Passive replacement of media

Information

  • Patent Grant
  • 11795432
  • Patent Number
    11,795,432
  • Date Filed
    Wednesday, December 20, 2017
    6 years ago
  • Date Issued
    Tuesday, October 24, 2023
    7 months ago
Abstract
Embodiments described herein generally relate to passively replacing media in a closed cell expansion system to reduce or prevent the dilution of chemical signaling used to inhibit signaling pathways that keep a cell population in the lag phase of cell growth. To prevent such dilution, active inlet fluid flow to the system may be halted. To replace fluid lost by the system, a bag containing media may be attached to the waste line in replacement of the waste or outlet bag connected thereto. By turning off one or more pumps, media from the replacement bag is added to the system at the rate of evaporation. Chemical signaling dilution may be prevented while conserving system resources. Enhancement of chemical signaling to reduce the lag phase of cell growth may further be accomplished by adding molecules, such as chemical-signaling proteins, from a direct source to the system.
Description
BACKGROUND

Cell Expansion Systems (CESs) are used to expand and differentiate cells. Cell expansion systems may be used to expand, e.g., grow, stem cells, such as mesenchymal stem cells, human mesenchymal stem cells, etc. Cell expansion systems may also expand other types of cells, such as bone marrow cells, for example. Stem cells which are expanded from donor cells may be used to repair or replace damaged or defective tissues and have broad clinical applications for a wide range of diseases. 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 using the passive replacement of media in a cell expansion system to conserve media and provide an environment conducive to encouraging cell growth. The expansion of cells, such as human mesenchymal stem cells, for example, uses external chemical signaling between the cells to initiate cell expansion by inhibiting lag phase signaling pathways internal to the cells. The expansion of other types of cells, such as Chinese hamster ovary (CHO) cells, for example, may be particularly sensitive to chemical signaling between the cells, according to embodiments. For example, CHO cells secrete cholecystokinin (CCK), a regulatory hormone responsible in part for cell culture maintenance and proliferation via chemical signaling. In embodiments, CCK may be small enough to pass through the microporous membrane of a hollow fiber bioreactor. Due to such ability to pass through the membrane, dilution of chemical signaling may occur regardless of inlet media addition to the intracapillary or extracapillary loop of a cell expansion system. To reduce or prevent the dilution of external chemical signaling in a closed, automated cell expansion system and, thus, reduce the lag phase of the cells, aspects of particular embodiments provide for passively replacing media by interrupting protocol procedures being executed and replacing a waste or outlet bag(s) used with the cell expansion system with a media bag(s). In embodiments, a bag containing base media may be attached to a waste line of the cell expansion system, in which such configuration allows base media to be added to the system at the rate of evaporation during conditions of no active inlet fluid flow. In embodiments, other types of replacement fluids are used in the media bag(s), such as, for example, complete media or cytokines or other cell-signaling protein molecules. In other embodiments, fluid may be passively replaced by interrupting protocol procedures being executed and allowing any fluid in the waste or outlet bag (assuming no constituents toxic to cell growth are present in the waste or outlet bag) to be passively added to the system at the rate of evaporation during conditions of no active inlet fluid flow. The passive addition of fluid avoids adding an excess amount of fluid, in which an excess amount of fluid may dilute the chemical signaling used to initiate cell expansion. Further, media constituents themselves may ultimately be conserved, resulting in increased system efficiencies and a savings of resources.


Embodiments of the present disclosure further relate to enhancing chemical signaling by adding a molecule(s), e.g. cell-signaling protein molecules, such as cytokines, according to embodiments, to the expanding cell population in a bioreactor. In an embodiment, cytokines, or other type of cell-signaling protein molecules, may be added to the bioreactor by, for example, welding a tubing line or other material connected to a cytokine source, or pre-filled with cytokines or other desired constituents, to a sampling coil or sample coil of the cell expansion system. The cytokines may thus be added to the bioreactor at the sample coil. Such direct addition results in a significant savings of cytokines, which may be costly, because a much higher amount of cytokines would need to be added to a media bag to compensate for dilution of the cytokines by the media than are needed when only the cytokine source itself replenishes the bioreactor. Further, cytokines tend to degrade quickly over time or with exposure to ultra-violet (UV) light, in which such degradation may be minimized by adding cytokines closer to the expanding cell population, e.g., at the sample coil of the bioreactor itself which is isolated from UV light sources. In such embodiments, the cytokines in the bioreactor may thus be maintained at a certain level while conserving resources.


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” