cell colony or cell cluster. In embodiments, the circulate to shear 2326 may cause cell colonies to incur a shear stress, causing one or more cells in the cell colony to break apart from the cell colony.


Process 2300 may next proceed to harvest operation 2328, in which the cells may be transferred to a harvest bag(s) or container(s). In embodiments, a therapeutic dose of cells may be harvested. In embodiments, the cells harvested at operation 2328 may be on the order of 1×109 cells. The harvested cells may have viabilities between about 75% and about 95%, in embodiments.


Process 2300 may then optionally proceed to allow for further processing/analysis 2330. Such further processing may include characterization of the phenotype(s), for example, of the harvested cells, e.g., T cells or Tregs. In one embodiment, the harvested cells may express biomarkers consistent with Tregs. For example, the cells may express CD4+, CD25+, and/or FoxP3+ biomarkers. In embodiments, the harvested cells may include the CD4+CD25+ phenotype at a frequency of above about 80%. In other embodiments, the cells may include the CD4+FoxP3+ phenotype at a frequency of above about 55%. Process 2300 may then terminate at END operation 2332.


The operational steps depicted in the above figures are offered for purposes of illustration and may be rearranged, combined into other steps, used in parallel with other steps, etc., according to embodiments of the present disclosure. Fewer or additional steps may be used in embodiments without departing from the spirit and scope of the present disclosure. Also, steps (and any sub-steps), such as priming, conditioning media, loading cells, for example, may be performed automatically in some embodiments, such as by a processor executing pre-programmed tasks stored in memory, in which such steps are provided merely for illustrative purposes. Further, the example pump rate settings for feeding cells depicted in FIG. 10B, for example, are offered for purposes of illustration. Other pump rates, flow rates, directions, etc. may be used in accordance with embodiments of the present disclosure.


Examples and further description of tasks and protocols, including custom tasks and pre-programmed tasks, for use with a cell expansion system are provided in U.S. patent application Ser. No. 13/269,323 (“Configurable Methods and Systems of Growing and Harvesting Cells in a Hollow Fiber Bioreactor System,” filed Oct. 7, 2011) and U.S. patent application Ser. No. 13/269,351 (“Customizable Methods and Systems of Growing and Harvesting Cells in a Hollow Fiber Bioreactor System,” filed Oct. 7, 2011), which are hereby incorporated by reference herein in their entireties for all that they teach and for all purposes.


Next, FIG. 24 illustrates example components of a computing system 2400 upon which embodiments of the present disclosure may be implemented. Computing system 2400 may be used in embodiments, for example, where a cell expansion system uses a processor to execute tasks, such as custom tasks or pre-programmed tasks performed as part of processes such as processes illustrated and/or described herein. In embodiments, pre-programmed tasks may include, follow “IC/EC Washout” and/or “Feed Cells,” for example.


The computing system 2400 may include a user interface 2402, a processing system 2404, and/or storage 2406. The user interface 2402 may include output device(s) 2408, and/or input device(s) 2410 as understood by a person of skill in the art. Output device(s) 2408 may include one or more touch screens, in which the touch screen may comprise a display area for providing one or more application windows. The touch screen may also be an input device 2410 that may receive and/or capture physical touch events from a user or operator, for example. The touch screen may comprise a liquid crystal display (LCD) having a capacitance structure that allows the processing system 2404 to deduce the location(s) of touch event(s), as understood by those of skill in the art. The processing system 2404 may then map the location of touch events to UI elements rendered in predetermined locations of an application window. The touch screen may also receive touch events through one or more other electronic structures, according to embodiments. Other output devices 2408 may include a printer, speaker, etc. Other input devices 2410 may include a keyboard, other touch input devices, mouse, voice input device, etc., as understood by a person of skill in the art.


Processing system 2404 may include a processing unit 2412 and/or a memory 2414, according to embodiments of the present disclosure. The processing unit 2412 may be a general purpose processor operable to execute instructions stored in memory 2414. Processing unit 2412 may include a single processor or multiple processors, according to embodiments. Further, in embodiments, each processor may be a multi-core processor having one or more cores to read and execute separate instructions. The processors may include general purpose processors, application specific integrated circuits (ASICs), field programmable gate arrays (FPGAs), other integrated circuits, etc., as understood by a person of skill in the art.


The memory 2414 may include any short-term or long-term storage for data and/or processor executable instructions, according to embodiments. The memory 2414 may include, for example, Random Access Memory (RAM), Read-Only Memory (ROM), or Electrically Erasable Programmable Read-Only Memory (EEPROM), as understood by a person of skill in the art. Other storage media may include, for example, CD-ROM, tape, digital versatile disks (DVD) or other optical storage, tape, magnetic disk storage, magnetic tape, other magnetic storage devices, etc., as understood by a person of skill in the art.


Storage 2406 may be any long-term data storage device or component. Storage 2406 may include one or more of the systems described in conjunction with the memory 2414, according to embodiments. The storage 2406 may be permanent or removable. In embodiments, storage 2406 stores data generated or provided by the processing system 2404.


EXAMPLES

The following description includes some examples of protocols/methods/processes that may be used with a cell expansion system, such as CES 500 (e.g., FIGS. 5A, 5B, 5C) and/or CES 600 (FIG. 6), for example, that implements aspects of the embodiments. Although specific features may be described in the examples, such examples are provided merely for illustrative and descriptive purposes. For example, while examples may provide for the expansion of T cells and/or Treg cells, other and/or additional cell types and/or combinations thereof may be used in other embodiments. Although specific parameters, features, and/or values are described, e.g., use of a CES, e.g., Quantum® Cell Expansion System, according to some embodiments, these parameters, features, and/or values, etc., are provided merely for illustrative purposes. The present disclosure is not limited to the examples and/or specific details provided herein.


Further, the examples provided herein are not intended to limit other embodiments, which may include different or additional steps, parameters, or other features. The example methods or protocols, including the steps (and any sub-steps), may be performed automatically in some embodiments, such as by a processor executing pre-programmed tasks stored in memory. In other embodiments, the steps (and any sub-steps) may be performed through the combination of automated and manual execution of operations. In further embodiments, the steps (and any sub-steps) may be performed by an operator(s) or user(s) or through other manual means.


While example data may be provided in such examples, such example data are provided for illustrative purposes and are not intended to limit other embodiments, which may include different steps, parameters, values, materials, or other features.


Example 1

Methods


General Treg Cell Culture


Immunomagnetic-isolated CD4+CD25+ Tregs may be acquired from healthy adult donor peripheral blood by leukapheresis (HemaCare Corporation, Van Nuys, Calif.) and may be subsequently expanded at Terumo BCT at a concentration of 1.0×105 cells/mL in sterile-filtered TexMACS™ GMP Medium supplemented using three T25 flasks (7 mL/flask) with the recombinant human IL-2 IS Premium grade cytokine at 200 IU/mL (Miltenyi Biotec GmbH, Bergisch Gladbach) and Gibco PSN 100X antibiotic mixture (ThermoFisher Scientific, Waltham, Mass.). The actively growing Treg cell suspension may be subsequently used as the inoculum in each of the three (3) Quantum Cell Expansion System experimental runs. Tregs for both the inoculum and the Quantum System expansion may be co-stimulated using a soluble tetrameric Immunocult™ human antibody CD3/CD28/CD2 cell activator complex (Stem Cell Technologies, Vancouver, BC) at 25 μL/mL in the absence of microbeads. Co-stimulation may be performed on Days 0 and 9 for the Treg inoculum and on Day 0 for the Quantum System Treg expansion. The Quantum System HFM bioreactor may be characterized by an intracapillary loop volume of 177.1 mL and surface area of 21,000 cm2.


Quantum System Treg Expansion


According to embodiments, two (2 L) bags of sterile filtered media may be prepared for the Treg scale up expansion in the Quantum System using the Quantum Media Bag 4 L Set (Cat. 21021). One 2 L bag of complete media containing TexMACS GMP, IL-2 and PSN antibiotics may be used to supply the IC compartment and one 2 L of base media containing TexMACS GMP and PSN antibiotics may be used to supply the EC inlet compartment of the bioreactor. After priming the Quantum System with PBS (Lonza Cat. 17-516Q, Walkersville, Md.), media bags may be connected to the appropriate IC and EC inlet lines using the TSCD-Q Terumo Sterile Welder. The complete media may be protected from exposure to light.


The total cell load for each run (4.5-6.5×107 Tregs) may be resuspended, using aseptic technique, in 50 mL of complete medium with a Quantum Cell Inlet Bag (Cat. 21020) for introduction into the Quantum System bioreactor. Additional disposable bags, such as the Quantum CES Media Bag 4 L (Cat. 21021) and Waste Bag 4 L (Cat. 21023) may also be used during the Treg scale-up expansion runs.


At the completion of the “Load Cells Centrally without Circulation” Task, the Quantum System runs (n=3) may be seeded with Tregs at a concentration of 2.5-3.7×105 cells/mL in 177 mL of complete medium or an average of 2.1-3.1×103 cells/cm2 within the lumen or the intracapillary (IC) compartment of the hollow fiber membrane bioreactor.


Day 0-4:


Example Quantum Custom Task


Feed Cells, Modified.


IC/EC Exchange & Condition Media for Regulatory T Cells, Example.


Attach the TexMACS GMP Complete Medium with IL-2 supplement (200 IU/mL) Media bag to the IC Media line of the Quantum System with the Terumo BCT TSCD-Q sterile welder. Attach TexMACS Base Media to EC Media line. Perform IC/EC Washout and Condition Media Tasks respectively. Complete Media may be used for IC Exchange or Washout and Base Media may be used for the EC Exchange or Washout to conserve the amount of IL-2 and activator complex.


Place system on modified “Feed Cells” prior to introducing cells. Increase the IC inlet rate (Q1) and IC circulation rate (Q2) in a matched rate to 0.2, 0.3, and 0.4 mL/min on Days 5, 6, and 7, but opposite direction on Days 4, 5, 6 or as needed to keep the lactate level between 5-8 mmol/L.









TABLE 1







Feed Cells, Modified, Example.


Table 1: Feed Cells










Setting
Step 1







IC Inlet
IC Media



IC Inlet Rate


0.1





IC Circulation Rate


−0.1




EC Inlet
None



EC Inlet Rate
 0.00



EC Circulation Rate


100
   




Outlet
EC Waste



Rocker Control


No Motion:
()




Stop Condition
Manual



Estimated Fluid
Unknown



Omit or Include
Include










Example Quantum Custom Task:


Load Cells Centrally without Circulation, Example.


Purpose: This task enables suspension cells to be centrally distributed within the bioreactor membrane while allowing flow on the extracapillary (EC) circulation loop. The pump flow rate to the IC loop may be set to zero.


Prior to loading the cells into the Quantum systems using the Load Cells without Circulation, enter the modifications to the task.









TABLE 2







Load Cells Centrally without Circulation, Modifications, Example


Table 2: Solutions for Loading Suspension Cells













Volume





(estimate based on factory



Bag
Solution in Bag
default values)







Cell Inlet


50 mL


N/A



Reagent
None
N/A



IC Media


Serum-Free Media


6 mL/hour



Wash
None
N/A



EC Media
None
N/A

















TABLE 3







Load Cells Centrally without Circulation, Example


Table 3: Custom Task 8 Load Cells without Circulation











Setting
Step 1
Step 2







IC Inlet
Cell
IC Media



IC Inlet Rate


50




50





IC Circulation Rate
0
0



EC Inlet
None
None



EC Inlet Rate
   0.00
   0.00



EC Circulation Rate
30
30



Outlet


IC Waste




IC Waste





Rocker Control


In Motion:




In Motion:






(−90 to 180°)
(−90 to 180°)






Dwell Time




Dwell Time








1 sec




1 sec





Stop Condition
Empty Bag
IC Volume:





57.1 mL



Estimated Fluid
Unknown
<0.1 L



Omit or Include
Include
Include










Return to default cell feeding tasks as needed and continue with the expansion protocol using Feed Cells Task.









TABLE 4







Feed Cells, Modified, Example.


Table 4: Feed Cells










Setting
Step 1







IC Inlet
IC Media



IC Inlet Rate


0.1





IC Circulation Rate


−0.1




EC Inlet
None



EC Inlet Rate
 0.00



EC Circulation Rate


100
   




Outlet
EC Waste



Rocker Control


No Motion:
()




Stop Condition
Manual



Estimated Fluid
Unknown



Omit or Include
Include










Day 4 or Later:


Resuspension of Treg Cells During Cell Culture or Prior to Harvest, Example.


A purpose of this modified Circulation Task is to uniformly resuspend those cells that may be loosely adhered during culture or prior to initiating the Harvest Task.


In addition, this task may be used to shear Treg cell colonies every two (2) days during cell culture in order to maintain uniform cell density and nutrient diffusion beginning on or after Day 4. If the task is used to shear colonies during the culture process, the Quantum System may be returned to the modified “Feed Cells” Task.









TABLE 5







Circulation and Resuspension of Cells, Return Cells to Bioreactor, & Feed, Example


Table 5: Custom Task 6 Settings to Resuspend Settled Cells











Setting
Step 1
Step 2
Step 3
Step 4





IC Inlet


None




IC Media




IC Media




IC Media




IC Inlet Rate
0


0.1




100




0.1




IC Circulation Rate


300




−0.1



−70




−0.1



EC Inlet


None




None




None




None




EC Inlet Rate
0


0
  

0


0
  



EC Circulation Rate


100




100
   



100




100
   



Outlet


EC Outlet




EC Outlet




EC Outlet




EC Outlet




Rocker Control


In Motion:




Stationary




In Motion:




In Motion:





(−90 to 180°)

(−90 to 180°)
(−90 to 180°)





Dwell Time:





Dwell Time:




Dwell Time:







1 sec





1 sec




1 sec




Stop Condition


Time:




Time:




IC Volume




Manual







4 min




1 min




150 mL




Estimated Fluid
Unknown
0.1 L
0.2 L
Unknown


Omit or Include
Include
Include
Include
Include









Harvest Quantum Harvest Task with Modification, Example.









TABLE 6







Harvest, Modification, Example


Table 6: Harvest, Modified











Setting
Step 1
Step 2







IC Inlet


None


IC Media



IC Inlet Rate
0
400 



IC Circulation Rate


300


−70 



EC Inlet


None




IC Media





EC Inlet Rate
0
60



EC Circulation Rate


100


30



Outlet


EC Outlet


Harvest



Rocker Control


In Motion:


In Motion:




(−90° to 180°,)
(−90° to 180°)






Dwell Time:


Dwell Time:






1 sec


1 sec



Stop Condition


Time:


IC Volume:






4 minutes


378 mL



Estimated Fluid


IC Media: <0.1 L


IC Media: 0.5 L



Omit or Include


Include


Include










Harvested cells may be removed from the Quantum System by RF welding for further evaluation and analysis.


Post-Harvest Analysis


Harvested cells may be enumerated with a Vi-CELL XR 2.04 Cell Viability Analyzer (Beckman Coulter) over a range of 5-50 μm and quantified for membrane integrity by trypan blue dye exclusion.


Metabolism


Regulatory T cell metabolism may be monitored from the Quantum EC sample port daily by i-STAT handheld analyzer (Abbott Point of Care, Princeton, N.J.) using G Cartridge for glucose and lactate concentrations using i-STAT G Cartridge (Cat. 03P83-25) and i-STAT CG4+ Cartridge (Cat. 03P85-50) respectively.


Cell Surface Biomarker Expression


Human Regulatory T cells (natural and induced) compose a small subset (2-10%) of all T cells in human cord blood and peripheral blood. Functionally, Tregs are responsible for maintaining immunological homeostasis which includes the modulation of immune tolerance in both innate and adoptive responses. Moreover, the expression of the transcriptional regulator forkhead box P3 (FoxP3) gene product is known to correlate with the CD4+CD25+FoxP3+CD127lo/− Treg phenotype and the immune suppression of antigen presentation cells (APCs) and effector T cells (Teff). IL-2 binding to the CD25/IL-2 receptor (Rα) and the activation of STATS transcriptional factor is used for Foxp3 induction. FoxP3 suppression upregulates the activity of several genes such as CTLA-4, TNFRSF18 and IL2RA and downregulates IL-2 via its association with histone acetylase KAT5 and histone deacetylase HDAC7.


Treg phenotype frequency of the harvested cell surface biomarkers may be quantified by flow cytometry. To this end, the cells may be stained with the following antibody conjugates and gated against viable, unstained cells: Fixable Viability Dye eFluor® 780 (eBioscience 65-0865), mouse anti-human CD4-PE (BD Pharmingen 561844), anti-CD4-Alexa Fluor 647 (BD Pharmingen 557707), anti-CD4-FITC (BD Pharmingen 561842), anti-CD4-FITC (BD Pharmingen 555346), anti-CD25-PE (BD Pharmingen 555432), anti-CD127-PE (BD Pharmingen 557938), anti-CD45RO-PE (BD Pharmingen 347967), and anti-FoxP3-Alexa Fluor 647 (BD Pharmingen 560045). Specimen data may be acquired on a bead-compensated BD Canto II flow cytometer equipped with FACSDiva v6.1.3 software using 1×106 cells and 20,000 total events per sample.


Experimental Flow, Example. Referring to FIG. 25, an example of experimental flow according to the present disclosure is depicted. The experimental flow example begins with Treg Cell Source using, for example, Peripheral Blood from Leukopheresis. Immunomagnetic Bead Isolation occurs next using, for example. CD4*CD25* T cells. Resuspension occurs next using, for example. TexMACS Medium+rhlL-2+PSN. Co-stimulation occurs next using, for example, Soluble Anti-CD3/CD28/CD2 Activator Complex. Last, the Quantum System is utilized to perform, for example, Treg Cell Expansion and Harvest≤14 Days using, for example, Lactate, Glucose, and O2.


Possible Results


Preliminary studies with Tregs in static culture may show that these cells tend to form micro-colonies on the order of 100 μm in diameter. Separating these cells during the medium exchange process every two days may help to limit cellular necrosis and return the cells to a high density, single cell suspension by using a 1,000 μL pipet tip that has an ID of 762 μm. Alternatively, the process of maintaining a single cell suspension may be accomplished more efficiently in an automated HFM bioreactor where the fiber lumen ID is on the order of 200 μm, such as in the Quantum System, with the aid of a preprogrammed, daily circulation task. In addition, this automated feeding task may reduce the likelihood of contamination while maintaining continuous nutrient flow to the Treg culture since it may be performed in a functionally closed system.


Possible Treg Cell Density and Viability









TABLE 7





T25 Flask Possible Harvest


















Cell Density
3.11 × 106 cells/mL 




8.71 × 105 cells/cm2



Viability
81.90%



Stimulation Cycles
2  



Doublings
4.9

















TABLE 8







Quantum Possible Harvest









Cell Density














Harvest Bag
3.61 × 106 cells/m



Bioreactor
 1.24 × 107 cells/mL




≥7.33 × 104 cells/cm2



Viability
84.80%



Stimulation Cycles
1  



Doublings
4.6



Doubling Time
41.6 hours










Preliminary Cell Seeding Density Experiments


In preparation for the expansion of immunomagnetic selected cells from the Donor in the automated bioreactor, a series of static growth experiments may be performed to determine if stimulated Tregs may be cultured at a seeding density of less than 1.0×106 cells/mL. This portion of the study may be performed by seeding 18 wells of a 24-well tissue culture plate 1.0×105 cells/mL or well in TexMACS GMP medium supplemented with IL-2 (200 IU/mL) and PSN antibiotics. These cells may also be co-stimulated with the soluble anti-CD3/CD28/CD2 mAb complex on Day 0 and Day 9 at 25 μL/mL. Cells may be manually harvested and counted on Day 14 by Vi-CELL XR.









TABLE 9







Summary of possible Treg cell seeding static plate test.
















Average





Treg Plate
Average Treg
Average Treg
Treg





Samples
Seeding
Harvest
Harvest


Treg Plate Harvest


(1 mL
Day 0
Day 14
Day14
Treg Cell
Treg Cell
Cell Viability


each)
(viable cells/mL)
(viable cells/mL)
(cells/cm2)
DS
DT (hours)
(Trypan Blue Exclusion)





n = 18
1.00 × 105
2.65 × 106
1.33 × 106
4.7
71.0
61.5%




SE 4.14 × 104





Abbreviations:


Doublings (DS),


Doubling Time (DT).






After harvest, the cell samples may be pooled for Treg biomarker analysis by flow cytometry. Possible results may show that the frequencies of CD4+C25+, CD4+CD127, and CD4+FoxP3+ phenotype may be respectively 90.9%, 79.7%, and 31.6% in static culture. The CD4+CD25+ phenotype (>70%) is generally the most reliable determinant for Treg biomarker identification since FoxP3+ detection is highly dependent on the permeabilization method and cell viability.


The data from this static plate test may suggest that human Tregs may be expanded, with cell seeding densities on the order of 105 cells/mL, when cultured in the presence of a soluble co-stimulation anti-CD3/CD28/CD2 mAb complex and serum-free medium.


Treg Metabolism


Regulatory T cells are dependent on mitochondrial metabolism and have the ability to oxidize multiple carbon sources, i.e., lipid or glucose. Tregs are known to shift their metabolism from fatty acid oxidation (FAO) to glycolysis when they enter a highly proliferative state as a result of mTOR regulation of glycolysis and fatty acid metabolism. Moreover, it has been shown that glycolysis may be necessary for the generation and suppressive functionality of human inducible Tregs by modulating the expression of FoxP3 variants through IL-2/STATS/enolase-1 promoter signaling and 2-Deoxy-D-glucose inhibition studies. Accordingly, monitoring the glucose and lactate levels may facilitate the adjustment of Quantum System media flow rates to support Treg expansion in the hollow fiber bioreactor. Initially, the Tregs may be thought to transiently reduce their metabolic rate before they enter the cell cycle and proliferate. This may be supported by the transient reduction of glycolysis and mTOR activity in freshly isolated human Tregs before TCR stimulation. Specifically, mTORC1 may be thought to increase the expression of glucose transporters such as Glut-1-mediated glucose transport as a consequence of the upregulated mTOR pathway.


The possible results of three (3) expansions from three separate Treg cell aliquots may indicate that the glucose consumption and lactate generate may appear to correlate within each Quantum System run. All three of the ex vivo expansion runs may show that glucose consumption in Tregs may increase above background levels by Day 1 and 2 out of 3 runs may show that the lactate generation levels may increase above background levels by Day 2. One run, with reduced cell viability at thaw, may generate a lagging lactate generation rate which may be reflected in the cell harvest yield. Maximum glucose consumption rates for the 2 out of 3 runs, in the most actively growing Treg cultures, may be 1.618 and 2.342 mmol/day on Days 8 and 7 respectively. The maximum lactate generation rates may be 2.406 and 3.156 mmol/day at the same time points.


Throughout the Treg expansion runs, an effort may be made to control the lactate values at 7 mmol/L by concurrently increasing both the IC Input (+) and IC circulation (−) pump rates from (±0.1 to ±0.4 mL/min) within the lumen of the hollow fiber membrane over Days 4-8. The lowest glucose levels during the course of the Treg cell expansions may range from 264 mg/dL on Day 7 (Q1584) to 279 mg/dL on Day 8 (Q1558). The base glucose concentration, in the cell growth formulated medium for these feasibility expansions, may be 325 to 335 mg/dL which may be found to be supportive when used in conjunction with the Quantum System flow rate adjustments.


Regulatory T Cell Biomarker Expression


The evaluation of the Treg cell harvest by flow cytometry may be centered on the CD4+CD25+FoxP3+ T cell subsets in this feasibility study. In T lymphocytes, the human CD4 gene, on Chromosome 12, encodes for a membrane glycoprotein which interacts with the major histocompatibility complex class II and functions to initiate the early phase of T cell activation. In regulatory T cells, the human CD25 (IL2R) gene, on Chromosome 10, encodes for the IL-2 receptor and functions by sequestering the cytokine IL-2. In regulatory T cells, the forkhead/winged-helix box P3 human gene, on the Chromosome X, encodes for the FoxP3 transcriptional factor which may be essential for Treg suppressor function. FoxP3 gene product binds to the promoter region of CD25, CTLA-4 and IL-2, IL7R, IFN-γ genes thereby upregulating CD25 and CTLA-4 and repressing IL-2, IL7R, and IFN-γ gene transcription. The CD127 gene encodes for the IL-7 receptor and Tregs may be generally characterized by low CD127 (IL-7R) expression when compared to conventional T cells. However, certain Treg subsets are known to express high CD127 levels during in vitro and in vivo activation which may correlate to a higher Treg survival when the cells are incubated with IL-7. The CD45RO gene product is expressed on naive thymus derived Tregs that upon activation lose CD45RA and express CD45RO.









TABLE 10







Possible regulatory T cell biomarker expression


as a percent of parent population.













Quantum System
Seeding
Harvest






Expansion Run
Viability
Viability
CD4+CD45RO+
CD4+CD25+
CD4+CD127low
CD4+FoxP3+
















Q1558
81.9%
84.8%
72.4%
86.7%
40.1%
58.2%


Q1567*
49.6%
69.8%
55.3%
79.3%
74.2%
*5.6%


Q1584
90.7%
94.6%
72.8%
90.5%
41.0%
64.9%


Average (n = 3)



85.5%

*42.9%


Average (n = 2)*





*61.6%





*Low cell viability at thaw/harvest and incomplete permeabilization on Q1567 cells for FoxP3+ frequency.






The average expression of the CD4+CD25+ Treg phenotype frequency may be 85.5% in the cells harvested from the Quantum System which may compare favorably with the published CD4+CD25+ release criteria of >70%. In the 01567 Treg expansion, the elevated frequency of the CD4+CD127low population (74.2%) may be a reflection of the low cell viability in this particular thawed cell sample since these cells may be cultured only with IL-2 as a cytokine supplement, according to an embodiment. In cells expanded by the two Quantum System runs with seeding and harvest viability above 80%, the CD4+FoxP3+ expression frequency may be 61.6%. This finding may be consistent with the published release specification of 60% for FoxP3+. Furthermore, the results of the two billion cell expansions may compare favorably with the CD3+CD45+ (87.30%), CD25+ (47.76%), and FoxP3+ (59.64%) biomarker expression in the original donor Treg cell specimen which may be received from HemaCare BioResearch Products.


Additional flow cytometry analysis may be performed on cryopreserved Treg cells from the 01584 expansion run by a third-party laboratory, for example, using fluorescence Minus One (FMO) gating, different stains, and different instrumentation. FMO control is a type of gating control use to interpret cell populations by taking into account the spread of all the fluorochromes in the data plots minus the one used to quantify the frequency of a particular marker. For example, the flow results from the third-party laboratory may indicate that the CD4+CD25+ Treg cell population frequency may be 95.4% from the 01584 run which may compare favorably with the 90.5% which may be found by the Terumo BCT CES Laboratory. Incomplete staining with the alternative anti-FoxP3-PE clone stain may limit the third-party laboratory quantification of this internal biomarker, but the dot-plots may suggest that there may be a subpopulation of high expressing FoxP3+ Tregs in the 01584 specimen that may not be observed in the Control Treg cell reference sample. Although interesting, additional studies may be needed to confirm these observations.


Harvest Yield


The possible average diameter of viable (trypan blue exclusion) Treg cells at Quantum System harvest may be 10.89, 11.04, and 11.06 μm respectively across the Q1558, Q1567, and Q1584 runs over a range of 5-50 μm as defined with 50 samples for each run. This may compare to an average cell diameter of 11.91, 12.40, and 7.83 μm respectively from flasks at the time of bioreactor seeding.


These possible cell diameter data may suggest that there may be more uniformity in the diameter of the cells harvested from the Quantum System than there may be in the diameter of the cells which may be expanded in the inoculum flasks.


The Treg Quantum System possible harvest data are summarized in Table 11. Moreover, the impact of the CD4+CD25+ cell viability at the point of seeding the bioreactor may be evident when comparing the results of Q1554/1584 harvests with the Q1567 harvest. There may be a 32-41% higher viability in the bioreactor inoculum for the Q1554/1584 expansion runs versus the viability for the Q1567 run. This may be due to a variation in the original cell isolation, cryopreservation technique or the length of storage since the cell aliquots that may be used in this study (HemaCare PB425C-2; Lot 14034019) may be derived from the same donor collection on Feb. 11, 2014.









TABLE 11







Possible expansion of Treg cells from inoculum flasks to Quantum System harvests.

















Treg
Treg
Treg



Treg Viability
Treg Viability
Tregs
DS (11 Days)
DS (7-8 Days)
DT (hours)


Quantum
Flask
Quantum
Quantum
Flask
Quantum
Quantum


Run
InoculumA
HarvestA
HarvestA
InoculumB
HarvestB
HarvestB
















Q1554
81.9%
84.8%
1.82 × 109
5.0
4.8
38.5


Q1567
49.6%
69.8%
1.59 × 108
4.4
1.8
101.3


Q1584
90.7%
94.6%
1.30 × 109
4.7
4.6
35.7





Abbreviations:


DS—Population Doublings,


DT—Population Doubling Time in hours.



AHarvest data may be based on Vi-Cell XR counts with Trypan Blue for membrane integrity.




BNote: the Treg cell inoculum from flasks may receive two (2) rounds costimulation on Days −0 and −9; whereas, the Tregs which may be harvested from the Quantum Systems may receive one (1) round of costimulation on Day-0.







The objective of this feasibility study may be to determine if the Quantum System may support the expansion of Tregs in the range of 7.0×107-1.4×109 cells with commercially available supplements. Two of the three bioreactor harvests from Q1554 and Q1584 may generate an average of 1.56×109 Tregs, using a soluble anti-CD3/CD28/CD2 mAb co-stimulator complex, from a seeding density of <1.0×106 cells/mL in less than eight (8) days. This may translate into an average harvest cell density of 8.81×106 cells/mL or 7.43×104 cells/cm2 in the IC loop of the Quantum System bioreactor over the Q1554/1584 runs.


Possible Conclusions


The results of this feasibility study may be exploratory in nature and may not necessarily be designed to cover all technical options. For example, one could consider the reduction of inoculum Treg co-stimulation from two (2) to one (1) activation events. As such, the methods which may be used in the automated Quantum System expansion of immunomagnetic-isolated regulatory T cells may be open to modification. Our attempt here may be to define certain technical aspects of the culture process that may be conducive to further study in the upscale expansion of Tregs within the Quantum System platform. Within this context, the possible study findings may suggest that these possible conclusions or observations may be reasonable and may be helpful in the production of regulatory T cells for research, development, or production purposes.


Human Tregs, as identified as FoxP3+/CD25+, may be cultured and may be expanded with a soluble co-stimulatory anti-CD3/CD28/CD2 monoclonal antibody (mAb) T cell complex, in the absence of co-stimulatory mAb-coated beads, when supplemented with the cytokine IL-2 in the Quantum System automated hollow fiber bioreactor.


Human Tregs may be efficiently expanded in the Quantum system from cell seeding densities of less than 1×106 cells/mL or less than 6.6×104 cells/cm2. To this end, the objective of harvesting Tregs within the range of 7.0×108 to 1.4×109 cells in less than 14 days may be achieved with an average (n=3) of 1.09×109 total cells. An average of 85.5% of the cells may express the Treg CD4+CD25+ phenotype and an average of 42.9% may express CD4+FoxP3+ phenotype (n=3). In the two (2) billion cell Quantum System expansions, an average of 61.6% of the total cells may express the CD4+FoxP3+ phenotype. One of the three Quantum system Treg cell expansion runs may be validated for CD4+CD25+ expression by a third-party laboratory human IMSR due to the limited number of cells.


Human Tregs may be successfully cultured and may be expanded in the Quantum System by centrally seeding the cells within the lumen (IC loop) of an automated hollow fiber bioreactor.


Media IC input (+0.1 to +0.4 mL/min) and IC circulation (−0.0 to −0.4 mL/min) may be adjusted in parallel to support the Treg cell expansion process in order to maintain lactate levels 7 mmol/L and to maintain the single cell suspension of the Treg culture by shearing cell micro-colonies at an IC circulation rate of 300 mL/min through the lumen of the Quantum System HFM bioreactor under functional closed conditions.


Example 2

The tables below provide example task settings (e.g., flow rates, angular rotation, outlet, etc.) for different components (e.g., pumps, rocker, valves, etc.) of a cell expansion system over several days of performing an example protocol for the expansion of T cells. The protocol may follow the following sequence:


Day 0: Load Set, Prime, Add Media, Load Cells, and begin feeding


Day 3: Add a bolus of cytokines to the IC loop while re-distributing the cells. Begin feeding again.


Day 6: Add a bolus of cytokines to the IC loop while re-distributing the cells. Begin feeding again.


Day 9: Add a bolus of cytokines to the IC loop while re-distributing the cells. Begin feeding again.


Day 11-13: Harvest; reload remaining cells. Harvest (Day 14)


Table(s) of settings: changes made compared to example factory settings are highlighted in bold and underline









TABLE 12







IL-2 Concentration and Amount in Complete Media, Example


Complete Media









Volume
IL-2
IL-2


(mL)
(IU/mL)
(IU)





2000
200
4E+05
















TABLE 13







Volumes of Bolus Additions and IL-2 Amounts, Example


Bolus Additions










Volume
IL-2



(mL)
(IU)













Day 0 (cell load)
100
1E+05


Day 3
150
2E+05


Day 6
150
2E+05


Day 9
150
4E+05
















TABLE 14







Settings Day 0-Day 2, Example









Day 0















Load cells with Uniform
Cells
Feed



Load Cell

Suspension
in BR
cells














Expansion

IC EC
Condition media
(1e8 lymphocytes)
Custom 1


















Set
Prime
washout
Step 1
Step 2
Step 1
Step 2
Step 3
Step 1
Step 2



STEP 1
STEP 2
STEP 3
STEP 4
STEP 5
STEP 6
STEP 7
STEP 8
STEP 9
STEP 10





















Task
IC inlet
Default
Default


EC


None
None
Cell
IC
None
IC
IC Media


Settings

settings
settings


media





Media

Media




IC inlet


100
0
0
25
25
0
100
0.1



rate













IC circ


−17
100
100
150
150
200
−70
1



rate













EC inlet


EC
EC
EC
None
None
None
None
None






Media
Media
Media








EC inlet


148
0.1
0.1
0
0
0
0
0



rate













EC circ


−1.7
250
30
30
30
30
30
100



rate













Outlet


IC and
EC outlet
EC outlet
EC
EC
EC
EC
EC outlet






EC


outlet
outlet
outlet
outlet







outlet










Rocker


In
Stationary
Stationary
In
In
In
In
Stationary






Motion
(0°)
(0°)
Motion
Motion
Motion
Motion
(0°)






(−90°,


(−90°,
(−90°,
(−90°,
(−90°,







180°, 1 sec)


180°,
180°,
180°,
180°, 1 sec)










1 sec)
1 sec)
1 sec)





Stop


Exchange
Time (10 min)
Manual
Empty
IC
Time
IC
Manual



condition


(2.5 IC

(20-50 min)
bag
volume
(2 min)
Volume
(4320 min)






volume;



(22 ml)

(120 mL)







2.5 EC













volume)
























Extra
Necessary
NA
2 L
1300 mL
6 mL/hr EC Media
100 mL
22 mL

120 mL
432 mL


Information
volume

(PBS)
EC


IC


IC Media






Media


Media



















Time
10 min
35 min
5 min
30-60 min
7 min
2 min
3 days
















TABLE 15







Settings Day 3-Day 5, Example


Day 3











Cells in





BR
Feed cells









Add Bag Contents
Custom 2












Step 1
Step 2
Step 1
Step 2



STEP 11
STEP 12
STEP 13
STEP 14















Task
IC inlet
Reagent
IC Media
IC Media
IC Media


Settings
IC inlet
30
30
100
 0.1



rate



IC circ


100




100


−70
−0.1



rate



EC inlet
None
None
None
None



EC inlet
 0
 0
 0
0 



rate



EC circ


100




100


 30
150   



rate



Outlet
EC outlet
EC outlet
EC outlet
EC outlet



Rocker


In Motion




In Motion


In Motion
In Motion




(−90°, 180°,
(−90°, 180°,
(−90°, 180°,
(0°, 180°,






1 sec
)



1 sec
)

1 sec)
3600 sec)



Stop
Empty
IC Volume
IC Volume
Manual



condition
Bag
(22 mL)
(120 mL)
(4320 min)


Extra
Necessary
150 mL
22 mL IC
120 mL
432 mL IC


Informati
volume

Media

Media












Time
5 min
2 min
3 days

















TABLE 16







Settings Day 6-Day 8, Example









Day 6














Cells in






Add Bag Contents
BR
Feed cells
Feed cells
Feed cells











Step 1

Custom 3














STEP
Step 2
Step 1
Step 2
Step 3
Step 4



15
STEP 16
STEP 17
STEP 18
STEP 20
STEP 22

















Task Settings
IC inlet
Reagent
IC Media
IC Media
IC Media
IC Media
IC Media



IC inlet
30
30
100
0.1
0.1
0.1



rate









IC circ


100




100


−70
−0.1
−0.1
−0.1



rate









EC inlet
None
None
None
EC Media
EC Media
EC Media



EC inlet
 0
 0
0
0.1
0.2
0.3



rate









EC circ


100




100


30
200
200
250



rate









Outlet
EC
EC outlet
EC outlet
EC outlet
EC outlet
EC outlet




outlet








Rocker


In




In

Motion


In Motion
In Motion
In Motion
In Motion






Motion




(−90°,


(−90°,
(0°, 180°,
(0°, 180°,
(0°, 180°,






(−90°,




180°,

1

sec)


180°, 1 sec)
3600 sec)
3600 sec)
3600 sec)






180°,

1

sec)










Stop
Empty
IC
IC
Time
Time
Manual



condition
Bag
Volume
Volume
(1440 min)
(1440 min)






(22 mL)
(120 mL)





Extra
Necessary
150 mL
22 mL IC
120 mL
144 mL IC
144 mL IC
144 mL IC


Information
volume

Media

Media
Media
Media







144 mL
288 mL
436 mL







EC Media
EC Media
EC Media












Time
4 min
2 min
3 days
















TABLE 17







Settings Day 9-Day 10, Example









Day 9











Cells in





BR
Feed cells
Feed cells










Add Bag Contents
Custom 3













Step 1
Step 2
Step 1
Step 2
Step 3



STEP 23
STEP 24
STEP 17
STEP 18
STEP 20
















Task Settings
IC inlet
Reagent
IC Media
IC Media
IC Media
IC Media



IC inlet
30
30
100
0.1
0.1



rate








IC circ


100




100


−70
−0.1
−0.1



rate








EC inlet
None
None
None
EC Media
EC Media



EC inlet
 0
 0
0
0.1
0.2



rate








EC circ


100




100


30
200
200



rate








Outlet
EC
EC outlet
EC outlet
EC outlet
EC outlet




outlet







Rocker


In




In

Motion


In Motion
In Motion
In Motion






Motion




(−90°,


(−90°,
(0°, 180°,
(0°, 180°,






(−90°,




180°,

1

sec)


180°, 1 sec)
3600 sec)
3600 sec)






180°,

1

sec)









Stop
Empty
IC
IC
Time
Time



condition
Bag
Volume
Volume
(1440 min)
(1440 min)





(22 mL)
(120 mL)




Extra
Necessary
150 mL
22 mL IC
120 mL
144 mL IC
144 mL IC


Information
volume

Media

Media
Media







144 mL
288 mL







EC Media
EC Media












Time
4 min
2 min
2 days
















TABLE 18







Settings Day 11, Example









Harvest
















Cells in




Mix

Load cells with Uniform Suspension
BR
Feed cells












Custom 4

(Harvest Product)
Custom 2















Step 1
Harvest
Step 1
Step 2
Step 3
Step 1
Step 2



STEP 31
STEP 32
STEP 33
STEP 34
STEP 35
STEP 13
STEP 14


















Task Settings
IC inlet
None
EC
Cell
IC Media
None
IC Media
IC Media





Media








IC inlet
0


100


25
25
0
100
0.1



rate










IC circ rate
200


−20


150
150
200
−70
−0.1



EC inlet
None
EC
None
None
None
None
None





Media








EC inlet
0


100


0
0
0
0
0



rate










EC circ rate
200
30
30
30
30
30
150



Outlet
EC
Harvest
EC outlet
EC outlet
EC outlet
EC outlet
EC outlet




outlet









Rocker
In
In
In Motion
In Motion
In Motion
In Motion
In Motion




Motion
Motion
(−90°,
(−90°, 180°,
(−90°,
(−90°,
(0°, 180°,




(−90°,
(−90°,
180°, 1 sec)
1 sec)
180°, 1 sec)
180°, 1 sec)
3600 sec)




180°, 1 sec)
180°, 1 sec)








Stop
Time (3 min)


IC


Empty
IC volume
Time (2 min)
IC
Manual



condition



Volume


bag
(22 ml)

Volume
(1440 min)







(400

mL)





(120 mL)



Extra
Necessary

800 mL
(variable)
22 mL IC

120 mL
144 mL IC


Information
volume

EC
mL
Media


Media





Media



















Time
3 min
4 min
10 min
2 min
1 days
















TABLE 19







Settings Day 12, Example









Harvest
















Cells in




Mix

Load cells with Uniform Suspension
BR
Feed cells












Custom 4

(Harvest Product)
Custom 2















Step 1
Harvest
Step 1
Step 2
Step 3
Step 1
Step 2



STEP 31
STEP 32
STEP 33
STEP 34
STEP 35
STEP 13
STEP 14


















Task Settings
IC inlet
None
EC
Cell
IC Media
None
IC Media
IC Media





Media








IC inlet
0


100


25
25
0
100
0.1



rate










IC circ rate
200


−20


150
150
200
−70
−0.1



EC inlet
None
EC
None
None
None
None
None





Media








EC inlet
0


100


0
0
0
0
0



rate










EC circ rate
200
30
30
30
30
30
150



Outlet
EC
Harvest
EC outlet
EC outlet
EC outlet
EC outlet
EC outlet




outlet









Rocker
In
In
In Motion
In Motion
In Motion
In Motion
In Motion




Motion
Motion
(−90°,
(−90°, 180°,
(−90°,
(−90°,
(0°, 180°,




(−90°,
(−90°,
180°, 1 sec)
1 sec)
180°, 1 sec)
180°, 1 sec)
3600 sec)




180°, 1 sec)
180°, 1 sec)








Stop
Time (3 min)


IC


Empty
IC volume
Time (2 min)
IC
Manual



condition



Volume


bag
(22 ml)

Volume
(1440 min)







(400

mL)





(120 mL)



Extra
Necessary

800 mL
(variable)
22 mL IC

120 mL
144 mL IC


Information
volume

EC
mL
Media


Media





Media



















Time
3 min
4 min
10 min
2 min
1 days
















TABLE 20







Settings Day 13, Example









Harvest
















Cells in




Mix

Load cells with Uniform Suspension
BR
Feed cells












Custom 4

(Harvest Product)
Custom 2















Step 1
Harvest
Step 1
Step 2
Step 3
Step 1
Step 2



STEP 31
STEP 32
STEP 33
STEP 34
STEP 35
STEP 13
STEP 14


















Task Settings
IC inlet
None
EC
Cell
IC Media
None
IC Media
IC Media





Media








IC inlet
0


100


25
25
0
100
0.1



rate










In circ rate
200


−20


150
150
200
−70
−0.1



EC inlet
None
EC
None
None
None
None
None





Media








EC inlet
0


100


0
0
0
0
0



rate










EC circ rate
200
30
30
30
30
30
150



Outlet
EC
Harvest
EC
EC outlet
EC outlet
EC outlet
EC outlet




outlet

outlet







Rocker
In
In
In
In Motion
In Motion
In Motion
In Motion




Motion
Motion
Motion
(−90°, 180°,
(−90°,
(−90°,
(0°, 180°,




(−90°,
(−90°,
(−90°,
1 sec)
180°, 1 sec)
180°, 1 sec)
3600 sec)




180°, 1 sec)
180°, 1 sec)
180°, 1 sec)







Stop
Time (3 min)


IC


Empty
IC volume
Time (2 min)
IC
Manual



condition



Volume


bag
(22 ml)

Volume
(1440 min)







(400

mL)





(120 mL)



Extra
Necessary

800 mL
(variable)
22 mL IC

120 mL
144 mL IC


Information
volume

EC
mL
Media


Media





Media



















Time
3 min
4 min
10 min
2 min
1 days
















TABLE 21







Settings Day 14, Example


Harvest










Mix




Custom 4



Step 1
Harvest



STEP 31
STEP 32
















Task
IC inlet
None
EC Media



Settings
IC inlet rate
 0


100






IC circ rate
200


−20






EC inlet
None
EC Media




EC inlet rate
 0


100






EC circ rate
200
30




Outlet
EC outlet
Harvest




Rocker
In Motion
In Motion





(−90°, 180°,
(−90°, 180°,





1 sec)
1 sec)




Stop
Time


IC Volume






condition
(3 min)
(400 mL)



Extra
Necessary

800 mL



Information
volume

EC Media




Time
3 min
4 min










It will be apparent to those skilled in the art that various modifications and variations may be made to the methods and structure of the present invention without departing from its scope. Thus, it should be understood that the invention is not to be limited to the specific examples given. Rather, the invention is intended to cover modifications and variations within the scope of the following claims and their equivalents.

Claims
  • 1. A method of expanding cells in a cell expansion system, the method comprising: loading a first volume of a first fluid into the cell expansion system, wherein the first fluid comprises a plurality of cells, and wherein the cell expansion system comprises a cell growth chamber;based on the first volume of the first fluid, determining a second volume of a second fluid to load into a portion of a first fluid circulation path to position the first volume of the first fluid in a first portion of the cell growth chamber, wherein the second fluid comprises media, and wherein the first portion of the cell growth chamber comprises a central portion of the cell growth chamber;based on the determining the second volume of the second fluid, loading the second volume of the second fluid into the cell expansion system to position the first volume of the first fluid in the central portion of the cell growth chamber;loading a third volume of a third fluid for feeding the plurality of cells; andexpanding the plurality of cells.
  • 2. The method of claim 1, wherein the cell expansion system comprises a second fluid circulation path.
  • 3. The method of claim 1, wherein the second volume of the second fluid in the first fluid circulation path flows through an intracapillary space of the cell growth chamber.
  • 4. The method of claim 3, wherein a fourth fluid in the second fluid circulation path flows through an extracapillary space of the cell growth chamber.
  • 5. The method of claim 1, wherein the first volume of the first fluid is loaded without activating an intracapillary circulation pump.
  • 6. The method of claim 1, wherein the first volume of the first fluid is the same as the second volume of the second fluid.
  • 7. The method of claim 1, wherein the first volume of the first fluid is different from the second volume of the second fluid.
  • 8. The method of claim 1, wherein the cell growth chamber comprises a hollow fiber membrane.
  • 9. The method of claim 1, further comprising: stimulating the plurality of cells with an activator to activate the plurality of cells.
  • 10. The method of claim 9, wherein the activator comprises a soluble activator.
  • 11. The method of claim 9, wherein the activator comprises one or more beads.
  • 12. The method of claim 1, wherein the plurality of cells comprises non-adherent cells.
  • 13. The method of claim 12, wherein the plurality of cells comprises T cells.
  • 14. The method of claim 13, wherein the plurality of cells comprises a subpopulation of T cells.
  • 15. The method of claim 14, wherein the subpopulation of T cells comprises regulatory T cells (Tregs).
  • 16. The method of claim 1, wherein the loading the first volume of the first fluid comprises loading the first volume of the first fluid without activating a circulation pump.
  • 17. The method of claim 1, further comprising: determining to reposition the first volume of the first fluid in the cell growth chamber; andbased on the determining to reposition the first volume of the first fluid, loading a fourth volume of a fourth fluid into the first fluid circulation path.
  • 18. The method of claim 1, wherein the second volume of the second fluid includes a first amount of the second fluid in a first portion of the first fluid circulation path.
  • 19. The method of claim 18, wherein the second volume of the second fluid includes a second amount of the second fluid in a second portion of the first fluid circulation path.
  • 20. The method of claim 19, wherein the second portion of the first fluid circulation path comprises an intracapillary space between an inlet port of the cell growth chamber and the central portion of the cell growth chamber.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to, and the benefit of, the following applications: U.S. Provisional Application Ser. No. 62/479,721, filed on Mar. 31, 2017, and entitled, “Expanding Cells;” U.S. Provisional Application Ser. No. 62/479,760, filed on Mar. 31, 2017, and entitled “Cell Expansion;” U.S. Provisional Application Ser. No. 62/479,788, filed on Mar. 31, 2017, and entitled, “Expanding Cells in a Bioreactor;” U.S. Provisional Application Ser. No. 62/549,871, filed on Aug. 24, 2017, and entitled, “Expansion of Cells;” and U.S. Provisional Application Ser. No. 62/647,361, filed on Mar. 23, 2018, and entitled, “Expansion of Cells.” The disclosures of the above-identified applications are hereby incorporated by reference in their entireties as if set forth herein in full for all that they teach and for all purposes.

US Referenced Citations (1075)
Number Name Date Kind
2997077 Rodrigues Aug 1961 A
3013435 Rodrigues Dec 1961 A
3067915 Shapiro et al. Dec 1962 A
3191807 Rodrigues Jun 1965 A
3283727 Rodrigues Nov 1966 A
3701717 Ingvorsen Oct 1972 A
3821087 Knazek et al. Jun 1974 A
3896061 Tanzawa et al. Jul 1975 A
4173415 Wyatt Nov 1979 A
4301010 Eddleman et al. Nov 1981 A
4301118 Eddleman et al. Nov 1981 A
4391912 Yoshida et al. Jul 1983 A
4412990 Lundblad et al. Nov 1983 A
4418691 Yannas et al. Dec 1983 A
4439322 Sonoda et al. Mar 1984 A
4439901 Eddleman Apr 1984 A
4440853 Michaels et al. Apr 1984 A
4478829 Landaburu et al. Oct 1984 A
4486188 Altshuler et al. Dec 1984 A
4509695 Bessman Apr 1985 A
4585654 Landaburu et al. Apr 1986 A
4618586 Walker et al. Oct 1986 A
4629686 Gruenberg Dec 1986 A
4647539 Bach Mar 1987 A
4650766 Harm et al. Mar 1987 A
4670544 Schwinn et al. Jun 1987 A
4722902 Harm et al. Feb 1988 A
4727059 Binder et al. Feb 1988 A
4789658 Yoshimoto et al. Dec 1988 A
4804628 Cracauer et al. Feb 1989 A
4828706 Eddleman May 1989 A
4885087 Kopf Dec 1989 A
4889812 Guinn et al. Dec 1989 A
4894342 Guinn et al. Jan 1990 A
4897358 Carrasco Jan 1990 A
4910139 Chang et al. Mar 1990 A
4918019 Guinn Apr 1990 A
4960521 Keller Oct 1990 A
4973558 Wilson et al. Nov 1990 A
4988623 Schwarz et al. Jan 1991 A
5015585 Robinson May 1991 A
5019054 Clement et al. May 1991 A
5079168 Amiot Jan 1992 A
5081035 Halberstadt Jan 1992 A
5126238 Gebhard et al. Jun 1992 A
5130141 Law et al. Jul 1992 A
5149544 Gentile et al. Sep 1992 A
5156844 Aebischer et al. Oct 1992 A
5162225 Sager et al. Nov 1992 A
5169930 Ruoslahti et al. Dec 1992 A
5192553 Boyse et al. Mar 1993 A
5197985 Caplan et al. Mar 1993 A
5202254 Amiot et al. Apr 1993 A
5225346 Matsumiya et al. Jul 1993 A
5226914 Caplan et al. Jul 1993 A
5240614 Ofsthun et al. Aug 1993 A
5240861 Bieri Aug 1993 A
5252216 Folena-Wasserman et al. Oct 1993 A
5283058 Faustman Feb 1994 A
5310676 Johansson et al. May 1994 A
5324428 Flaherty Jun 1994 A
5330915 Wilson et al. Jul 1994 A
5342752 Platz et al. Aug 1994 A
5399493 Emerson et al. Mar 1995 A
5416022 Amiot May 1995 A
5422197 Zito Jun 1995 A
5436151 McGlave et al. Jul 1995 A
5437994 Emerson et al. Aug 1995 A
5439757 Zito Aug 1995 A
5459069 Palsson et al. Oct 1995 A
5460964 McGlave et al. Oct 1995 A
H1509 Eran et al. Dec 1995 H
5478739 Slivka et al. Dec 1995 A
5486359 Caplan et al. Jan 1996 A
5496659 Zito Mar 1996 A
5507949 Ho Apr 1996 A
5510257 Sirkar et al. Apr 1996 A
5512180 Ho Apr 1996 A
5527467 Ofsthun et al. Jun 1996 A
5541105 Melink et al. Jul 1996 A
5543316 Zawadzka et al. Aug 1996 A
5545492 Zito Aug 1996 A
5549674 Humes et al. Aug 1996 A
5571720 Grandies et al. Nov 1996 A
5581687 Lyle et al. Dec 1996 A
5591625 Gerson et al. Jan 1997 A
5593580 Kopf Jan 1997 A
5595909 Hu et al. Jan 1997 A
5599703 Davis et al. Feb 1997 A
5605822 Emerson et al. Feb 1997 A
5605829 McGlave et al. Feb 1997 A
5605835 Hu et al. Feb 1997 A
5622857 Goffe Apr 1997 A
5626731 Cooley et al. May 1997 A
5627070 Gruenberg May 1997 A
5631006 Melink et al. May 1997 A
5635386 Palsson et al. Jun 1997 A
5635387 Fei et al. Jun 1997 A
5643736 Bruder et al. Jul 1997 A
5646043 Emerson et al. Jul 1997 A
5653887 Wahl et al. Aug 1997 A
5654186 Cerami et al. Aug 1997 A
5656421 Gebhard et al. Aug 1997 A
5658995 Kohn et al. Aug 1997 A
5667985 O'Leary et al. Sep 1997 A
5670147 Emerson et al. Sep 1997 A
5670351 Emerson et al. Sep 1997 A
5674750 Kraus et al. Oct 1997 A
5684712 Goffe et al. Nov 1997 A
5686289 Humes et al. Nov 1997 A
5688687 Palsson et al. Nov 1997 A
5695989 Kalamasz Dec 1997 A
5700289 Breitbart et al. Dec 1997 A
5705534 D'Agostino et al. Jan 1998 A
5707859 Miller et al. Jan 1998 A
5712163 Parenteau et al. Jan 1998 A
5728581 Schwartz et al. Mar 1998 A
5733541 Taichman et al. Mar 1998 A
5733542 Haynesworth et al. Mar 1998 A
5736396 Bruder et al. Apr 1998 A
5744347 Wagner et al. Apr 1998 A
5750651 Oppermann et al. May 1998 A
5753506 Johe May 1998 A
5763194 Slowiaczek et al. Jun 1998 A
5763197 Tsukamoto et al. Jun 1998 A
5763261 Gruenberg Jun 1998 A
5763266 Palsson et al. Jun 1998 A
5766944 Ruiz Jun 1998 A
5772994 Ildstad et al. Jun 1998 A
5783075 Eddleman et al. Jul 1998 A
5783216 Faustman Jul 1998 A
5785912 Cooley et al. Jul 1998 A
5804446 Cerami et al. Sep 1998 A
5806529 Reisner et al. Sep 1998 A
5807686 Wagner et al. Sep 1998 A
5811094 Caplan et al. Sep 1998 A
5811397 Francavilla et al. Sep 1998 A
5817773 Wilson et al. Oct 1998 A
5821218 Toback et al. Oct 1998 A
5827735 Young et al. Oct 1998 A
5827740 Pittenger Oct 1998 A
5830921 Cooley et al. Nov 1998 A
5833979 Schinstine et al. Nov 1998 A
5837258 Grotendorst Nov 1998 A
5837539 Caplan et al. Nov 1998 A
5840502 Van Vlasselaer Nov 1998 A
5840576 Schinstine et al. Nov 1998 A
5840580 Terstappen et al. Nov 1998 A
5842477 Naughton et al. Dec 1998 A
5843633 Yin et al. Dec 1998 A
5846796 Cerami et al. Dec 1998 A
5853247 Shroyer Dec 1998 A
5853717 Schinstine et al. Dec 1998 A
5855608 Brekke et al. Jan 1999 A
5855613 Antanavich et al. Jan 1999 A
5855619 Caplan et al. Jan 1999 A
5858747 Schinstine et al. Jan 1999 A
5858782 Long et al. Jan 1999 A
5861315 Nakahata Jan 1999 A
5866115 Kanz et al. Feb 1999 A
5866420 Talbot et al. Feb 1999 A
5868930 Kopf Feb 1999 A
5882295 Kope Mar 1999 A
5882918 Goffe Mar 1999 A
5882929 Fofonoff et al. Mar 1999 A
5888807 Palsson et al. Mar 1999 A
5902741 Purchio et al. May 1999 A
5906827 Khouri et al. May 1999 A
5906934 Grande et al. May 1999 A
5908782 Marshak et al. Jun 1999 A
5908784 Johnstone et al. Jun 1999 A
5912177 Turner et al. Jun 1999 A
5914108 Tsukamoto et al. Jun 1999 A
5922597 Verfaillie et al. Jul 1999 A
5922847 Broudy et al. Jul 1999 A
5925567 Kraus et al. Jul 1999 A
5928945 Seliktar et al. Jul 1999 A
5935849 Schinstine et al. Aug 1999 A
5938929 Shimagaki et al. Aug 1999 A
5939323 Valentini et al. Aug 1999 A
5942225 Bruder et al. Aug 1999 A
5955353 Amiot Sep 1999 A
5958763 Goffe Sep 1999 A
5965436 Thiede et al. Oct 1999 A
5972703 Long et al. Oct 1999 A
5980795 Klotzer et al. Nov 1999 A
5981211 Hu et al. Nov 1999 A
5981708 Lawman et al. Nov 1999 A
5985653 Armstrong et al. Nov 1999 A
5994129 Armstrong et al. Nov 1999 A
5998184 Shi Dec 1999 A
6001585 Gramer Dec 1999 A
6001643 Spaulding Dec 1999 A
6001647 Peck et al. Dec 1999 A
6004743 Kenyon et al. Dec 1999 A
6010696 Caplan et al. Jan 2000 A
6015554 Galy Jan 2000 A
6022540 Bruder et al. Feb 2000 A
6022742 Kopf Feb 2000 A
6022743 Naughton et al. Feb 2000 A
6027743 Khouri et al. Feb 2000 A
6029101 Yoshida et al. Feb 2000 A
6030836 Thiede et al. Feb 2000 A
6040180 Johe Mar 2000 A
6045818 Cima et al. Apr 2000 A
6048721 Armstrong et al. Apr 2000 A
6048727 Kopf Apr 2000 A
6049026 Muschler Apr 2000 A
6054121 Cerami et al. Apr 2000 A
6060270 Humes May 2000 A
6066317 Yang et al. May 2000 A
6071691 Hoekstra et al. Jun 2000 A
6074366 Rogers et al. Jun 2000 A
6077708 Collins et al. Jun 2000 A
6082364 Balian et al. Jul 2000 A
6083747 Wong et al. Jul 2000 A
6086643 Clark et al. Jul 2000 A
6087113 Caplan et al. Jul 2000 A
6096532 Armstrong et al. Aug 2000 A
6096537 Chappel Aug 2000 A
6103117 Shimagaki et al. Aug 2000 A
6103522 Torok-Storb et al. Aug 2000 A
6110176 Shapira Aug 2000 A
6110482 Khouri et al. Aug 2000 A
6114307 Jaspers et al. Sep 2000 A
6117985 Thomas et al. Sep 2000 A
6120491 Kohn et al. Sep 2000 A
6127141 Kopf Oct 2000 A
6129911 Faris Oct 2000 A
6143293 Weiss et al. Nov 2000 A
6146360 Rogers et al. Nov 2000 A
6146888 Smith et al. Nov 2000 A
6149902 Artavanis-Tsakonas et al. Nov 2000 A
6149906 Mosca Nov 2000 A
6150164 Humes Nov 2000 A
6152964 Van Blitterswijk et al. Nov 2000 A
6162643 Wille, Jr. Dec 2000 A
6165225 Antanavich et al. Dec 2000 A
6165785 Ogle et al. Dec 2000 A
6174333 Kadiyala et al. Jan 2001 B1
6174526 Cerami et al. Jan 2001 B1
6174666 Pavlakis et al. Jan 2001 B1
6179871 Halpern Jan 2001 B1
6197325 MacPhee et al. Mar 2001 B1
6197575 Griffith et al. Mar 2001 B1
6200606 Peterson et al. Mar 2001 B1
6214369 Grande et al. Apr 2001 B1
6214574 Kopf Apr 2001 B1
6224860 Brown May 2001 B1
6225119 Qasba et al. May 2001 B1
6225368 D'Agostino et al. May 2001 B1
6228117 De Bruijn et al. May 2001 B1
6228607 Kersten et al. May 2001 B1
6228635 Armstrong et al. May 2001 B1
6238908 Armstrong et al. May 2001 B1
6239157 Mbalaviele May 2001 B1
6242252 Reid et al. Jun 2001 B1
6248319 Zsebo et al. Jun 2001 B1
6248587 Rodgers et al. Jun 2001 B1
6255112 Thiede et al. Jul 2001 B1
6258597 Bachovchin et al. Jul 2001 B1
6258778 Rodgers et al. Jul 2001 B1
6261549 Fernandez et al. Jul 2001 B1
6280718 Kaufman et al. Aug 2001 B1
6280724 Moore Aug 2001 B1
6281012 McIntosh et al. Aug 2001 B1
6281195 Rueger et al. Aug 2001 B1
6287864 Bagnis et al. Sep 2001 B1
6291249 Mahant et al. Sep 2001 B1
6297213 Oppermann et al. Oct 2001 B1
6299650 Van Blitterswijk et al. Oct 2001 B1
6306424 Vyakarnam et al. Oct 2001 B1
6306575 Thomas et al. Oct 2001 B1
6322784 Pittenger et al. Nov 2001 B1
6322786 Anderson Nov 2001 B1
6326198 Emerson et al. Dec 2001 B1
6326201 Fung et al. Dec 2001 B1
6328765 Hardwick et al. Dec 2001 B1
6328960 McIntosh et al. Dec 2001 B1
6333029 Vyakarnam et al. Dec 2001 B1
6335195 Rodgers et al. Jan 2002 B1
6338942 Kraus et al. Jan 2002 B2
6340592 Stringer Jan 2002 B1
6342370 Connolly et al. Jan 2002 B1
6355239 Bruder et al. Mar 2002 B1
6358252 Shapira Mar 2002 B1
6361997 Huss Mar 2002 B1
6365149 Vyakarnam et al. Apr 2002 B2
6368636 McIntosh et al. Apr 2002 B1
6372210 Brown Apr 2002 B2
6372244 Antanavich et al. Apr 2002 B1
6372494 Naughton et al. Apr 2002 B1
6372892 Ballinger et al. Apr 2002 B1
6376742 Zdrahala et al. Apr 2002 B1
6379953 Bruder et al. Apr 2002 B1
6387367 Davis-Sproul et al. May 2002 B1
6387369 Pittenger et al. May 2002 B1
6387693 Rieser et al. May 2002 B2
6387964 D'Agostino et al. May 2002 B1
6392118 Hammang et al. May 2002 B1
6394812 Sullivan et al. May 2002 B1
6399580 Elias et al. Jun 2002 B1
6410320 Humes Jun 2002 B1
6414219 Denhardt et al. Jul 2002 B1
6416496 Rogers et al. Jul 2002 B1
6417205 Cooke et al. Jul 2002 B1
6419829 Ho et al. Jul 2002 B2
6420138 Gentz et al. Jul 2002 B1
6423681 Barasch et al. Jul 2002 B1
6426332 Rueger et al. Jul 2002 B1
6428802 Atala Aug 2002 B1
6429012 Kraus et al. Aug 2002 B1
6429013 Halvorsen et al. Aug 2002 B1
6432653 Okarma Aug 2002 B1
6432711 Dinsmore et al. Aug 2002 B1
6440407 Bauer et al. Aug 2002 B1
6440734 Pykett et al. Aug 2002 B1
6451562 Ruben et al. Sep 2002 B1
6454811 Sherwood et al. Sep 2002 B1
6455678 Yin et al. Sep 2002 B1
6458585 Vachula et al. Oct 2002 B1
6458589 Rambhatla et al. Oct 2002 B1
6461495 Morrissey et al. Oct 2002 B1
6461853 Zhu Oct 2002 B1
6464983 Grotendorst Oct 2002 B1
6465205 Hicks, Jr. Oct 2002 B2
6465247 Weissman et al. Oct 2002 B1
6465249 Reya et al. Oct 2002 B2
6468794 Uchida et al. Oct 2002 B1
6472200 Mitrani Oct 2002 B1
6475481 Talmadge Nov 2002 B2
6479064 Atala Nov 2002 B1
6482231 Abatangelo et al. Nov 2002 B1
6482411 Ahuja et al. Nov 2002 B1
6482645 Atala Nov 2002 B2
6482926 Thomas et al. Nov 2002 B1
6488925 Ruben et al. Dec 2002 B2
6491918 Thomas et al. Dec 2002 B1
6495129 Li et al. Dec 2002 B1
6495364 Hammang et al. Dec 2002 B2
6497875 Sorrell et al. Dec 2002 B1
6498034 Strobl Dec 2002 B1
6506574 Rambhatla et al. Jan 2003 B1
6511510 de Bruijn et al. Jan 2003 B1
6511767 Calver et al. Jan 2003 B1
6511958 Atkinson et al. Jan 2003 B1
6514514 Atkinson et al. Feb 2003 B1
6524452 Clark et al. Feb 2003 B1
6528052 Smith et al. Mar 2003 B1
6528245 Sanchez-Ramos et al. Mar 2003 B2
6531445 Cohen et al. Mar 2003 B1
6534084 Vyakarnam et al. Mar 2003 B1
6537807 Smith et al. Mar 2003 B1
6541024 Kadiyala et al. Apr 2003 B1
6541249 Wager et al. Apr 2003 B2
6544506 Reisner Apr 2003 B2
6548734 Glimcher et al. Apr 2003 B1
6555324 Olweus et al. Apr 2003 B1
6555374 Gimble et al. Apr 2003 B1
6559119 Burgess et al. May 2003 B1
6562616 Toner et al. May 2003 B1
6565843 Cohen et al. May 2003 B1
6566126 Cadwell May 2003 B2
6569421 Hodges May 2003 B2
6569427 Boyse et al. May 2003 B1
6569428 Isner et al. May 2003 B1
6569654 Shastri et al. May 2003 B2
6576188 Rose et al. Jun 2003 B1
6576428 Assenmacher et al. Jun 2003 B1
6576464 Gold et al. Jun 2003 B2
6576465 Long Jun 2003 B1
6582471 Bittmann et al. Jun 2003 B1
6582955 Martinez et al. Jun 2003 B2
6586192 Peschle et al. Jul 2003 B1
6589728 Csete et al. Jul 2003 B2
6589786 Mangano et al. Jul 2003 B1
6593123 Wright et al. Jul 2003 B1
6596274 Abatangelo et al. Jul 2003 B1
6599300 Vibe-Hansen et al. Jul 2003 B2
6599520 Scarborough et al. Jul 2003 B2
6610535 Lu et al. Aug 2003 B1
6613798 Porter et al. Sep 2003 B1
6616912 Eddleman et al. Sep 2003 B2
6617070 Morrissey et al. Sep 2003 B1
6617152 Bryhan et al. Sep 2003 B2
6617159 Cancedda et al. Sep 2003 B1
6623749 Williams et al. Sep 2003 B2
6623942 Ruben et al. Sep 2003 B2
6624108 Clark et al. Sep 2003 B1
6626950 Brown et al. Sep 2003 B2
6627191 Bartelmez et al. Sep 2003 B1
6629003 Frizzell et al. Sep 2003 B1
6632425 Li et al. Oct 2003 B1
6632620 Makarovskiy Oct 2003 B1
6632934 Moreadith et al. Oct 2003 B1
6638765 Rosenberg Oct 2003 B1
6642019 Anderson et al. Nov 2003 B1
6642048 Xu et al. Nov 2003 B2
6642049 Chute et al. Nov 2003 B1
6642201 Khavinson et al. Nov 2003 B1
6645489 Pykett et al. Nov 2003 B2
6645727 Thomas et al. Nov 2003 B2
6645763 Kobayashi et al. Nov 2003 B2
6649189 Talmadge et al. Nov 2003 B2
6649595 Clackson et al. Nov 2003 B2
6649631 Orme et al. Nov 2003 B1
6653105 Triglia et al. Nov 2003 B2
6653134 Prockop et al. Nov 2003 B2
6660523 Blom et al. Dec 2003 B2
6662805 Frondoza et al. Dec 2003 B2
6667034 Palsson et al. Dec 2003 B2
6667176 Funk et al. Dec 2003 B1
6670169 Schob et al. Dec 2003 B1
6670175 Wang et al. Dec 2003 B2
6673603 Baetge et al. Jan 2004 B2
6673606 Tennekoon et al. Jan 2004 B1
6677306 Veis et al. Jan 2004 B1
6683192 Baxter et al. Jan 2004 B2
6685936 McIntosh et al. Feb 2004 B2
6685971 Xu Feb 2004 B2
6686198 Melton et al. Feb 2004 B1
6689324 Inoue Feb 2004 B2
6690981 Kawachi et al. Feb 2004 B1
6696575 Schmidt et al. Feb 2004 B2
6699716 Sullivan et al. Mar 2004 B2
6703017 Peck et al. Mar 2004 B1
6703209 Baetscher et al. Mar 2004 B1
6706293 Quintanilla Almagro et al. Mar 2004 B1
6709864 Pittenger et al. Mar 2004 B1
6712850 Vyakarnam et al. Mar 2004 B2
6719969 Hogaboam et al. Apr 2004 B1
6719970 Costantino et al. Apr 2004 B1
6720340 Cooke et al. Apr 2004 B1
6730314 Jeschke et al. May 2004 B2
6730315 Usala et al. May 2004 B2
6730510 Roos et al. May 2004 B2
6733746 Daley et al. May 2004 B2
6734000 Chin et al. May 2004 B2
6740493 Long et al. May 2004 B1
6759039 Tsang et al. Jul 2004 B2
6759245 Toner et al. Jul 2004 B1
6761883 Weissman et al. Jul 2004 B2
6761887 Kavalkovich et al. Jul 2004 B1
6767699 Polo et al. Jul 2004 B2
6767737 Wilson et al. Jul 2004 B1
6767738 Gage et al. Jul 2004 B1
6767740 Sramek et al. Jul 2004 B2
6770478 Crowe et al. Aug 2004 B2
6777227 Ricci et al. Aug 2004 B2
6777231 Katz et al. Aug 2004 B1
6780612 Ford et al. Aug 2004 B1
6787355 Miller et al. Sep 2004 B1
6790455 Chu et al. Sep 2004 B2
6793939 Badylak Sep 2004 B2
6797269 Mosca et al. Sep 2004 B2
6797514 Berenson et al. Sep 2004 B2
6800480 Bodnar et al. Oct 2004 B1
6802971 Gorsuch et al. Oct 2004 B2
6805860 Alt Oct 2004 B1
6809117 Enikolopov et al. Oct 2004 B2
6811773 Gentz et al. Nov 2004 B1
6811776 Kale et al. Nov 2004 B2
6814961 Jensen et al. Nov 2004 B1
6821513 Fleming Nov 2004 B1
6821790 Mahant et al. Nov 2004 B2
6828145 Avital et al. Dec 2004 B2
6833269 Carpenter Dec 2004 B2
6835377 Goldberg et al. Dec 2004 B2
6835566 Smith et al. Dec 2004 B2
6838284 de Bruijn et al. Jan 2005 B2
6841150 Halvorsen et al. Jan 2005 B2
6841151 Stringer Jan 2005 B2
6841294 Morrissey et al. Jan 2005 B1
6841355 Livant Jan 2005 B2
6841386 Kraus et al. Jan 2005 B2
6841542 Bartelmez et al. Jan 2005 B2
6844011 Faustman Jan 2005 B1
6844187 Weschler et al. Jan 2005 B1
6849051 Sramek et al. Feb 2005 B2
6849255 Gazit et al. Feb 2005 B2
6849454 Kelly et al. Feb 2005 B2
6849662 Enikolopov et al. Feb 2005 B2
6852308 Kohn et al. Feb 2005 B2
6852321 Colucci et al. Feb 2005 B2
6852533 Rafii et al. Feb 2005 B1
6855242 Comninellis et al. Feb 2005 B1
6855542 DiMilla et al. Feb 2005 B2
6863900 Kadiyala et al. Mar 2005 B2
6866843 Habener et al. Mar 2005 B2
6872389 Faris Mar 2005 B1
6875430 McIntosh et al. Apr 2005 B2
6887600 Morrissey et al. May 2005 B2
6887704 Peled et al. May 2005 B2
6908763 Akashi et al. Jun 2005 B1
6911201 Merchav et al. Jun 2005 B1
6914279 Lu et al. Jul 2005 B2
6939955 Rameshwar Sep 2005 B2
6943008 Ma Sep 2005 B1
6944522 Karmiy et al. Sep 2005 B2
6965018 Mikesell et al. Nov 2005 B2
6969308 Doi et al. Nov 2005 B2
6979308 McDonald et al. Dec 2005 B1
6979321 Geis et al. Dec 2005 B2
6988004 Kanno et al. Jan 2006 B2
7008394 Geise et al. Mar 2006 B2
7015037 Furcht et al. Mar 2006 B1
7029666 Bruder et al. Apr 2006 B2
7033339 Lynn Apr 2006 B1
7033823 Chang Apr 2006 B2
7041493 Rao May 2006 B2
7045098 Stephens May 2006 B2
7052517 Murphy et al. May 2006 B2
7056493 Kohn et al. Jun 2006 B2
7112441 Uemura et al. Sep 2006 B2
7118672 Husain et al. Oct 2006 B2
7122178 Simmons et al. Oct 2006 B1
7160719 Nyberg Jan 2007 B2
7169295 Husain et al. Jan 2007 B2
7172696 Martinez et al. Feb 2007 B1
7175763 Husain et al. Feb 2007 B2
7192776 Stephens Mar 2007 B2
7195711 Gorsuch et al. Mar 2007 B2
7250154 Kohn et al. Jul 2007 B2
7270996 Cannon et al. Sep 2007 B2
7271234 Kohn et al. Sep 2007 B2
7294259 Cote et al. Nov 2007 B2
7300571 Cote et al. Nov 2007 B2
7303676 Husain et al. Dec 2007 B2
7303677 Cote et al. Dec 2007 B2
7341062 Chachques et al. Mar 2008 B2
7358001 Morrissey et al. Apr 2008 B2
7361493 Hammond et al. Apr 2008 B1
7368169 Kohn et al. May 2008 B2
7378271 Bader May 2008 B2
7399872 Webster et al. Jul 2008 B2
7416884 Gemmiti et al. Aug 2008 B2
7425440 Malinge et al. Sep 2008 B2
7435586 Bartlett et al. Oct 2008 B2
7438902 Habener et al. Oct 2008 B2
7439057 Frangos et al. Oct 2008 B2
7452529 Brown, Jr. et al. Nov 2008 B2
7491388 Mc Intosh et al. Feb 2009 B1
7494811 Wolfinbarger, Jr. et al. Feb 2009 B2
7514074 Pittenger et al. Apr 2009 B2
7514075 Hedrick et al. Apr 2009 B2
7524676 Reiter et al. Apr 2009 B2
7531351 Marx et al. May 2009 B2
7534609 Merchav et al. May 2009 B2
7572374 Gorsuch et al. Aug 2009 B2
7579179 Bryhan et al. Aug 2009 B2
7585412 Gorsuch et al. Sep 2009 B2
7588938 Ma Sep 2009 B2
7598075 Smith et al. Oct 2009 B2
7608447 Cohen et al. Oct 2009 B2
7659118 Furcht et al. Feb 2010 B2
7678573 Merchav et al. Mar 2010 B2
7682822 Noll et al. Mar 2010 B2
7682823 Runyon Mar 2010 B1
7718430 Antwiler May 2010 B2
7722896 Kohn et al. May 2010 B2
D620732 Andrews Aug 2010 S
7838122 Kohn et al. Nov 2010 B2
7838289 Furcht et al. Nov 2010 B2
7892829 Pittenger et al. Feb 2011 B2
7919307 Klaus et al. Apr 2011 B2
7927587 Blazer et al. Apr 2011 B2
7989851 Lu et al. Aug 2011 B2
8008528 Kohn et al. Aug 2011 B2
8034365 Baluca Oct 2011 B2
8075881 Verfaillie et al. Dec 2011 B2
8147824 Maziarz et al. Apr 2012 B2
8147863 Kohn et al. Apr 2012 B2
8158120 Pittenger et al. Apr 2012 B2
8158121 Pittenger et al. Apr 2012 B2
8252280 Verfaillie et al. Aug 2012 B1
8252887 Bolikal et al. Aug 2012 B2
8288159 Warren et al. Oct 2012 B2
8288590 Kohn et al. Oct 2012 B2
8298823 Warren et al. Oct 2012 B2
8309347 Antwiler Nov 2012 B2
8321145 Antwiler Nov 2012 B2
8361453 Uhrich et al. Jan 2013 B2
8377683 Lu et al. Feb 2013 B2
8383397 Wojciechowski et al. Feb 2013 B2
8383806 Rameshwar Feb 2013 B2
8399245 Leuthaeuser et al. Mar 2013 B2
8415449 Kohn et al. Apr 2013 B2
8435781 Kodama May 2013 B2
8461289 Kohn et al. Jun 2013 B2
8476399 Bolikal et al. Jul 2013 B2
8486621 Luo et al. Jul 2013 B2
8486695 Danilkovitch et al. Jul 2013 B2
8492140 Smith et al. Jul 2013 B2
8492150 Parker et al. Jul 2013 B2
8524496 Meiron et al. Sep 2013 B2
8529888 Meiron et al. Sep 2013 B2
8540499 Page et al. Sep 2013 B2
8551511 Brandom et al. Oct 2013 B2
8580249 Blazar et al. Nov 2013 B2
8678638 Wong Mar 2014 B2
8785181 Antwiler Jul 2014 B2
8852570 Pittenger et al. Oct 2014 B2
8852571 Pittenger et al. Oct 2014 B2
8852572 Pittenger et al. Oct 2014 B2
8852573 Pittenger et al. Oct 2014 B2
8852574 Pittenger et al. Oct 2014 B2
8852575 Pittenger et al. Oct 2014 B2
9109193 Galliher et al. Aug 2015 B2
9220810 Ma et al. Dec 2015 B2
9260698 Antwiler Feb 2016 B2
9441195 Wojciechowski et al. Sep 2016 B2
9534198 Page et al. Jan 2017 B2
9677042 Stanton, IV et al. Jun 2017 B2
9725689 Stanton, IV et al. Aug 2017 B2
9732313 Hirschel et al. Aug 2017 B2
10093956 Hirschel et al. Oct 2018 B2
10494421 Castillo Dec 2019 B2
20010017188 Cooley et al. Aug 2001 A1
20010020086 Hubbell et al. Sep 2001 A1
20010021516 Wei et al. Sep 2001 A1
20010029046 Beaulieu Oct 2001 A1
20010033834 Wilkison et al. Oct 2001 A1
20010036663 Kraus et al. Nov 2001 A1
20010041687 Mruk Nov 2001 A1
20010044413 Pierce et al. Nov 2001 A1
20010049139 Lagasse et al. Dec 2001 A1
20020015724 Yang et al. Feb 2002 A1
20020018804 Austin et al. Feb 2002 A1
20020028510 Sanberg et al. Mar 2002 A1
20020031757 Ohgushi et al. Mar 2002 A1
20020037278 Ueno et al. Mar 2002 A1
20020045260 Hung et al. Apr 2002 A1
20020064869 Ebner et al. May 2002 A1
20020076400 Katz et al. Jun 2002 A1
20020077687 Ahn Jun 2002 A1
20020082698 Parenteau et al. Jun 2002 A1
20020116054 Lundell et al. Aug 2002 A1
20020128581 Vishnoi et al. Sep 2002 A1
20020128582 Farrell et al. Sep 2002 A1
20020128583 Min et al. Sep 2002 A1
20020128584 Brown et al. Sep 2002 A1
20020130100 Smith Sep 2002 A1
20020132343 Lum Sep 2002 A1
20020139743 Critz et al. Oct 2002 A1
20020142457 Umezawa et al. Oct 2002 A1
20020146678 Benvenisty Oct 2002 A1
20020146817 Cannon et al. Oct 2002 A1
20020150989 Greene et al. Oct 2002 A1
20020151056 Sasai et al. Oct 2002 A1
20020159981 Peled et al. Oct 2002 A1
20020160032 Long et al. Oct 2002 A1
20020160510 Hariri Oct 2002 A1
20020168765 Prockop et al. Nov 2002 A1
20020169408 Beretta et al. Nov 2002 A1
20020182241 Borenstein et al. Dec 2002 A1
20020182664 Dolecek et al. Dec 2002 A1
20020188962 Denhardt et al. Dec 2002 A1
20020197240 Chiu Dec 2002 A1
20030021850 Xu Jan 2003 A1
20030022390 Stephens Jan 2003 A1
20030027330 Lanza et al. Feb 2003 A1
20030027331 Yan et al. Feb 2003 A1
20030032143 Neff et al. Feb 2003 A1
20030036168 Ni et al. Feb 2003 A1
20030037836 Blatt et al. Feb 2003 A1
20030040113 Mizuno et al. Feb 2003 A1
20030049236 Kassem et al. Mar 2003 A1
20030054331 Fraser et al. Mar 2003 A1
20030059851 Smith Mar 2003 A1
20030059939 Page et al. Mar 2003 A1
20030069650 Karmiy et al. Apr 2003 A1
20030078345 Morrisey Apr 2003 A1
20030082795 Shuler et al. May 2003 A1
20030086915 Rader et al. May 2003 A1
20030089471 Gehr et al. May 2003 A1
20030092101 Ni et al. May 2003 A1
20030101465 Lawman et al. May 2003 A1
20030103957 McKerracher Jun 2003 A1
20030104568 Lee Jun 2003 A1
20030113813 Heidaran et al. Jun 2003 A1
20030113910 Levanduski Jun 2003 A1
20030124091 Tuse et al. Jul 2003 A1
20030124721 Cheatham et al. Jul 2003 A1
20030130593 Gonzalez Jul 2003 A1
20030133918 Sherley Jul 2003 A1
20030138950 McAllister et al. Jul 2003 A1
20030143727 Chang Jul 2003 A1
20030148152 Morrisey Aug 2003 A1
20030149011 Ackerman et al. Aug 2003 A1
20030152558 Luft et al. Aug 2003 A1
20030157078 Hall et al. Aug 2003 A1
20030157709 DiMilla et al. Aug 2003 A1
20030161817 Young et al. Aug 2003 A1
20030166272 Abuljadayel Sep 2003 A1
20030170214 Bader Sep 2003 A1
20030180296 Salcedo et al. Sep 2003 A1
20030185817 Thomas et al. Oct 2003 A1
20030202938 Rameshwar Oct 2003 A1
20030203483 Seshi Oct 2003 A1
20030204323 Morrisey Oct 2003 A1
20030211602 Atala Nov 2003 A1
20030211603 Earp et al. Nov 2003 A1
20030216718 Hamblin et al. Nov 2003 A1
20030219898 Sugaya et al. Nov 2003 A1
20030223968 Yang Dec 2003 A1
20030224420 Hellerstein et al. Dec 2003 A1
20030224510 Yamaguchi et al. Dec 2003 A1
20030225010 Rameshwar Dec 2003 A1
20030232432 Bhat Dec 2003 A1
20030232752 Freeman et al. Dec 2003 A1
20030235909 Hariri et al. Dec 2003 A1
20040009158 Sands et al. Jan 2004 A1
20040009589 Levenberg et al. Jan 2004 A1
20040010231 Leonhardt et al. Jan 2004 A1
20040014209 Lassar et al. Jan 2004 A1
20040018174 Palasis Jan 2004 A1
20040018617 Hwang Jan 2004 A1
20040023324 Sakano et al. Feb 2004 A1
20040023370 Yu et al. Feb 2004 A1
20040032430 Kung et al. Feb 2004 A1
20040033214 Young et al. Feb 2004 A1
20040033599 Rosenberg Feb 2004 A1
20040037811 Penn et al. Feb 2004 A1
20040037815 Clarke et al. Feb 2004 A1
20040038316 Kaiser et al. Feb 2004 A1
20040053869 Andrews et al. Mar 2004 A1
20040062753 Rezania et al. Apr 2004 A1
20040063205 Xu Apr 2004 A1
20040067585 Wang et al. Apr 2004 A1
20040071668 Bays et al. Apr 2004 A1
20040072259 Scadden et al. Apr 2004 A1
20040077079 Storgaard et al. Apr 2004 A1
20040079248 Mayer et al. Apr 2004 A1
20040087016 Keating et al. May 2004 A1
20040091936 West May 2004 A1
20040096476 Uhrich et al. May 2004 A1
20040097408 Leder et al. May 2004 A1
20040101959 Marko et al. May 2004 A1
20040107453 Furcht et al. Jun 2004 A1
20040110286 Bhatia Jun 2004 A1
20040115804 Fu et al. Jun 2004 A1
20040115806 Fu Jun 2004 A1
20040120932 Zahner Jun 2004 A1
20040121461 Honmou et al. Jun 2004 A1
20040121464 Rathjen et al. Jun 2004 A1
20040126405 Sahatjian et al. Jul 2004 A1
20040128077 Koebler et al. Jul 2004 A1
20040131601 Epstein et al. Jul 2004 A1
20040132184 Dennis et al. Jul 2004 A1
20040136967 Weiss et al. Jul 2004 A1
20040137612 Baksh Jul 2004 A1
20040137613 Vacant et al. Jul 2004 A1
20040143174 Brubaker Jul 2004 A1
20040143863 Li et al. Jul 2004 A1
20040151700 Harlan et al. Aug 2004 A1
20040151701 Kim et al. Aug 2004 A1
20040151706 Shakhov et al. Aug 2004 A1
20040151729 Michalopoulos et al. Aug 2004 A1
20040152190 Sumita Aug 2004 A1
20040161419 Strom et al. Aug 2004 A1
20040171533 Zehentner et al. Sep 2004 A1
20040180347 Stanton et al. Sep 2004 A1
20040191902 Hambor et al. Sep 2004 A1
20040197310 Sanberg et al. Oct 2004 A1
20040197375 Rezania et al. Oct 2004 A1
20040208786 Kevy et al. Oct 2004 A1
20040214275 Soejima et al. Oct 2004 A1
20040219134 Naughton et al. Nov 2004 A1
20040219136 Hariri Nov 2004 A1
20040219563 West et al. Nov 2004 A1
20040224403 Bhatia Nov 2004 A1
20040229351 Rodriguez et al. Nov 2004 A1
20040234972 Owens et al. Nov 2004 A1
20040235158 Bartlett et al. Nov 2004 A1
20040235160 Nishikawa et al. Nov 2004 A1
20040235166 Prockop et al. Nov 2004 A1
20040242469 Lee et al. Dec 2004 A1
20040258669 Dzau et al. Dec 2004 A1
20040259242 Malinge et al. Dec 2004 A1
20040259254 Honmou et al. Dec 2004 A1
20040260058 Scheek et al. Dec 2004 A1
20040260318 Hunter et al. Dec 2004 A1
20040265996 Schwarz et al. Dec 2004 A1
20050002914 Rosen et al. Jan 2005 A1
20050003460 Nilsson et al. Jan 2005 A1
20050003527 Lang et al. Jan 2005 A1
20050003534 Huberman et al. Jan 2005 A1
20050008624 Peled et al. Jan 2005 A1
20050008626 Fraser et al. Jan 2005 A1
20050009178 Yost et al. Jan 2005 A1
20050009179 Gemmiti et al. Jan 2005 A1
20050009181 Black et al. Jan 2005 A1
20050013804 Kato et al. Jan 2005 A1
20050014252 Chu et al. Jan 2005 A1
20050014253 Ehmann et al. Jan 2005 A1
20050014254 Kruse Jan 2005 A1
20050014255 Tang et al. Jan 2005 A1
20050019801 Rubin et al. Jan 2005 A1
20050019908 Hariri Jan 2005 A1
20050019910 Takagi et al. Jan 2005 A1
20050019911 Gronthos et al. Jan 2005 A1
20050026836 Dack et al. Feb 2005 A1
20050031587 Tsutsui et al. Feb 2005 A1
20050031595 Peled et al. Feb 2005 A1
20050031598 Levenberg et al. Feb 2005 A1
20050032122 Hwang et al. Feb 2005 A1
20050032207 Wobus et al. Feb 2005 A1
20050032209 Messina et al. Feb 2005 A1
20050032218 Gerlach Feb 2005 A1
20050036980 Chaney et al. Feb 2005 A1
20050037488 Mitalipova et al. Feb 2005 A1
20050037490 Rosenberg et al. Feb 2005 A1
20050037492 Xu et al. Feb 2005 A1
20050037493 Mandalam et al. Feb 2005 A1
20050037949 O'Brien et al. Feb 2005 A1
20050106119 Brandom et al. May 2005 A1
20050106127 Kraus et al. May 2005 A1
20050112447 Fletcher et al. May 2005 A1
20050112762 Hart et al. May 2005 A1
20050118712 Tsai et al. Jun 2005 A1
20050130297 Sarem et al. Jun 2005 A1
20050136093 Denk Jun 2005 A1
20050137517 Blickhan et al. Jun 2005 A1
20050142162 Hunter et al. Jun 2005 A1
20050149157 Hunter et al. Jul 2005 A1
20050152946 Hunter et al. Jul 2005 A1
20050158289 Simmons et al. Jul 2005 A1
20050172340 Logvinov et al. Aug 2005 A1
20050175665 Hunter et al. Aug 2005 A1
20050175703 Hunter et al. Aug 2005 A1
20050178395 Hunter et al. Aug 2005 A1
20050178396 Hunter et al. Aug 2005 A1
20050180957 Scharp et al. Aug 2005 A1
20050181502 Furcht et al. Aug 2005 A1
20050182463 Hunter et al. Aug 2005 A1
20050183731 Hunter et al. Aug 2005 A1
20050186244 Hunter et al. Aug 2005 A1
20050186671 Cannon et al. Aug 2005 A1
20050187140 Hunter et al. Aug 2005 A1
20050196421 Hunter et al. Sep 2005 A1
20050208095 Hunter et al. Sep 2005 A1
20050244963 Teplyashin Nov 2005 A1
20050249731 Aslan et al. Nov 2005 A1
20050255118 Wehner Nov 2005 A1
20050261674 Nobis et al. Nov 2005 A1
20050277577 Hunter et al. Dec 2005 A1
20050281790 Simmons et al. Dec 2005 A1
20050282733 Prins et al. Dec 2005 A1
20050283844 Furcht et al. Dec 2005 A1
20060002900 Binder et al. Jan 2006 A1
20060008452 Simmons et al. Jan 2006 A1
20060019389 Yayon et al. Jan 2006 A1
20060054941 Lu et al. Mar 2006 A1
20060083720 Fraser et al. Apr 2006 A1
20060099198 Thomson et al. May 2006 A1
20060166364 Senesac Jul 2006 A1
20060172008 Yayon et al. Aug 2006 A1
20060193840 Gronthos et al. Aug 2006 A1
20060228798 Verfaillie et al. Oct 2006 A1
20060233834 Guehenneux et al. Oct 2006 A1
20060239909 Anderson et al. Oct 2006 A1
20060258586 Sheppard et al. Nov 2006 A1
20060258933 Ellis et al. Nov 2006 A1
20060259998 Brumbley et al. Nov 2006 A1
20060280748 Buckheit Dec 2006 A1
20060286077 Gronthos et al. Dec 2006 A1
20070005148 Barofsky et al. Jan 2007 A1
20070011752 Paleyanda Jan 2007 A1
20070042462 Hildinger Feb 2007 A1
20070065938 Gronthos et al. Mar 2007 A1
20070105222 Wolfinbarger et al. May 2007 A1
20070116612 Williamson May 2007 A1
20070117180 Morikawa et al. May 2007 A1
20070122904 Nordon May 2007 A1
20070123996 Sugaya et al. May 2007 A1
20070160583 Lange et al. Jul 2007 A1
20070166834 Williamson et al. Jul 2007 A1
20070178071 Westenfelder Aug 2007 A1
20070192715 Kataria et al. Aug 2007 A1
20070196421 Hunter et al. Aug 2007 A1
20070197957 Hunter et al. Aug 2007 A1
20070198063 Hunter et al. Aug 2007 A1
20070202485 Nees et al. Aug 2007 A1
20070203330 Kretschmar et al. Aug 2007 A1
20070208134 Hunter et al. Sep 2007 A1
20070258943 Penn et al. Nov 2007 A1
20070274970 Gordon et al. Nov 2007 A1
20070275457 Granchelli et al. Nov 2007 A1
20070295651 Martinez et al. Dec 2007 A1
20070298015 Beer et al. Dec 2007 A1
20070298497 Antwiler Dec 2007 A1
20080003663 Bryhan et al. Jan 2008 A1
20080009458 Dornan et al. Jan 2008 A1
20080032398 Cannon et al. Feb 2008 A1
20080050770 Zhang et al. Feb 2008 A1
20080063600 Aguzzi et al. Mar 2008 A1
20080064649 Rameshwar Mar 2008 A1
20080069807 Jy et al. Mar 2008 A1
20080095676 Andretta Apr 2008 A1
20080095690 Liu Apr 2008 A1
20080103412 Chin May 2008 A1
20080110827 Cote et al. May 2008 A1
20080113426 Smith et al. May 2008 A1
20080113440 Gurney et al. May 2008 A1
20080153077 Henry Jun 2008 A1
20080160597 van der Heiden et al. Jul 2008 A1
20080166808 Nyberg Jul 2008 A1
20080181879 Catelas et al. Jul 2008 A1
20080190857 Beretta et al. Aug 2008 A1
20080194017 Esser et al. Aug 2008 A1
20080206831 Coffey et al. Aug 2008 A1
20080220522 Antwiler Sep 2008 A1
20080220523 Antwiler Sep 2008 A1
20080220524 Noll et al. Sep 2008 A1
20080220526 Ellison et al. Sep 2008 A1
20080221443 Ritchie et al. Sep 2008 A1
20080227189 Bader Sep 2008 A1
20080227190 Antwiler Sep 2008 A1
20080248572 Antwiler Oct 2008 A1
20080254533 Antwiler Oct 2008 A1
20080268165 Fekety et al. Oct 2008 A1
20080268538 Nordon et al. Oct 2008 A1
20080306095 Crawford Dec 2008 A1
20090004738 Merchav et al. Jan 2009 A1
20090011399 Fischer Jan 2009 A1
20090047289 Denhardt et al. Feb 2009 A1
20090074728 Gronthos et al. Mar 2009 A1
20090075881 Catelas et al. Mar 2009 A1
20090076481 Stegmann et al. Mar 2009 A1
20090081770 Srienc et al. Mar 2009 A1
20090081797 Fadeev et al. Mar 2009 A1
20090092608 Ni et al. Apr 2009 A1
20090098103 Madison et al. Apr 2009 A1
20090098645 Fang et al. Apr 2009 A1
20090100944 Newby Apr 2009 A1
20090104163 Deans et al. Apr 2009 A1
20090104653 Paldus et al. Apr 2009 A1
20090104692 Bartfeld et al. Apr 2009 A1
20090104699 Newby et al. Apr 2009 A1
20090118161 Cruz May 2009 A1
20090181087 Kraus et al. Jul 2009 A1
20090183581 Wilkinson et al. Jul 2009 A1
20090191627 Fadeev et al. Jul 2009 A1
20090191632 Fadeev et al. Jul 2009 A1
20090191634 Martin et al. Jul 2009 A1
20090203065 Gehman et al. Aug 2009 A1
20090203129 Furcht et al. Aug 2009 A1
20090203130 Furcht et al. Aug 2009 A1
20090214382 Burgess et al. Aug 2009 A1
20090214481 Muhs et al. Aug 2009 A1
20090214652 Hunter et al. Aug 2009 A1
20090215022 Page et al. Aug 2009 A1
20090227024 Baker et al. Sep 2009 A1
20090227027 Baker et al. Sep 2009 A1
20090233334 Hildinger et al. Sep 2009 A1
20090233353 Furcht et al. Sep 2009 A1
20090233354 Furcht et al. Sep 2009 A1
20090258379 Klein et al. Oct 2009 A1
20090269841 Wojciechowski et al. Oct 2009 A1
20090270725 Leimbach et al. Oct 2009 A1
20090280153 Hunter et al. Nov 2009 A1
20090280565 Jolicoeur et al. Nov 2009 A1
20090291890 Madison et al. Nov 2009 A1
20100009409 Hubbell et al. Jan 2010 A1
20100021954 Deshayes et al. Jan 2010 A1
20100021990 Edwards et al. Jan 2010 A1
20100028311 Motlagh et al. Feb 2010 A1
20100042260 Antwiler Feb 2010 A1
20100075410 Desai et al. Mar 2010 A1
20100086481 Baird et al. Apr 2010 A1
20100090971 Choi et al. Apr 2010 A1
20100092536 Hunter et al. Apr 2010 A1
20100093607 Dickneite Apr 2010 A1
20100111910 Rakoczy May 2010 A1
20100129376 Denhardt et al. May 2010 A1
20100129912 Su et al. May 2010 A1
20100136091 Moghe et al. Jun 2010 A1
20100144634 Zheng et al. Jun 2010 A1
20100183561 Sakthivel et al. Jul 2010 A1
20100183585 Van Zant et al. Jul 2010 A1
20100203020 Ghosh Aug 2010 A1
20100230203 Karayianni Sep 2010 A1
20100248366 Fadeev et al. Sep 2010 A1
20100278933 Sayeski et al. Nov 2010 A1
20100285453 Goodrich Nov 2010 A1
20100285590 Verfaillie et al. Nov 2010 A1
20100291180 Uhrich Nov 2010 A1
20100291181 Uhrich et al. Nov 2010 A1
20100297234 Sugino et al. Nov 2010 A1
20100304427 Faris et al. Dec 2010 A1
20100304482 Deshayes et al. Dec 2010 A1
20100310524 Bechor et al. Dec 2010 A1
20100316446 Runyon Dec 2010 A1
20110060463 Selker et al. Mar 2011 A1
20110085746 Wong et al. Apr 2011 A1
20110111498 Oh et al. May 2011 A1
20110129447 Meretzki et al. Jun 2011 A1
20110129486 Meiron Jun 2011 A1
20110143433 Oh et al. Jun 2011 A1
20110159584 Gibbons et al. Jun 2011 A1
20110171182 Abelman Jul 2011 A1
20110171659 Furcht et al. Jul 2011 A1
20110177595 Furcht et al. Jul 2011 A1
20110212493 Hirschel et al. Sep 2011 A1
20110256108 Meiron et al. Oct 2011 A1
20110256160 Meiron et al. Oct 2011 A1
20110293583 Aberman Dec 2011 A1
20120028352 Oh et al. Feb 2012 A1
20120051976 Lu et al. Mar 2012 A1
20120058554 Deshayes et al. Mar 2012 A1
20120064047 Verfaillie et al. Mar 2012 A1
20120064583 Edwards et al. Mar 2012 A1
20120086657 Stanton, IV et al. Apr 2012 A1
20120089930 Stanton, IV et al. Apr 2012 A1
20120118919 Cianciolo May 2012 A1
20120122220 Merchav et al. May 2012 A1
20120135043 Maziarz et al. May 2012 A1
20120145580 Paruit et al. Jun 2012 A1
20120156779 Anneren et al. Jun 2012 A1
20120178885 Kohn et al. Jul 2012 A1
20120189713 Kohn et al. Jul 2012 A1
20120208039 Barbaroux et al. Aug 2012 A1
20120219531 Oh et al. Aug 2012 A1
20120219737 Sugino et al. Aug 2012 A1
20120226013 Kohn et al. Sep 2012 A1
20120231519 Bushman et al. Sep 2012 A1
20120237557 Lewitus et al. Sep 2012 A1
20120295352 Antwiler Nov 2012 A1
20120308531 Pinxteren et al. Dec 2012 A1
20120315696 Luitjens et al. Dec 2012 A1
20130004465 Aberman Jan 2013 A1
20130039892 Aberman Feb 2013 A1
20130058907 Wojciechowski et al. Mar 2013 A1
20130059383 Dijkhuizen Borgart et al. Mar 2013 A1
20130101561 Sabaawy Apr 2013 A1
20130102070 Antwiler Apr 2013 A1
20130143313 Niazi Jun 2013 A1
20130157353 Dijkhuizen Borgart et al. Jun 2013 A1
20130259843 Duda et al. Oct 2013 A1
20130319575 Mendyk Dec 2013 A1
20130323213 Meiron et al. Dec 2013 A1
20130337558 Meiron et al. Dec 2013 A1
20140004553 Parker et al. Jan 2014 A1
20140017209 Aberman et al. Jan 2014 A1
20140030805 Kasuto et al. Jan 2014 A1
20140051162 Nankervis Feb 2014 A1
20140051167 Nankervis et al. Feb 2014 A1
20140112893 Tom et al. Apr 2014 A1
20140186937 Smith et al. Jul 2014 A1
20140193895 Smith et al. Jul 2014 A1
20140193911 Newby et al. Jul 2014 A1
20140242039 Meiron et al. Aug 2014 A1
20140248244 Danilkovitch et al. Sep 2014 A1
20140315300 Oh et al. Oct 2014 A1
20140342448 Nagels Nov 2014 A1
20150004693 Danilkovitch et al. Jan 2015 A1
20150024492 Antwiler Jan 2015 A1
20150104431 Pittenger et al. Apr 2015 A1
20150111252 Hirschel et al. Apr 2015 A1
20150125138 Karnieli et al. May 2015 A1
20150140653 Jones et al. May 2015 A1
20150140654 Nankervis et al. May 2015 A1
20150175950 Hirschel et al. Jun 2015 A1
20150225685 Hirschel et al. Aug 2015 A1
20150247122 Tom et al. Sep 2015 A1
20150259749 Santos et al. Sep 2015 A1
20160333314 Nguyen et al. Nov 2016 A1
20160362650 Wojciechowski et al. Dec 2016 A1
20160362652 Page et al. Dec 2016 A1
20170267966 Stanton, IV et al. Sep 2017 A1
20170335270 Stanton, IV et al. Nov 2017 A1
20180010082 Jagues et al. Jan 2018 A1
20180030398 Castillo Feb 2018 A1
20180155668 Hirschel et al. Jun 2018 A1
20190194628 Rao et al. Jun 2019 A1
Foreign Referenced Citations (323)
Number Date Country
1016332 Aug 1977 CA
102406926 Apr 2012 CN
3833925 Sep 1989 DE
4007703 Sep 1991 DE
10244859 Apr 2004 DE
10327988 Jul 2004 DE
102012200939 Jul 2013 DE
0220650 May 1987 EP
0201086 Jan 1992 EP
0224734 Mar 1992 EP
750938 Jan 1997 EP
906415 Apr 1999 EP
959980 Dec 1999 EP
1007631 Jun 2000 EP
1028737 Aug 2000 EP
1028991 Aug 2000 EP
1066052 Jan 2001 EP
1066060 Jan 2001 EP
1084230 Mar 2001 EP
1147176 Oct 2001 EP
1220611 Jul 2002 EP
1223956 Jul 2002 EP
1325953 Jul 2003 EP
1437404 Jul 2004 EP
1437406 Jul 2004 EP
1447443 Aug 2004 EP
1452594 Sep 2004 EP
1062321 Dec 2004 EP
1484080 Dec 2004 EP
1498478 Jan 2005 EP
1036057 Oct 2005 EP
1605044 Dec 2005 EP
1756262 Feb 2007 EP
1771737 Apr 2007 EP
1882030 Jan 2008 EP
1908490 Apr 2008 EP
1971679 Sep 2008 EP
1991668 Nov 2008 EP
2200622 Jun 2010 EP
2208782 Jul 2010 EP
2264145 Dec 2010 EP
2027247 Jan 2011 EP
2303293 Apr 2011 EP
2311938 Apr 2011 EP
2331957 Jun 2011 EP
2334310 Jun 2011 EP
2334783 Jun 2011 EP
2361968 Aug 2011 EP
2366775 Sep 2011 EP
2465922 Jun 2012 EP
2481819 Aug 2012 EP
2548951 Jan 2013 EP
2561066 Feb 2013 EP
2575831 Apr 2013 EP
2591789 May 2013 EP
2624845 Aug 2013 EP
2626417 Aug 2013 EP
2641606 Sep 2013 EP
2689008 Jan 2014 EP
2694639 Feb 2014 EP
2697362 Feb 2014 EP
2739720 Jun 2014 EP
2807246 Dec 2014 EP
1414671 Nov 1975 GB
2297980 Aug 1996 GB
2360789 Oct 2001 GB
3285 May 2007 HU
H02245177 Sep 1990 JP
H03047074 Feb 1991 JP
2003052360 Feb 2003 JP
2003510068 Mar 2003 JP
2005278564 Oct 2005 JP
2007000038 Jan 2007 JP
2012-506257 Mar 2012 JP
5548207 Jul 2014 JP
5548207 Jul 2014 JP
2016-536998 Dec 2016 JP
2016-537001 Dec 2016 JP
2019-516029 Jun 2019 JP
2019-525765 Sep 2019 JP
101228026 Jan 2013 KR
2015-0002762 Jan 2015 KR
101504392 Mar 2015 KR
101548790 Aug 2015 KR
101553040 Sep 2015 KR
2017-0076679 Jul 2017 KR
2018-0027501 Mar 2018 KR
102027596 Oct 2019 KR
115206 Apr 2003 MY
8602379 Apr 1986 WO
8801643 Mar 1988 WO
WO 8912676 Dec 1989 WO
9002171 Mar 1990 WO
WO-9013306 Nov 1990 WO
WO-9105238 Apr 1991 WO
9107485 May 1991 WO
WO-9106641 May 1991 WO
WO-9109194 Jun 1991 WO
9110425 Jul 1991 WO
9210564 Jun 1992 WO
WO-9425571 Nov 1994 WO
9504813 Feb 1995 WO
9521911 Aug 1995 WO
WO 9524468 Sep 1995 WO
9527041 Oct 1995 WO
WO-9629395 Sep 1996 WO
WO-9639035 Dec 1996 WO
WO-9705826 Feb 1997 WO
9716527 May 1997 WO
WO-9729792 Aug 1997 WO
9740137 Oct 1997 WO
WO-9739104 Oct 1997 WO
WO-1997-040137 Oct 1997 WO
WO 9822588 May 1998 WO
WO-9831403 Jul 1998 WO
9853046 Nov 1998 WO
WO-9851317 Nov 1998 WO
WO-9851785 Nov 1998 WO
WO-9905180 Feb 1999 WO
WO-9924391 May 1999 WO
WO-9924490 May 1999 WO
WO-9927167 Jun 1999 WO
WO-9949015 Sep 1999 WO
WO-0006704 Feb 2000 WO
WO-0009018 Feb 2000 WO
WO-OO16420 Mar 2000 WO
WO-OO17326 Mar 2000 WO
WO-0029002 May 2000 WO
WO-0032225 Jun 2000 WO
WO-0044058 Jul 2000 WO
WO 0046354 Aug 2000 WO
WO-0054651 Sep 2000 WO
WO-0056405 Sep 2000 WO
WO-0059933 Oct 2000 WO
WO-0069449 Nov 2000 WO
0075275 Dec 2000 WO
WO-0075196 Dec 2000 WO
WO-0077236 Dec 2000 WO
WO-2001 000783 Jan 2001 WO
WO-2001 011011 Feb 2001 WO
WO-2001 018174 Mar 2001 WO
WO-2001021766 Mar 2001 WO
0123520 Apr 2001 WO
WO-2001 025402 Apr 2001 WO
WO-2001 029189 Apr 2001 WO
WO-0122810 Apr 2001 WO
WO-2001 034167 May 2001 WO
WO-2001049851 Jul 2001 WO
WO-2001 054706 Aug 2001 WO
0194541 Dec 2001 WO
WO-2001 -094541 Dec 2001 WO
0228996 Apr 2002 WO
WO-2002042422 May 2002 WO
WO-2002057430 Jul 2002 WO
WO-2002092794 Nov 2002 WO
WO-2002101385 Dec 2002 WO
WO-2003010303 Feb 2003 WO
WO-2003014313 Feb 2003 WO
WO-2003016916 Feb 2003 WO
03024587 Mar 2003 WO
WO-2003023018 Mar 2003 WO
WO-2003023019 Mar 2003 WO
WO-2003025167 Mar 2003 WO
WO-2003029402 Apr 2003 WO
WO 03039459 May 2003 WO
WO-2003040336 May 2003 WO
WO-2003042405 May 2003 WO
WO-2003046161 Jun 2003 WO
WO-2003055989 Jul 2003 WO
WO-2003061685 Jul 2003 WO
WO-2003061686 Jul 2003 WO
WO-2003068961 Aug 2003 WO
WO-2003072064 Sep 2003 WO
WO-2003078609 Sep 2003 WO
WO-2003078967 Sep 2003 WO
WO-2003080816 Oct 2003 WO
WO-2003082145 Oct 2003 WO
WO-2003085099 Oct 2003 WO
WO-2003089631 Oct 2003 WO
WO-2003091398 Nov 2003 WO
WO-2003095631 Nov 2003 WO
03105663 Dec 2003 WO
WO-2004001697 Dec 2003 WO
WO-2004012226 Feb 2004 WO
WO-2004016779 Feb 2004 WO
WO-2004018526 Mar 2004 WO
WO-2004018655 Mar 2004 WO
WO-2004026115 Apr 2004 WO
WO-2004029231 Apr 2004 WO
WO-2004042023 May 2004 WO
WO-2004042033 May 2004 WO
WO-2004042040 May 2004 WO
WO-2004044127 May 2004 WO
WO-2004044158 May 2004 WO
WO-2004046304 Jun 2004 WO
WO-2004050826 Jun 2004 WO
WO-2004053096 Jun 2004 WO
WO-2004055155 Jul 2004 WO
WO-2004056186 Jul 2004 WO
WO-2004065616 Aug 2004 WO
WO-2004069172 Aug 2004 WO
WO-2004070013 Aug 2004 WO
WO-2004072264 Aug 2004 WO
WO-2004073633 Sep 2004 WO
WO-2004074464 Sep 2004 WO
WO-2004076642 Sep 2004 WO
WO-2004076653 Sep 2004 WO
2004090112 Oct 2004 WO
WO-2004087870 Oct 2004 WO
WO-2004094588 Nov 2004 WO
WO-2004096975 Nov 2004 WO
WO-2004104166 Dec 2004 WO
WO-2004106499 Dec 2004 WO
WO-2004113513 Dec 2004 WO
WO-2005001033 Jan 2005 WO
WO-2005001081 Jan 2005 WO
WO-2005003320 Jan 2005 WO
WO-2005007799 Jan 2005 WO
WO-2005010172 Feb 2005 WO
WO-2005011524 Feb 2005 WO
WO-2005012480 Feb 2005 WO
WO-2005012510 Feb 2005 WO
WO-2005012512 Feb 2005 WO
WO-05014775 Feb 2005 WO
WO-2005028433 Mar 2005 WO
WO-05044972 May 2005 WO
WO-2005056747 Jun 2005 WO
WO-05051316 Jun 2005 WO
WO-2005063303 Jul 2005 WO
WO-2005075636 Aug 2005 WO
2005087915 Sep 2005 WO
WO 2005104755 Nov 2005 WO
WO-2005107760 Nov 2005 WO
WO-2006009291 Jan 2006 WO
2006026835 Mar 2006 WO
WO-2006032075 Mar 2006 WO
WO-2006032092 Mar 2006 WO
WO 2006037022 Apr 2006 WO
WO-2006108229 Oct 2006 WO
WO-2006113881 Oct 2006 WO
WO-061 24021 Nov 2006 WO
WO-20061 21445 Nov 2006 WO
WO-061 29312 Dec 2006 WO
WO 2007038572 Apr 2007 WO
WO 2007059473 May 2007 WO
WO-2007115367 Oct 2007 WO
WO-2007115368 Oct 2007 WO
WO 2007117765 Oct 2007 WO
2007136821 Nov 2007 WO
2007139742 Dec 2007 WO
2007139746 Dec 2007 WO
2007139747 Dec 2007 WO
2007139748 Dec 2007 WO
WO-2008006168 Jan 2008 WO
WO-2008011664 Jan 2008 WO
WO-200801 7128 Feb 2008 WO
WO-2008028241 Mar 2008 WO
WO-08040812 Apr 2008 WO
WO 2008073635 Jun 2008 WO
WO-2008116261 Oct 2008 WO
WO-20081 49129 Dec 2008 WO
WO-2009026635 Mar 2009 WO
WO-09058146 May 2009 WO
WO-09080054 Jul 2009 WO
WO-09081408 Jul 2009 WO
WO-2009140452 Nov 2009 WO
WO-09132457 Nov 2009 WO
WO-2009144720 Dec 2009 WO
WO-10005527 Jan 2010 WO
WO-201 0019886 Feb 2010 WO
WO-10014253 Feb 2010 WO
WO-10019997 Feb 2010 WO
WO-201 0026573 Mar 2010 WO
WO-201 0026574 Mar 2010 WO
WO-201 0026575 Mar 2010 WO
WO-201 0036760 Apr 2010 WO
WO 2010036760 Apr 2010 WO
WO-201 0059487 May 2010 WO
WO-10061377 Jun 2010 WO
WO-10068710 Jun 2010 WO
WO-10071826 Jun 2010 WO
WO-10083385 Jul 2010 WO
WO-10111255 Sep 2010 WO
WO-101 19036 Oct 2010 WO
WO-10123594 Oct 2010 WO
WO-201 1025445 Mar 2011 WO
WO 2011098592 Aug 2011 WO
WO-201 1132087 Oct 2011 WO
WO 2011130617 Oct 2011 WO
WO-201 1147967 Dec 2011 WO
WO-201 2072924 Jun 2012 WO
WO-201 2127320 Sep 2012 WO
WO-201 2138968 Oct 2012 WO
WO-201 2140519 Oct 2012 WO
2012171026 Dec 2012 WO
2012171030 Dec 2012 WO
WO-2012168295 Dec 2012 WO
WO 2013085682 Jun 2013 WO
WO-201 3110651 Aug 2013 WO
WO-201 4037862 Mar 2014 WO
WO-2014037863 Mar 2014 WO
WO-201 4068508 May 2014 WO
WO-201 4128306 Aug 2014 WO
WO-201 4128634 Aug 2014 WO
WO-2014131846 Sep 2014 WO
WO-2014141111 Sep 2014 WO
WO-201 5004609 Jan 2015 WO
WO 2015059714 Apr 2015 WO
2015-073913 May 2015 WO
WO 2015069943 May 2015 WO
WO-201 5118148 Aug 2015 WO
WO-201 5118149 Aug 2015 WO
WO 2015118148 Aug 2015 WO
WO 2015118149 Aug 2015 WO
WO-201 5131143 Sep 2015 WO
WO-2015158868 Oct 2015 WO
WO 2016130940 Aug 2016 WO
2016-183350 Nov 2016 WO
WO-201 7072201 May 2017 WO
WO 2017072201 May 2017 WO
WO 2017158611 Sep 2017 WO
WO 2017207822 Dec 2017 WO
WO 2018183426 Oct 2018 WO
Non-Patent Literature Citations (307)
Entry
Sheu et al., “Large-scale production of lentiviral vector in a closed system hollow fiber bioreactor”, 2015, Molecular Therapy—Methods and Clinical Development 2, p. 1-7.
Biovest International, “AutovaxIDTM: advanced hollow fibre bioreactors with automated lactate control yield higher density monoclonal antibody production”, VWRbioMarke, No. 21, Sep. 2008, pp. 10-11.
Chang et al., “Membrane Bioreactors: Present and Prospects”, Advances in Biochemical Engineering, 1991, vol. 44, pp. 27-64.
Chang, Ho Nam, “Membrane Bioreactors: Engineering Aspects”, Biotech. Adv., 1987, vol. 5, pp. 129-145.
Clausen et al. , “Lactate as an Indicator of Terminating Time in Insect Cell Culture Baculovirus Expression Vector Systems”, Biotechnology Techniques, vol. 10, No. 10, Oct. 1996, pp. 721-726.
Edgington, Stephen M., “New Horizons for Stem-Cell Bioreactors”, Biotechnology, Oct. 1992, vol. 10, pp. 1099-1106.
Gastens et al., “Good Manufacturing Practice-Compliant Expansion of Marrow-Derived Stem and Progenitor Cells for Cell Therapy”, Cell Transplantation, 2007, vol. 16, pp. 685-696.
Gerlach, J.C. et al., “Comparison of hollow fibre membranes for hepatocyte immobilization in bioreactors,” The International Journal of Artificial Organs, 1996, vol. 19 No. 10, pp. 610-616.
Gloeckner et al., “New Miniaturized Hollow-Fiber Bioreacter for in Vivo Like Cell Culture, Cell Expansion, and Production of Cell-Derived Products”, Biotechnol. Prog., Aug. 21, 2001, vol. 17, No. 5, pp. 828-831.
Gramer et al., “Screening Tool for Hollow-Fiber Bioreactor Process Development”, Biotechnol. Prog., 1998, vol. 14, pp. 203-209.
Grayson et al., “Effects of Hypoxia on Human Mesenchymal Stem Cell Expansion and Plasticity in 3D Constructs”, J. Cellular Physiology, 2006, 207:331-339.
Hirschel et al., “An Automated Hollow Fiber System for the Large Scale Manufacture of Mammalian Cell Secreted Product”, Large Scale Cell Culture Technology, ed. Bjorn K. Lydersen, Hanser Publishers, 1987, pp. 113-144.
Lloyd, J.R. et al., “Hollow-Fibre bioreactors compared to batch and chemostat culture for the production of a recombinant toxoid by a marine Vibrio,” Appl. Microbiol Biotechnol, Aug. 1997, vol. 48, pp. 155-161.
Neumann, Detlef et al., “Bioreaktorsteurung mit grafischer Bedienoberflache,” ATP Automatisierungstechnische Praxis, Mar. 1995, pp. 16-23, vol. 37, No. 3, Munchen, DE. (English language translation included).
Nielsen, Lars Keld, “Bioreactors for Hematopoietic Cell Culture”, Annu. Rev. Biomed. Eng., 1999, vol. 1, pp. 129-152.
Ozturk et al., “Real-Time Monitoring and Control of Glucose and Lactate Concentrations in a Mammalian Cell Perfusion Reactor”, Biotechnology and Bioengineering, vol. 53, No. 4, Feb. 20, 1997, pp. 372-378.
Pörtner et al., “An Overview on Bioreactor Design, Prototyping and Process Control for Reproducible Three-Dimensional Tissue Culture”, Drug Testing in Vitro: Breakthroughs and Trends in Cell Culture Technology, ed. Uwe Marx and Volker Sandig, 2007, Wiley-VCH, pp. 53-78.
Sauer, I. et al., “Extracorporeal liver support based on primary human liver cells and albumin dialysis—treatment of patient with primary grall non function,” Journal of Hepatology, Oct. 2003, vol. 39 No. 4, pp. 649-653.
Wang et al., “Influence of Oxygen on the Proliferation and Metabolism of Adipose Derived Adult Stem Cells”, J. Cellular Physiology, 2005, 204:184-161.
Zhao et al., “Effects of Oxygen Transport on 3-D human Mesenchymal Stem Cell Metabolic Activity in Perfusion and Static Cultures: Experiments and Mathematical Model”, Biotechnol. Prog, 2005, 27, 1269-1280.
Zhao et al., “Perfusion Bioreactor System for Human Mesenchymal Stem Cell Tissue Engineering: Dynamic Cell Seeding and Construct Development”, Biotechnology and Bioengineering, Aug. 20, 2005, vol. 91, No. 4, pp. 482-493.
Abumiya et al., “Shear Stress Induces Expression of Vascular Endothelial Growth Factor Receptor Flk-1/KDR Through the CT-Rich Sp1 Binding Site,” Ateriosclerosis, Thrombosis, and Vascular Biology, vol. 22, Jun. 2002, pp. 907-913.
Akiyama et al., “Ultrathin Poly(N-isopropylacrylamide) Grafted Layer on Polystyrene Surfaces for Cell Adhesion/Detachment Control,” Langmuir, vol. 20, No. 13, May 26, 2004, pp. 5506-5511.
Akram et al., “Mesenchymal Stem Cells Promote Alveolar Epithelial Cell Wound Repair in vitro through Distinct Migratory and Paracrine Mechanisms,” Respiratory Research, vol. 14, No. 9, 2013, pp. 1-16.
Anamelechi et al., “Streptavidin Binding and Endothelial Cell Adhesion to Biotinylated Fibronectin,” Langmuir, vol. 23, No. 25, Dec. 4, 2007, pp. 12583-12588.
Azar et al., “Heart Rates of Male and Female Sprague—Dawley and Spontaneously Hypertensive Rats Housed Singly or in Groups,” Journal of the American Association for Laboratory Animal Science, vol. 50, No. 2, Mar. 2011, pp. 175-184.
Beacher-Allan et al., “CD4+CD25high Regulatory Cells in Human Peripheral Blood,” The Journal of Immunology, vol. 167, 2001, pp. 1245-1253.
Boitano et al., “Aryl Hydrocarbon Receptor Antagonists Promote the Expansion of Human Hematopoietic Stem Cells,” Science, vol. 329, No. 5997, published Sep. 10, 2010. corrected May 6, 2011, pp. 1345-1348.
Brunstein et al., “Infusion of ex vivo Expanded T Regulatory Cells in Adults Transplanted with Umbilical Cord Blood: Safety Profile and Detection Kinetics,” Blood, vol. 117, No. 3, Jan. 20, 2011, pp. 1061-1070.
Bryce et al., “In vitro Micronucleus Assay Scored by Flow Cytometry Provides a Comprehensive Evaluation of Cytogenetic Damage and Cytotoxicity,” Mutation Research, vol. 630, Mar. 19, 2007, pp. 78-91.
Bryce et al., “Interlaboratory Evaluation of a Flow Cytometric, High Content in vitro Micronucleus Assay,” Mutation Research, vol. 650, Jan. 7, 2008, pp. 181-195.
Camacho Villa et al., “CD133+CD34+ and CD133+CD38+ Blood Progenitor Ceils as Predictors of Platelet Engraftment in Patients Undergoing Autologous Peripheral Blood Stem Cell Transplantation,” Transfusion and Apheresis Science, vol. 46, 2012, pp. 239-244.
Cano et al., “Immobilization of endo-1,4-β-xylanase on Polysulfone Acrylate Membranes: Synthesis and Characterization,” Journal of Membrane Science, vol. 280, Feb. 28, 2006, pp. 383-388.
Carvell et al., “Monitoring Live Biomass in Disposable Bioreactors,” BioProcess International, vol. 14, No. 3, Mar. 2016, pp. 40-48.
Carvell et al., “On-line Measurements and Control of Viable Cell Density in Cell Culture Manufacturing Processes Using Radio Frequency Impedance,” Cytotechnology, 2006, vol. 50, pp. 35-48.
Cuchiara et al., “Covalent Immobilization of SCF and SDF1α for in vitro Culture of Hematopoietic Progenitor Cells,” Acta Biomaterials, vol. 9, No. 12, Dec. 2013, pp. 9258-9269.
Da Silva et al., “Smart Thermoresponsive Coatings and Surfaces for Tissue Engineering: Switching Cell-Material Boundaries,” Trends in Biotechnology, vol. 15, No. 12, 2007, pp. 577-583.
Hao et al., “A Functional Comparison of CD34+ CD38− Cells in Cord Blood and Bone Marrow,” Blood, vol. 86, No. 10, Nov. 15, 1995, pp. 3745-3753.
Harimoto et al., “Novel Approach for Achieving Double-Layered Cell Sheets Co-Culture: Overlaying Endothelial Cell Sheets onto Monolayer Hepatocytes Utilizing Temperature-Responsive Culture Dishes,” Journal of Biomedical Material Research, vol. 62, 2002, pp. 464-470.
Högstedt et al., “Frequency and Size Distribution of Micronuclei in Lymphocytes Stimulated with Phytohemagglutinin and Pokeweed Mitogen in Workers Exposed to Piperazine,” Hereditas, vol. 109, 1998, pp. 139-142.
Itkin et al., “SDF-1 Keeps HSC Quiescent at Home,” Blood, vol. 117, No. 2, Jan. 13, 2011. pp. 373-374.
Jang et al., “Syndecan-4 Proteoliposomes Enhance Fibroblast Growth Factor-2 (FGF-2)-Induced Proliferation, Migration, and Neovascularization of Ischemic Muscle,” PNAS, vol. 109, No. 5, Jan. 31, 2012, pp. 1679-1684.
Johansson et al., “Pancreatic Islet Survival and Engraftment Is Promoted by Culture on Functionalized Spider Silk Matrices,” PLoS ONE, Jun. 19, 2015, pp. 1-21.
Klein et al., “Affinity Membranes Prepared from Hydrophilic Coatings on Microporous Polysulfone Hollow Fibers,” Journal of Membrane Science, vol. 90, 1994, pp. 69-80.
Koestenbauer et al., “Protocols for Hematopoietic Stem Cell Expansion from Umbilical Cord Blood,” Cell Transplantation, vol. 18, May 6, 2009, pp. 1059-1068.
Koller et al., “Clinical-scale Human Umbilical Cord Blood Cell Expansion in a Novel Automated Perfusion Culture System,” Bone Marrow Transplantation, vol. 21, 1998, pp. 653-663.
Lang et al., “Generation of Hematopoietic Humanized Mice in the Newborn BALB/C-Rag2null Il2rynull Mouse Model: A Multivariable Optimization Approach,” Clinical Immunology, vol. 140, Apr. 14, 2011, pp. 102-116.
Lataillade et al., “Chemokine SDF-1 Enhances Circulating CD341 Cell Proliferation in Synergy with Cytokines: Possible Role in Progenitor Survival,” Blood, vol. 95, No. 3, Feb. 1, 2000, pp. 756-768.
Lee et al., “Long-Term Outcomes Following CD19 CAR T Cell Therapy for B-ALL Are Superior in Patients Receiving a Fludarabine/Cyclophosphamide Preparative Regimen and Post-CAR Hematopoietic Stem Cell Transplantation,” Blood, vol. 128, No. 22, Dec. 2, 2016, Ab. 218.
Li et al., “Heparin-induced Conformation Changes of Fibronectin within the Extracellular Matrix Promote hMSC Osteogenic Differentiation,” Biomaterials Science, vol. 3, 2015, pp. 73-84.
Malin et al., “Noninvasive Prediction of Glucose by Near-Infrared Diffuse Reflectance Spectroscopy,” Clinical Chemistry, vol. 45, No. 9, 1999, pp. 1651-1658.
Marek-Trzonkowska et al., “Administration of CD4+ CD25high CD127− Regulatory T Cells Preserves β-Cell Function in Type 1 Diabetes in Children,” Diabetes Care, vol. 35, No. 9, Sep. 2012, pp. 1817-1820.
Murugappan et al., “Human Hematopoietic Progenitor Cells Grow Faster under Rotational Laminar Flows,” Biotechnology Progress—Cell Culture & Tissue Engineering, Online, Apr. 22, 2010.
Nelson et al., “Emergent Patterns of Growth Controlled by Multicellular Form and Mechanics,” PNAS, vol. 102, No. 33, Aug. 16, 2005, pp. 11594-11599.
Nicolette et al., “In Vitro Micronucleus Screening of Pharmaceutical Candidates by Flow Cytometry in Chinese Hamster V79 Cells,” Environmental and Molecular Mutagenesis, vol. 52, Oct. 20, 2010, pp. 355-362.
Nugent et al., “Adventitial Endothelial Implants Reduce Matrix Metalloproteinase-2 Expression and Increase Luminal Diameter in Porcine Arteriovenous Grafts,” Journal of Vascular Surgery, vol. 46, No. 3, Sep. 2007, pp. 548-556.e2.
Okano et al., “Mechanism of Cell Detachment from Temperature-Modulated, Hydrophilic-Hydrophobic Polymer Surfaces,” Biomaterials, vol. 16, No. 4, 1995, pp. 297-303.
Putnam et al., “Expansion of Human Regulatory T-Cells from Patients with Type 1 Diabetes,” Diabetes, vol. 58, Mar. 2009, pp. 652-662.
Rodrigues et al.. “Stem Cell Cultivation in Bioreactors,” Biotechnology Advances, vol. 29, Jun. 25, 2011, pp. 815-829.
Ronco et al., “Blood and Dialysate Flow Distributions in Hollow-Fiber Hemodialyzers Analyzed by Computerized Helical Scanning Technique,” Journal of the American Society of Nephrology, vol. 13, 2002, pp. S53-S61.
Ryu et al., “Near-infrared Light Responsive Synthetic c-di-GMP Module for Optogenetic Applications,” ACS Synthetic Biology, vol. 3, Jan. 28, 2014, pp. 802-810.
Shimizu et al., “Fabrication of Pulsatile Cardiac Tissue Grafts Using a Novel 3-Dimensional Cell Sheet Manipulation Technique and Temperature-Responsive Cell Culture Surfaces,” Circulation Research, vol. 90, Feb. 22, 2002, e40-e48, pp. 1-9.
Smith et al., “Expansion of Neutrophil Precursors and Progenitors in Suspension Cultures of CD34+ Cells Enriched from Human Bone Marrow,” Experimental Hematology, vol. 21, 1993, pp. 870-877.
Takezawa et al., “Cell Culture on a Thermo-responsive Polymer Surface,” Nature, Bio/Technology, vol. 8, Sep. 1990, pp. 854-856.
Tiziani et al., “Metabolomic Profiling of Drug Response in Acute Myeloid Leukaemia Cell lines,” PLoS ONE, vol. 4, Issue 1, Jan. 22, 2009, e4251.
First Office Action for corresponding Chinese Patent Application No. 201880030588.1 dated Sep. 30, 2022, with English translation (9 Pages).
Abumiya, et al. at National Cardiovascular Center Research Institute in Japan, suggest that subjecting human umbilical vein endothelial cells (HUVECs) to laminar shear stress for a period of 8 hours increased the relative expression of VEGFR-2 mRNA (Ateriosclerosis, Thrombosis, and Vascular Biology, 2002).
Afzali B, Edozie FC, Fazekasova H, Scotta C, Mitchell PJ, Canavan JB, Kordasti SY, Chana PS, Ellis R, Lord GM, John S, Hilton R, Lechler RI, Lombardi G. Comparison of regulatory T cells in hemodialysis patients and healthy controls: implications for cell therapy in transplantation. Clin J Am Soc Nephrol. 2013;8(8):1396-405.
Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the Cell. 4th edition. New York: Garland Science; 2002. Fibroblasts and Their Transformations: The Connective-Tissue Cell Family. Available from: https://www.ncbi.nlm.nih.gov/books/NBK26889.
Alenazi, Noof A., et al. “Modified polyether-sulfone membrane: A mini review.” Designed monomers and polymers 20.1 (2017): 532-546.
Almeida L, Lochner M, Berod L, SparwasserT. Metabolic pathways in T cell activation and lineage differentiation. Semin Immunol. 2016;28(5):514-524.
Amy Putnam, Todd M. Brusko, Michael R. Lee, Weihong Liu, Gregory L. Szot, Taumoha Ghosh, Mark A. Atkinson, and Jeffrey A. Bluestone. Expansion of human regulatory T-Cells from patients with Type 1 Diabetes. Diabetes, 58: 652-662, 2009.
Anurathapan et al., “Engineered T cells for cancer treatment,” Cytotherapy, vol. 16, pp. 713-733,2014.
Aronowski J, Samways E, Strong R, Rhoades HM, Grotta JC. An alternative method for the quantitation of neuronal damage after experimental middle cerebral artery occlusion in rats: Analysis of behavioral deficit. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 1996;16:705-713.
Arrigoni, Chiara, et al. “Rotating versus perfusion bioreactor for the culture of engineered vascular constructs based on hyaluronic acid.” Biotechnology and bioengineering 100.5 (2008): 988-997.
Bai/Delaney (Nohla Therapeutics) showed that expanding Cord Blood-derived CD34+CD38-CD45RA- HSPCs in a biodegradable zwitterionic hydrogel with a rNotch ligand cocktail for 24 days mitigated HSPC differentiation and promoted self-renewal of lymphoid and myeloid cell phenotypes in an NSG mouse model (Nature Medicine, 2019).
Ballas CB, Zielske SP, Gerson SL (2002) Adult bone marrow stem cells for cell and gene therapies: implications for greater use. J Cell Biochem Suppl 38: 20-28.
Ballke C, Gran E, Baekkevold ES, Jahnsen FL. Characterization of Regulatory T-Cell Markers in CD4+ T Cells of the Upper Airway Mucosa. PLoS One. 2016;11(2):e0148826.
Baraniak PR, McDevitt TC (2010) Stem cell paracrine actions and tissue regeneration. Regen Med 5(1): 121-143.
Barckhausen C, Rice B, Baila S, et al. (2016) GMP-Compliant Expansion of Clinical-Grade Human Mesenchymal Stromal/Stem Cells Using a Closed Hollow Fiber Bioreactor. Methods Mol Biol 1416: 389-412.
Barker et al. “CD34+ Cell Content of 126 341 Cord Blood Units in the US Inventory: Implications for Transplantation and Banking,” blood Advances, vol. 3, No. 8, pp. 1267-1271, Apr. 23, 2019.
Bazarian JJ, Cernak I, Noble-Haeusslein L, Potolicchio S, Temkin N. Long-term neurologic outcomes after traumatic brain injury. The Journal of head trauma rehabilitation. 2009;24:439-451.
Bending D, Pesenacker AM, Ursu S, Wu Q, Lorn H, Thirugnanabalan B, Wedderbum LR. Hypomethylation at the regulatory T cell-specific demethylated region in CD25hi T cells is decoupled from FOXP3 expression at the inflamed site in childhood arthritis. J Immunol. 2014;193(6):2699-708.
Berendse M, Grounds MD, Lloyd CM (2003) Myoblast structure affects subsequent skeletal myotube morphology and sarcomere assembly. Exp Cell Res 291(2): 435-450.
Bernard, A., Payton, Mar. 1995. “Fermentation and Growth of Escherichia coli for Optimal Protein Production”, .
Berney SM, Schaan T, Wolf RE, van der Heyde H, Atkinson Tp. CD2 (OKT11) augments CD3-mediated intracellular signaling events in human T lymphocytes. J Investig Med. 2000;48(2):102-9.
Bioheart Clinical Trial Clinica 1302 Apr. 18, 2008.
Biomolecular and Cellular Interactions with the Hollow Fiber Membrane Currently Used in the Quantum® Cell Expansion System. 12th NJ Symposium on Biomaterials Science, Oct. 6-7, 2014, New Brunswick, NJ.
Blache C, Chauvin JM, Marie-Cardine A, Contentin N, Pommier P, Dedreux I, Francois S, Jacquot S, Bastit D, Boyer 0. Reduced frequency of regulatory T cells in peripheral blood stem cell compared to bone marrow transplantations. Biol Blood Marrow Transplant. 2010;16(3):430-4.
Bluestone et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Science Translational Medicine 7(315):1-34, 2015.
Bluestone JA, Tang Q. Treg cells-the next frontier of cell therapy. Science. 2018;362(6411):154-155.
Blum S, Moore AN, Adams F, Dash Pk. A mitogen-activated protein kinase cascade in the ca1/ca2 subfield of the dorsal hippocampus is essential for long-term spatial memory. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1999; 19:3535-3544.
Bojun Li et al. Heparin-induced conformation changes of fibronectin within the extracellular matrix promote hMSC osteogenic differentiation. Biomaterials Science 3: 73-84, 2015.
Boquest AC, Shahdadfar A, Brinchmann JE, Collas P. Isolation of Stromal Stem Cells from Human Adipose Tissue. Methods Mol Biol. 2006;325:35-46. doi: 10.1385/1-59745-005-7:35. PMID: 16761717.
Borden, M. and Longo, M., “Dissolution Behavior of Lipid Monolayer-Coated, Air-Filled Microbubbles: Effect of Lipid Hydrophobic Chain Length,” Langmuir, vol. 18, pp. 9225-9233, 2002.
Bourke, Sharon L., and Joachim Kohn. “Polymers derived from the amino acid L-tyrosine: polycarbonates, polyarylates and copolymers with poly (ethylene glycol).” Advanced drug delivery reviews 55.4 (2003): 447-466.
Brand, K. and Hermfisse, U., “Aerobic Glycolysis by Proliferating Cells: a Protective Strategy against Reactive Oxygen Species,” The FASEB Journal, vol. 11, pp. 388-395, Apr. 1997.
Brentjens et al., “CD19-Targeted T Cells Rapidly Induce Molecular Remission in Adults with Chemotherapy-Refractory Acute Lympohblastic Leukemia,” Science Translational Medicine, vol. 5, Issue 177, pp. 1-9, Mar. 20, 2013.
Brentjens et al., “Safety and Persistance of Adoptively Transferred Autologous CD19-Target T Cells in Patients with Relapsed or Chemotherapy Refractory B-Cell Leukemias,” Blood, vol. 118, No. 18, pp. 4817-4828, Nov. 3, 2011.
C. H. Weaver, et al. An Analysis of Engraftment Kinetics as a function of the CD34 Content of the Peripheral Blood Progenitor Cell Collections in 692 Patients After the Administration of Myeloblative Chemotherapy. Blood 86(10): 3691-3969, 1995.
Carswell, K. and Papoutsakis, E. “Culture of Human T Cells in Stirred Bioreactors for Cellular Immunotherapy Applications: Shear, Proliferation, and the IL-2 Receptor,” Biotechnology and Bioengineering, vol. 68, No. 3, pp. 329-338, May 5, 2000.
Celeste Nelson et al., Emergent patterns of growth controlled by multicellular form and mechanics, (in Christopher Chen's Lab demonstrated, in separate experiments, that curved surfaces with a radius of curvature (200 ?m) that is greater than the cell diameter and surfaces that have undulating special patterning (depressions) increase the patterned growth of ECs [PNAS 102(33): 11594-11599, 2005].
Chapman NM, Chi H. mTOR signaling, Tregs and immune modulation. Immunotherapy. 2014;6(12):1295-311.
Chaudhry A, Samstein RM, Treating P, Liang Y, Pils MC, Heinrich JM, Jack RS, Wunderlich FT, Bruning JC, Muller W, Rudensky AY. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 2011;34(4):566-78.
Chen, C. and Broden, M., “The Role of Poly(theylene glycol) Brush Architecture in Complement Activation on Targeted Microbubble Surfaces,” Biomaterials, vol. 32, No. 27, pp. 6579-6587, Jun. 17, 2011.
Choi W, Kwon SJ, Jin HJ, et al. (2017) Optimization of culture conditions for rapid clinical-scale expansion of human umbilical cord blood-derived mesenchymal stem cells. Clin Transl Med 6(1): 38.
Chullikana A, Majumdar AS, Gottipamula S, et al. (2015) Randomized, double-blind, phase l/ll study of intravenous allogeneic mesenchymal stromal cells in acute myocardial infarction. Cytotherapy 17(3): 250-261.
Coeshott C, Vang B, Jones M, Nankervis B. Large-scale expansion and characterization of CD3(+) T-cells in the Quantum((R)) Cell Expansion System. J Transl Med. 2019;17(1):258.
Coombes JL, Robinson NJ, Maloy KJ, Uhlig HH, Powrie F. Regulatory T cells and intestinal homeostasis. Immunol Rev. 2005;204:184-94.
Coquillard C. mTOR Signaling in Regulatory T cell Differentiation and Expansion. SOJ Immunology. 2015;3(1):1-10.
Creed JA, DiLeonardi AM, Fox DP, Tessier AR, Raghupathi R. Concussive brain trauma in the mouse results in acute cognitive deficits and sustained impairment of axonal function. Journal of neurotrauma. 2011 ;28:547-563.
Dash PK, Hochner B, Kandel ER. Injection of the camp-responsive element into the nucleus of aplysia sensory neurons blocks long-term facilitation. Nature. 1990;345:718-721.
Dash PK, Johnson D, Clark J, Orsi SA, Zhang M, Zhao J, Grill RJ, Moore AN, Pati S. Involvement of the glycogen synthase kinase-3 signaling pathway in tbi pathology and neurocognitive outcome. PloS one. 2011;6:e24648.
Dash PK, Mach SA, Blum S, Moore AN. Intrahippocampal wortmannin infusion enhances long-term spatial and contextual memories. Learn Mem. 2002;9:167-177.
Dash PK, Orsi SA, Zhang M, Grill RJ, Pati S, Zhao J, Moore AN. Valproate administered after traumatic brain injury provides neuroprotection and improves cognitive function in rats. PloS one. 2010;5:e11383.
Dash PK, Zhao J, Orsi SA, Zhang M, Moore AN. Sulforaphane improves cognitive function administered following traumatic brain injury. Neuroscience letters. 2009;460:103-107.
Davila et al., “Efficacy and Toxicity Management of 19-28z Car T Cell Therapy in B cell Acute Lymphoblastic Leukemia,” Science Translational Medicine, vol. 6, No. 224, pp. 1-10, Feb. 19, 2014.
Dejana E, Orsenigo F, Lampugnani MG. The role of adherens junctions and ve-cadherin in the control of vascular permeability. Journal of cell science. 2008;121:2115-2122.
Dejana E, Spagnuolo R, Bazzoni G. Interendothelial junctions and their role in the control of angiogenesis, vascular permeability and leukocyte transmigration. Thrombosis and haemostasis. 2001;86:308-315.
Dejana E, Toumier-Lasserve E, Weinstein BM. The control of vascular integrity by endothelial cell junctions: Molecular basis and pathological implications. Developmental cell. 2009;16:209-221.
Del Pino A, Ligero G, Lopez MB, et al. (2015) Morphology, cell viability, karyotype, expression of surface markers and plasticity of three primary cell line cultures before and after the cryostorage in LN2 and GN2. Cryobiology 70(1): 1-8.
Delaney, Colleen, et al. “Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution.” Nature medicine 16.2 (2010): 232-236.
Ding, Zhongli, Guohua Chen, and Allan S. Hoffman. “Synthesis and purification of thermally sensitive oligomer? enzyme conjugates of poly (N-isopropylacrylamide)? trypsin.” Bioconjugate chemistry 7.1 (1996): 121-125.
Dixon CE, Clifton GL, Lighthall JW, Yaghmai AA, Hayes RL. A controlled cortical impact model of traumatic brain injury in the rat. Journal of neuroscience methods. 1991;39:253-262.
Dominici M, Le Blanc K, Mueller I, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8(4): 315-317.
Durrani S, Konoplyannikov M, Ashraf M, Haider KH (2010) Skeletal myoblasts for cardiac repair. Regen Med 5(6): 919-932.
Esensten JH, Muller YD, Bluestone JA, Tang Q. Regulatory T-cell therapy for autoimmune and autoinflammatory diseases: The next frontier. J Allergy Clin Immunol. 2018;142(6):1710-1718.
Fakin R, Hamacher J, GuggerM, GazdharA, Moser H, Schmid RA. Prolonged amelioration of acute lung allograft rejection by sequential overexpression of human interleukin-10 and hepatocyte growth factor in rats. Exp Lung Res. 2011;37(9):555-62.
Fedorov et al., “PD-1- and CTLA-4-Based Inhibitory Chimeric Antigen Receptors (iCARs) Divert Off-Target Immunotherapy Responses,” Science Translational Medicine, vol. 5, No. 215, pp. 1-12, Dec. 11, 2013.
Ferreira LMR, Muller YD, Bluestone JA, Tang Q. Next-generation regulatory T cell therapy. Nat Rev Drug Discov. 2019;18(10):749-769.
Fischbach, Michael A., Jeffrey A. Bluestone, and Wendell A. Lim. “Cell-based therapeutics: the next pillar of medicine.” Science translational medicine 5.179 (2013): 179ps7-179ps7.
Fisk, Nicholas M., et al. “Can routine commercial cord blood banking be scientifically and ethically justified?.” PLoS medicine 2.2 (2005): e44.
Forbes Jun. 23, 2014 article “Will this man cure cancer?”.
Fowler DH. Rapamycin-resistant effector T-cell therapy. Immunol Rev. 2014;257(1):210-225.
Fraser H, Safinia N, Grageda N, Thirkell S, Lowe K, Fry LJ, Scotta C, Hope A, Fisher C, Hilton R, Game D, Harden P, Bushell A, Wood K, Lechler RI, Lombardi G. A Rapamycin-Based GMP-Compatible Process for the Isolation and Expansion of Regulatory T Cells for Clinical Trials. Mol Ther Methods Clin Dev. 2018;8:198-209.
Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002;16(6):769-777.
Fuchs A, Gliwinski M, Grageda N, Spiering R, Abbas AK, Appel S, Bacchetta R, Battaglia M, Berglund D, Blazar B, Bluestone JA, Bomhauser M, Ten Brinke A, Brusko TM, Cools N, Cuturi MC, Geissler E, Giannoukakis N, Golab K, Hafler DA, van Ham SM, Hester J et al. Minimum Information about T Regulatory Cells: A Step toward Reproducibility and Standardization. Front Immunol. 2017;8:1844.
G0211: Study for Gamma Irradiation of Bioreactor Membranes, undated, author unknown, 3 pages.
Galgani M, De Rosa V, La Cava A, Matarese G. Role of Metabolism in the Immunobiology of Regulatory T Cells. J Immunol. 2016;197(7):2567-2575.
Garlie, Nina K., et al. “T cells coactivated with immobilized anti-CD3 and anti-CD28 as potential immunotherapy for cancer.” Journal of immunotherapy (Hagerstown, Md.: 1997) 22.4 (1999): 336-345.
Gedaly R, De Stefano F, Turcios L, Hill M, Hidalgo G, Mitov Ml, Alstott MC, Butterfield DA, Mitchell HC, Hart J, Al-Attar A, Jennings CD, Marti F. mTOR Inhibitor Everolimus in Regulatory T Cell Expansion for Clinical Application in Transplantation. Transplantation. 2019;103(4):705-715.
Gimble, Jeffrey M., Adam J. Katz, and Bruce A. Bunnell. “Adipose-derived stem cells for regenerative medicine.” Circulation research 100.9 (2007): 1249-1260.
Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/mTOR. Genes Dev. 2001;15(7):807-826.
Godin, Michel, et al. “Measuring the mass, density, and size of particles and cells using a suspended microchannel resonator.” Applied physics letters 91.12 (2007): 123121.
Golab K, Leveson-Gower D, Wang XJ, Grzanka J, Marek-Trzonkowska N, Krzystyniak A, Millis JM, Trzonkowski P, Witkowski P. Challenges in cryopreservation of regulatory T cells (Tregs) for clinical therapeutic applications. Int Immunopharmacol. 2013;16(3):371-5.
Goldring CE, Duffy PA, Benvenisty N, Andrews PW, Ben-David U, Eakins R, French N, Hanley NA, Kelly L, Kitteringham NR, Kurth J, Ladenheim D, Laverty H, McBlane J, Narayanan G, Patel S, Reinhardt J, Rossi A, Sharpe M, Park BK. Assessing the safety of stem cell therapeutics. Cell stem cell. 2011;8:618-628.
Griesche, Nadine, et al. “A simple modification of the separation method reduces heterogeneity of adipose-derived stem cells.” cells tissues organs 192.2 (2010): 106-115.
Gutcher I, Donkor MK, Ma Q, Rudensky AY, Flavell RA, Li MO. Autocrine transforming growth factor-betal promotes in vivo Th17 cell differentiation. Immunity. 2011 ;34(3):396-408.
Haack-Sorensen M, Follin B, Juhl M, et al. (2016) Culture expansion of adipose derived Stromal cells. A closed automated Quantum Cell Expansion System compared with manual flask-based culture. J Transl Med 14(1): 319.
Hall ED, Sullivan PG, Gibson TR, Pavel KM, Thompson BM, Scheff SW. Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: More than a focal brain injury. Journal of neurotrauma. 2005;22:252-265.
Hami et al., “GMP Production and Testing of Xcellerated T Cells for the Treatment of Patients with CLL,” Cytotherapy, pp. 554-562, 2004.
Hamm RJ, Dixon CE, Gbadebo DM, Singha AK, Jenkins LW, Lyeth BG, Hayes RL. Cognitive deficits following traumatic brain injury produced by controlled cortical impact. Journal of neurotrauma. 1992;9:11-20.
Hanley PJ, Mei Z, Durett AG, et al. (2014) Efficient manufacturing of therapeutic mesenchymal stromal cells with the use of the Quantum Cell Expansion System. Cytotherapy 16(8): 1048-1058.
He N, Fan W, Henriquez B, Yu RT, Atkins AR, Liddle C, Zheng Y, Downes M, Evans RM. Metabolic control of regulatory T cell (Treg) survival and function by Lkb1. Proc Natl Acad Sci USA. 2017;114(47):12542-12547.
He X, Landman S, Bauland SC, van den Dolder J, Koenen HJ, Joosten I. A TNFR2-Agonist Facilitates High Purity Expansion of Human Low Purity Treg Cells. PLoS One. 2016;11(5):e0156311.
Heskins, Michael, and James E. Guillet. “Solution properties of poly (N-isopropylacrylamide).” Journal of Macromolecular Science—Chemistry 2.8 (1968): 1441-1455.
Hill JA, Feuerer M, Tash K, Haxhinasto S, Perez J, Melamed R, Mathis D, Benoist C. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity. 2007;27(5):786-800.
Högstedt, Benkt, Anita Karlsson, and Anders Holmen. “Frequency and size distribution of micronuclei in lymphocytes stimulated with phytohemagglutinin and pokeweed mitogen in workers exposed to piperazine.” Hereditas 109.(1988): 139-142.
Hollyman et al., “Manufacturing Validation of Biologicall Functional T Cells Targeted to CD19 Antigen for Autologous Adoptive Cell Therapy,” J Immunother, vol. 32, No. 2, pp. 169-180, Feb.-Mar. 2009.
http://www.ucdenver.edU/academics/colleges/medicalschool/centers/cancercenter/Research/sharedresources/Animallmaging/smallanimalimaging/Pages/MRI.aspx.
ISCT Webinar “Volume Reduction technology for Large Scale Harvest or Post-thaw Manipulation of Cellular Therapeutics”.
Iwashima, Shigejiro, et al. “Novel culture system of mesenchymal stromal cells from human subcutaneous adipose tissue.” Stem cells and development 18.4 (2009): 533-544.
Jarocha D, Stangel-Wojcikiewicz K, Basta A, Majka M (2014) Efficient myoblast expansion for regenerative medicine use. Int J Mol Med 34(1): 83-91.
Jo CH, Lee YG, Shin WH, et al. (2014) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells 32(5): 1254-1266.
John Carvell, et al. Monitoring Live Biomass in Disposable Bioreactors, BioProcess International 14(3)s, Mar. 2016.
John Nicolette, et al. (Abbott Laboratories). In Vitro Micronucleus Screening of Pharmaceutical Candidates by Flow Cyto9metry in Chinese Hamster V79 Cells, Environmental and Molecular Mutagenesis 00:000-000, 2010.
John P. Carvell and Jason E. Dowd. On-line measurements and control of viable cell density in cell culture manufacturing processes using radio frequency impedance. Cytotechnology 50: 35-48, 2006.
Johnson, Patrick A., et al. “Interplay of anionic charge, poly (ethylene glycol), and iodinated tyrosine incorporation within tyrosine?derived polycarbonates: Effects on vascular smooth muscle cell adhesion, proliferation, and motility.” Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials 93.2 (2010): 505-514.
Johnston LC, Su X, Maguire-Zeiss K, Horovitz K, Ankoudinova I, Guschin D, Hadaczek P, Federoff HJ, Bankiewicz K, Forsayeth J. Human interleukin-10 gene transfer is protective in a rat model of Parkinson's disease. Mol Ther. 2008;16(8):1392-1399.
Jones M, Varella-Garcia M, Skokan M, et al. (2013) Genetic stability of bone marrow-derived human mesenchymal stromal cells in the Quantum System. Cytotherapy 15(11): 1323-1339.
Jones2016ISCT 2016 Poster 69.
Joy, Abraham, et al. “Control of surface chemistry, substrate stiffness, and cell function in a novel terpolymer methacrylate library.” Langmuir 27.5 (2011): 1891-1899.
Kalamasz et al., “Optimization of Human T-Cell Expansion Ex Vivo Using Magnetic Beads Conjugated with Anti-CD3 and Anti-CD28 Antibodies,” J Immunother, vol. 27, No. 5, pp. 405-418, Sep.-Oct. 2004.
Klapper et al., “Single-Pass, Closed-System Rapid Expansion of Lymphocyte Cultures for Adoptive Cell Therapy,” Journal of Immunological Methods, 345, pp. 90-99, Apr. 21, 2009.
Korpanty et al., “Tageting Vascular Enothelium with Avidin Microbubbles,” Ultrasound in Medicine and Biology, vol. 31, No. 9, pp. 1279-1283, May 24, 2005.
Krauss et al., “Signaling Takes a Breath—New Quantitative Perspectives on Bioenergetics and Signal Transduction,” Immunity, vol. 15, pp. 497-502, Oct. 2001.
Kulikov, A. V., et al. “Application of multipotent mesenchymal stromal cells from human adipose tissue for compensation of neurological deficiency induced by 3-nitropropionic acid in rats.” Bulletin of experimental biology and medicine 145.4 (2008): 514-519.
Kumar P, Marinelarena A, Raghunathan D, Ragothaman VK, Saini S, Bhattacharya P, Fan J, Epstein AL, Maker AV, Prabhakar BS. Critical role of 0X40 signaling in the TCR-independent phase of human and murine thymic Treg generation. Cell Mol Immunol. 2019;16(2):138-153.
Kwan, J. and Borden, M., “Lipid Monolayer Collapse and Microbubble Stability,” Advances in Colloid and Interface Science, vols. 183-184, pp. 82-99, Aug. 21, 2012.
Lee et al., “Continued Antigen Stimulation Is Not Required During CD4+ T Cell Clonal Expansion,” The Journal of Immunology, 168, pp. 1682-1689, 2002.
Lee, Jae W., et al. “Allogeneic human mesenchymal stem cells for treatment of E. coll endotoxin-induced acute lung injury in the ex vivo perfused human lung.” Proceedings of the national academy of Sciences 106.38 (2009): 16357-16362.
Levine, B., “T Lymphocyte Engineering ex vivo for Cancer and Infectious Disease,” Expert Opinion on Biological Therapy, vol. 4, No. 4, pp. 475-489, 2008.
Lum et al., “Ultrasound Radiation Force Enables Targeted Deposition of Model Drug Carriers Loaded on Microbubbles,” Journal of Controlled Release, 111, pp. 128-134, 2006.
M. R. Koller, et al. Clinical-scale human umbilical cord blood cell expansion in a novel automated perfusion culture system. Bone Marrow Transplantion 21:653-663, 1998.
Malone et al., “Characterization of Human Tumor-Infiltrating Lymphocytes Expanded in Hollow-Fiber Bioreactors for Immunotherapy of Cancer,” Cancer Biotherapy & Radiopharmaceuticals, vol. 16, No. 5, pp. 381-390, 2001.
Mao AS, Mooney DJ (2015) Regenerative medicine: current therapies and future directions. Proc Natl Acad Sci USA 112(47): 14452-14459.
Maria Streltsova, Dean Lee (Nationwide Children's Hospital, OSU, Columbus, OH) et al. (Int'l Journal of Molecular Sciences, 2019).
Markgraf CG, Clifton GL, Aguirre M, Chaney SF, Knox-Du Bois C, Kennon K, Verma N. Injury severity and sensitivity to treatment after controlled cortical impact in rats. Journal of neurotrauma. 2001;18:175-186.
Mathew et al. A Phase I Clinical Trials I with Ex Vivo Expanded Recipient Regulatory T cells in Living Donor Kidney Transplants. Nature, Scientific Reports 8:7428 (1-12), 2018.
Matthay, Michael A., et al. “Therapeutic potential of mesenchymal stem cells for severe acute lung injury.” Chest 138.4 (2010): 965-972.
Maynard CL, Harrington LE, Janowski KM, Oliver JR, Zindl CL, Rudensky AY, Weaver CT. Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3- precursor cells in the absence of interleukin 10. Nat Immunol. 2007;8(9):931-41.
McKenna DH, Jr., Sumstad D, Kadidlo DM, et al. Optimization of cGMP purification and expansion of umbilical cord blood-derived T-regulatory cells in support of first-in-human clinical trials. Cytotherapy 2017;19:250-62.
McLimans W, Kinetics of Gas Diffusion in Mammalian Cell Culture Systems. Biotechnology and Bioengineering 1968; 10:725-740.
McMurtrey, Richard J. “Analytic models of oxygen and nutrient diffusion, metabolism dynamics, and architecture optimization in three-dimensional tissue constructs with applications and insights in cerebral organoids.” Tissue Engineering Part C: Methods 22.3 (2016): 221-249.
Menge, Tyler, et al. “Mesenchymal stem cells regulate blood-brain barrier integrity through TIMP3 release after traumatic brain injury.” Science translational medicine 4.161 (2012): 161ra150-161ra150.
Miska J, Lee-Chang C, Rashidi A, Muroski ME, Chang AL, Lopez-Rosas A, Zhang P, Panek WK, Cordero A, Han Y, Ahmed AU, Chandel NS, Lesniak Ms. HIF-1 alpha Is a Metabolic Switch between Glycolytic-Driven Migration and Oxidative Phosphorylation-Driven Immunosuppression of Tregs in Glioblastoma. Cell Rep. 2019;27(1):226-237 e4.
Miyara M, Yoshioka Y, Kitoh A, Shima T, Wng K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899-911.
Murugappan, G., et al. “Human hematopoietic progenitor cells grow faster under rotational laminar flows.” Biotechnology progress 26.5 (2010): 1465-1473.
Nankervis B, Jones M, Vang B et al. (2018) Optimizing T Cell Expansion in a Hollow-Fiber Bioreactor. Curr Stem Cell Rep. Advanced online publication, https://doi.org/10.1007/s40778-018-0116-x.
Nankervis, Brian, et al. “Optimizing T cell expansion in a hollow-fiber bioreactor.” Current Stem Cell Reports 4.1 (2018): 46-51.
Nedoszytko B, Lange M, Sokolowska-Wojdylo M, Renke J, Trzonkowski P, Sobjanek M, Szczerkowska-Dobosz A, Niedoszytko M, Gorska A, Romantowski J, Czarny J, Skokowski J, Kalinowski L, Nowicki R. The role of regulatory T cells and genes involved in their differentiation in pathogenesis of selected inflammatory and neoplastic skin diseases. Part II: The Treg role in skin diseases pathogenesis. Postepy Dermatol Alergol. 2017;34(5):405-417.
Nehlin JO, Just M, Rustan AC (2011) Human myotubes from myoblast cultures undergoing senescence exhibit defects in glucose and lipid metabolism. Biogerontology 12: 349-365.
New victories for adult Stem Cell Research New York Feb. 6, 2007.
Newton R, Priyadharshini B, Turka LA. Immunometabolism of regulatory T cells. Nat Immunol. 2016;17(6):618-25.
Ng TH, Britton GJ, Hill EV, Verhagen J, Burton BR, Wraith DC. Regulation of adaptive immunity; the role of interleukin-10. Front Immunol. 2013;4:129.
Nikolaychik, V. V., M. M. Samet, and P. I. Lelkes. “A New, Cryoprecipitate Based Coating For Improved Endothelial Cell Attachment And Growth On Medical Grade Artificial Surfaces.” ASAIO Journal (American Society for Artificial Internal Organs: 1992) 40.3 (1994): M846-52.
Nish SA, Schenten D, Wunderlich FT, Pope SD, Gao Y, Hoshi N, Yu S, Yan X, Lee HK, Pasman L, Brodsky I, Yordy B, Zhao H, Bruning J, Medzhitov R. T cell-intrinsic role of IL-6 signaling in primary and memory responses. Elife. 2014;3:e01949.
Niwayama, Jun, et al. “Analysis of hemodynamics during blood purification therapy using a newly developed noninvasive continuous monitoring method.” Therapeutic Apheresis and Dialysis 10.4 (2006): 380-386.
Nugent, Helen M., et al. “Adventitial endothelial implants reduce matrix metalloproteinase-2 expression and increase luminal diameter in porcine arteriovenous grafts.” Journal of vascular surgery 46.3 (2007): 548-556.
Okano et al. (Tokyo Women's Medical College, Japan) demonstrated the recovery of endothelial cells and hepatocytes from plasma-treated polystyrene dishes grafted with PNIAAm (Journal of Biomedical Materials Research, 1993).
Onishi Y, Fehervari Z, Yamaguchi T, Sakaguchi S. Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. Proc Natl Acad Sci USA. 2008; 105(29): 10113-10118.
Onyszchuk G, LeVine SM, Brooks WM, Berman NE. Post-acute pathological changes in the thalamus and internal capsule in aged mice following controlled cortical impact injury: A magnetic resonance imaging, iron histochemical, and glial immunohistochemical study. Neuroscience letters. 2009;452:204-208.
Pacella I, Procaccini C, Focaccetti C, Miacci S, Timperi E, Faicchia D, Severa M, Rizzo F, Coccia EM, Bonacina F, Mitro N, Norata GD, Rossetti G, Ranzani V, Pagani M, Giorda E, Wei Y, Matarese G, Barnaba V, Piconese S. Fatty acid metabolism complements glycolysis in the selective regulatory T cell expansion during tumor growth. Proc Natl Acad Sci USA. 2018;115(28):E6546-E6555.
Parhi, Purnendu, Avantika Goias, and Erwin A. Vogler. “Role Of Proteins And Water In The Initial Attachment Of Mammalian Cells To Biomedical Surfaces: A Review.” Journal of Adhesion Science and Technology 24.5 (2010): 853-888.
Pati S, Gerber MH, Menge TD, Wataha KA, Zhao Y, Baumgartner JA, Zhao J, Letourneau PA, Huby MP, Baer LA, Salsbury JR, Kozar RA, Wade CE, Walker PA, Dash PK, Cox CS, Jr., Doursout MF, Holcomb JB. Bone marrow derived mesenchymal stem cells inhibit inflammation and preserve vascular endothelial integrity in the lungs after hemorrhagic shock. PloS one. 2011;6:e25171.
Pati S, Khakoo AY, Zhao J, Jimenez F, Gerber MH, Harting M, Redell JB, Grill R, Matsuo Y, Guha S, Cox CS, Reitz MS, Holcomb JB, Dash PK. Human mesenchymal stem cells inhibit vascular permeability by modulating vascular endothelial cadherin/beta-catenin signaling. Stem cells and development. 2011;20:89-101.
Peters JH, Preijers FW, Woestenenk R, Hilbrands LB, Koenen HJ, Joosten I. Clinical grade Treg: GMP isolation, improvement of purity by CD127 Depletion, Treg expansion, and Treg cryopreservation. PLoS One. 2008;3(9):e3161.
Peters, R.; Jones, M.; Brecheisen, M.; Startz, T.; Vang, B.; Nankervis, B.; Frank, N.; Nguyen, K. (2012) TerumoBCT. https://www.terumobct.com/location/north-america/products-and-services/Pages/Quantum-Materials.aspx.
Porter CM, Horvath-Arcidiacono JA, Singh AK, Horvath KA, Bloom ET, Mohiuddin MM. Characterization and expansion of baboon CD4+CD25+ Treg cells for potential use in a nonhuman primate xenotransplantation model. Xenotransplantation. 2007;14(4):298-308.
Povsic TJ, O'Connor CM, Henry T, et al. (2011) A double-blind, randomized, controlled, multicenter study to assess the safety and cardiovascular effects of skeletal myoblast implantation by catheter delivery in patients with chronic heart failure after myocardial infarction. Am Heart J 162(4): 654-662.
Prockop, Darwin J., Carl A. Gregory, and Jeffery L. Spees. “One strategy for cell and gene therapy: harnessing the power of adult stem cells to repair tissues.” Proceedings of the National Academy of Sciences 10O.suppl_1 (2003): 11917-11923.
Q. L. Hao, et al. A functional comparison of CD34+ CD38= cells in cord blood and bone marrow. Blood 86:3745-3753, 1995.
Rahmahwati, Nurlaela, Deana Wahyuningrum, and Anita Alni. “The Synthesis Of Polyethersulfone (PES) Derivatives For The Immobilization Of Lipase Enzyme.” Key Engineering Materials. Vol. 811. Trans Tech Publications Ltd, 2019.
Rey-Jurado, Emma, et al. “Assessing the importance of domestic vaccine manufacturing centers: an overview of immunization programs, vaccine manufacture, and distribution.” Frontiers in immunology 9 (2018): 26.
Roballo KC, Dhungana S, Z. J, Oakey J, Bushman J. Localized delivery of immunosuppressive regulatory T cells to peripheral nerve allografts promotes regeneration of branched segmental defects. Biomaterials. 2019;209:1-9.
Rodrigues, C., Fernandes, T., Diogo, M., Lobato da Silva, C., Cabral, J. Stem Cell Cultivation in Bioreactors. 2011. Biotechnology Advances v. 29, pp. 815-829.
Ronco C1, Levin N, Brendolan A, Nalesso F, Cruz D, Ocampo C, Kuang D, Bonello M, De Cal M, Corradi V, Ricci Z. Flow distribution analysis by helical scanning in polysulfone hemodialyzers: effects of fiber structure and design on flow patterns and solute clearances. Hemodial Int. Oct. 2006; 10(4):380-8.
Rosenblum MD, Way SS, Abbas AK. Regulatory T cell memory. Nat Rev Immunol. 2016;16(2):90-101.
Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, Treuting P, Siewe L, Roers A, Henderson WR, Jr., Muller W, Rudensky AY. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity. 2008;28(4):546-558.
Rudensky, Alexander Y. “Regulatory T cellsand Foxp3.” Immunological reviews 241.1 (2011): 260-268.
S. Koestenbauer, et al. Protocols for Hematopoietic Stem Cell Expansion from Umbilical Cord Blood. Cell Transplantation 18: 1059-1068, 2009.
S. L. Smith, et al. Expansion of neutrophil precursors and progenitors in suspension cultures of CD34+ cells enriched from human bone marrow. Experimental Hematology 21:870-877, 1993.
Safinia N, Grageda N, Scotta C, Thirkell S, Fry LJ, Vaikunthanathan T, Lechler RI, Lombardi G. Cell Therapy in Organ Transplantation: Our Experience on the Clinical Translation of Regulatory T Cells. Front Immunol. 2018;9:354.
Sahay A, Scobie KN, Hill AS, O'Carroll CM, Kheirbek MA, Burghardt NS, Fenton AA, Dranovsky A, Hen R. Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation. Nature. 2011;472:466-470.
Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155(3):1151-64.
Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T, Itoh M, Kuniyasu Y, Nomura T, Toda M, Takahashi T. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev. 2001;182:18-32.
Schild, Howard G. “Poly (N-isopropylacrylamide): experiment, theory and application.” Progress in polymer science 17.2 (1992): 163-249.
Schmitz R, Alessio A, Kina P. The Physics of PET/CT scanners. Imaging Research Laboratory, Department of Radiology, University of Washington http://depts.washington.edu/imreslab/education/Physics%20of%20PET.pdf.
Schwartz RH. T cell anergy. Annu Rev Immunol. 2003;21:305-34.
Shevkoplyas et al., “The Force Acting on a Superparamagnetic Bead due to an Applied Magnetic Field,” Lab on a Chip , 7, pp. 1294-1302, 2007.
Shimazu Y, Shimazu Y, Hishizawa M, Hamaguchi M, Nagai Y, Sugino N, Fujii S, Kawahara M, Kadowaki N, Nishikawa H, Sakaguchi S, Takaori-Kondo A. Hypomethylation of the Treg-Specific Demethylated Region in FOXP3 Is a Hallmark of the Regulatory T-cell Subtype in Adult T-cell Leukemia. Cancer Immunol Res. 2016;4(2): 136-45.
Shimizu et al. (TWMU & Heart Institute of Japan) described the detachment of avian cardiomyocytes from PIPAAm matrixes that were observed to pulse spontaneously with neovascularization in layered sheets three (3) weeks after transplantation (Circulation Research, 2002).
Sigma-Aldrich Cheimcals Mitomycin C (M4287) MSDS, v4.4, Jul. 7, 2011.
Sirsi, S. and Borden, M., “Microbubble Composition, Properties, and Biomedical Applications,” Bubble Science, Engineering & Technolology, vol. 1, No. 1-2, pp. 3-17, 2009.
Smith C, Okern G, Rehan S, et al. Ex vivo expansion of human T cells for adoptive immunotherapy using the novel Xeno-free CTS Immune Cell Serum Replacement. Clinical & Translational Immunology 2015;4:e31.
Somerville et al., “Clinical Scale Rapid Expansion of Lymphocytes for Adoptive Cell Transfer Therapy in the WAVE® Bioreactor,” Journal of Translational Medicine, vol. 10, No. 69, pp. 1-11, 2012.
Somerville, R. and Dudley, M., “Bioreactors Get Personal,” Oncolmmunology, vol. 1, No. 8, pp. 1435-1437, Nov. 2012.
Spectrum Labs KrosFlo Research IIi TFF System, undated, Spectrum Laboratories, Inc., 4 pages.
Stafano Tiziani, et al. Metabolomic Profiling of Drug Response in Acute Myeloid Leukaemia Cell lines. PLOSone 4(1): e4251 (Jan. 22, 2009).
StAR_Abstract, undated, author unknown, 1 page.
Startz et al May 2016 TBCT T-cell White Paper.
Startz, T., et al. “Maturation of dendritic cells from CD14+ monocytes in an automated functionally closed hollow fiber bioreactor system.” Cytotherapy 16.4 (2014): S29.
Steven M. Bryce, et al. (Litron Laboratories). In vitro micronucleus assay scored by flow cytometry provides a comprehensive evaluation of cytogenetic damage and cytotoxicity. Mutation Research 630(1-2): 78-91, 2007.
Steven M. Bryce, et al. (Novartis Pharma AG, Johnson & Johnson Pharmaceutical Research, GlaxoSmithKline). Interlaboratory evaluation of a flow cytometric, high content in vitro micronucleus assay. Genetic Toxicology and Environmental Mutagenesis 650: 181-195, 2008.
Stuart, Martien A. Cohen, et al. “Emerging applications ofstimuli-responsive polymer materials.” Nature materials 9.2 (2010): 101-113.
Su LF, Del Alcazar D, Stelekati E, Wherry EJ, Davis MM. Antigen exposure shapes the ratio between antigen-specific Tregs and conventional T cells in human peripheral blood. Proc Natl Acad Sci USA. 2016;113(41):E6192-E6198.
The effect of rocking rate and angle on T cell cultures grown in Xuri™ Cell Expansion Systems, Aug. 2014, GE Healthcare UK Limited, 4 pages.
Trzonkowski et al., “Ex Vivo Expansion of CD4+ CD25+ T Regulatory Cells for Immunosuppressive Therapy,” Cytometry Part A, 75A, pp. 175-188, 2009.
Trzonkowski, Piotr, et al. “First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+ CD25+ CD127? T regulatory cells.” Clinical immunology 133.1 (2009): 22-26.
Tsvetkov, Ts, et al. “Isolation and cryopreservation of human peripheral blood monocytes.” Cryobiology 23.6 (1986): 531-536.
Underwood, P. Anne, et al. “Effects of base material, plasma proteins and FGF2 on endothelial cell adhesion and growth.” Journal of Biomaterials Science, Polymer Edition 13.8 (2002): 845-862.
Urbich, et al. from the Goethe-Universitat, demonstrated that human endothelial cells increased VEGFR-2 mRNA expression when exposed to 5-15 dynes/cm2 of constant shear force for a period of 6-24 hours (FEBS, 2002).
Van der Net JB, Bushell A, Wood KJ, Harden PN. Regulatory T cells: first steps of clinical application in solid organ transplantation. Transpl Int. 2016;29(1):3-11.
Van der Windt GJ, Pearce EL. Metabolic switching and fuel choice during T-cell differentiation and memory development. Immunol Rev. 2012;249(1):27-42.
Vera et al., “Accelerated Production of Antigen-Specific T-Cells for Pre-Clinical and Clinical Applications Using Gas-Permeable Rapid Expansion Cultureware (G-Rex),” J Immunother, vol. 33, No. 3, pp. 305-315, Apr. 2010.
Villa, Alma Y. Camacho, et al. “CD133+ CD34+ and CD133+ CD38+ blood progenitor cells as predictors of platelet engraftment in patients undergoing autologous peripheral blood stem cell Transplantation.” Transfusion and Apheresis Science 46.3 (2012): 239-244.
Visser EP1, Disselhorst JA, Brom M, Laverman P, Gotthardt M, Oyen WJ, Boerman OC. Spatial resolution and sensitivity of the Inveon small-animal PET scanner. J Nucl Med. Jan. 2009;50(1):139-147.
Von Laer, D., et al. “Loss of CD38 antigen on CD34+ CD38+ cells during short-term culture.” Leukemia 14.5 (2000): 947-948.
Wagner Jr, John E., et al. “Phase l/ll trial of StemRegenin-1 expanded umbilical cord blood hematopoietic stem cells supports testing as a stand-alone graft.” Cell stem cell 18.1 (2016): 144-155.
Walker, Peter A., et al. “Direct intrathecal implantation of mesenchymal stromal cells leads to enhanced neuroprotection via an NF?B-mediated increase in interleukin-6 production.” Stem cells and development 19.6 (2010): 867-876.
Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger J, Green DR. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity. 2011;35(6):871-82.
Wang, Jiamian, John A. Jansen, and Fang Yang. “Electrospraying: possibilities and challenges of engineering carriers for biomedical applications—a mini review.” Frontiers in Chemistry 7 (2019): 258.
Ward H, Vigues S, Poole S, Bristow AF. The rat interleukin 10 receptor: cloning and sequencing of cDNA coding for the alpha-chain protein sequence, and demonstration by western blotting of expression in the rat brain. Cytokine. 2001;15(5):237-240.
Wawman, Rebecca Ellen, Helen Bartlett, and Ye Htun Oo. “Regulatory T cell metabolism in the hepatic microenvironment.” Frontiers in immunology 8 (2018): 1889.
Weber et al., “White Paper on Adoptive Cell Therapy for Cancer with Tumor-Infiltrating Lymphocytes: A Report of the CTEP Subcommittee on Adoptive Cell Therapy,” Clinical Cancer Research, vol. 17, No. 7, pp. 1664-1673, Apr. 1, 2011.
Weiss RA, Weiss MA, Beasley KL, Munavalli G (2007) Autologous cultured fibroblast injection for facial contour deformities: a prospective, placebo-controlled, Phase III clinical trial. Dermatol Surg 33(3): 263-268.
Widdel, F. 2010. “Theory and measurement of bacterial growth” http://www.mpi-bremen.de/Binaries/Binary13037/Wachstumsversuch.pdf.
Yamada, Noriko, et al. “Thermo?responsive polymeric surfaces; control of attachment and detachment of cultured cells.” Die Makromolekulare Chemie, Rapid Communications 11.11 (1990): 571-576.
Yang, Hee Seok, et al. “Suspension culture of mammalian cells using thermosensitive microcarrier that allows cell detachment without proteolytic enzyme treatment.” Cell transplantation 19.9 (2010): 1123-1132.
Yi, Zhuan, et al. “A readily modified polyethersulfone with amino-substituted groups: its amphiphilic copolymer synthesis and membrane application.” Polymer 53.2 (2012): 350-358.
Yoshinari, Masao, et al. “Effect of cold plasma-surface modification on surface wettability and initial cell attachment.” International Journal of Biomedical and Biological Engineering 3.10 (2009): 507-511.
Zappasodi et al., “The Effect Of Artificial Antigen-Presenting Cells with Preclustered Anit-CD28/-CD3/LFA-1 Monoclonal Antibodies on the Induction of ex vivo Expansion of Functional Human Antitumor T Cells,” Haematologica, vol. 93, No. 10, pp. 1523-1534, 2008.
Zemmour D, Zilionis R, Kiner E, Klein AM, Mathis D, Benoist C. Publisher Correction: Singlecell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat Immunol. 2018;19(6):645.
Zeng B, Kwak-Kim J, Liu Y, Liao AH. Treg cells are negatively correlated with increased memory B cells in pre-eclampsia while maintaining suppressive function on autologous B-cell proliferation. Am J Reprod Immunol. 2013;70(6):454-463.
Zheng, et al. at the University of Iowa have shown that the differential effects of pulsatile blood flow and cyclic stretch are an important growth stimulus (American Journal of Physiology—Heart and Circulatory Physiology, 2008).
Anamelechi, Charles C., et al. “Streptavidin binding and endothelial cell adhesion to biotinylated fibronectin.” Langmuir 23.25 (2007): 12583-12588.
Barker, Juliet N., et al. “CD34+ cell content of 126 341 cord blood units in the US inventory: implications for transplantation and banking.” Blood advances 3.8 (2019): 1267-1271.
Bluestone, Jeffrey A., et al. “Type 1 diabetes immunotherapy using polyclonal regulatory T cells.” Science translational medicine 7.315 (2015): 315ra189-315ra189.
Claudio G. Brunstein, Jeffrey S. Miller, Qing Cao, Daivd H. McKenna, Keii L. Hippen, Julie Curtsinger, Todd Defer, Bruce L. Levine, Carl H. June, Pablo Rubinstein, Philip B. McGIave, Bruce R. Blazar, and John E. Wagner. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood, 117(3): 1061-1070, 2010.
Lang, Julie, et al. “Generation of hematopoietic humanized mice in the newborn BALB/c-Rag2nullll2rYnull mouse model: a multivariable optimization approach.” Clinical Immunology 140.1 (2011): 102-116.
Kim, Do-Hyung, et al. “mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery.” Cell 110.2 (2002): 163-175.
Kishore M, Cheung KCP, Fu H, Bonacina F, Wang G, Coe D, Ward EJ, Colamatteo A, Jangani M, Baragetti A, Matarese G, Smith DM, Haas R, Mauro C, Wraith DC, Okkenhaug K, Catapano AL, De Rosa V, Norata GD, Marelli-Berg FM. Regulatory T Cell Migration Is Dependent on Glucokinase-Mediated Glycolysis. Immunity. 2017;47(5):875-889 e10.
Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz C, Floess S, Fritz V, Matias Ml, Yong C, Surh N, Marie JC, Huehn J, Zimmermann V, Kinet S, Dardalhon V, Taylor N. Glutamine-dependent alpha-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci Signal. 2015;8(396):ra97.
Lampugnani MG, Caveda L, Breviario F, Del Maschio A, Dejana E. Endothelial cell-to-cell junctions. Structural characteristics and functional role in the regulation of vascular permeability and leukocyte extravasation. Bailliere's clinical haematology. 1993;6:539-558.
Lataillade, Jean-Jacques, et al. “Chemokine SDF-1 enhances circulating CD34+ cell proliferation in synergy with cytokines: possible role in progenitor survival.” Blood, The Journal of the American Society of Hematology 95.3 (2000): 756-768.
Lindstein, Tullia, et al. “Regulation of lymphokine messenger RNA stability by a surface-mediated T cell activation pathway.” Science 244.4902 (1989): 339-343.
Liotta, Francesco, et al. “Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma.” BJU international 107.9 (2011): 1500-1506.
Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone Ja. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006;203(7):1701-1711.
Mathew, James M., et al. “A phase I clinical trial with ex vivo expanded recipient regulatory T cells in living donor kidney transplants.” Scientific reports 8.1 (2018): 1-12.
Ueda, Ryosuke, et al. “Interaction of natural killer cells with neutrophils exerts a significant antitumor immunity in hematopoietic stem cell transplantation recipients.” Cancer medicine 5.1 (2015): 49-60.
Jin, H., and J. Bae. “Neuropeptide Y regulates the hematopoietic stem cell microenvironment and prevents nerve injury in the bone marrow.” 22nd Annual ISCT Meeting (2016): S29.
Bai, Tao, et al. “Expansion of primitive human hematopoietic stem cells by culture in a zwitterionic hydrogel.” Nature medicine 25.10 (2019): 1566-1575.
Horwitz, Mitchell E., et al. “Phase l/ll study of stem-cell transplantation using a single cord blood unit expanded ex vivo with nicotinamide.” Journal of Clinical Oncology 37.5 (2019): 367-373.
Lee III, Daniel W., et al. “Long-term outcomes following CD19 Car T cell therapy for B-ALL are superior in patients receiving a fludarabine/cyclophosphamide preparative regimen and post-CAR hematopoietic stem cell transplantation.” Blood 128.22 (2016): 218.
Goh, Celeste, Sowmya Narayanan, and Young S. Hahn. “Myeloid-derived suppressor cells: the dark knight or the joker in viral infections?.” Immunological reviews 255.1 (2013): 210-221.
Pati, Shibani, and Todd E. Rasmussen. “Cellular therapies in trauma and critical care medicine: Looking towards the future.” PLoS Medicine 14.7 (2017): e1002343.
Pati, Shibani, et al. “Lyophilized plasma attenuates vascular permeability, inflammation and lung injury in hemorrhagic shock.” PloS one 13.2 (2018): e0192363.
Related Publications (1)
Number Date Country
20180291342 A1 Oct 2018 US
Provisional Applications (5)
Number Date Country
62479721 Mar 2017 US
62479760 Mar 2017 US
62479788 Mar 2017 US
62549871 Aug 2017 US
62647361 Mar 2018 US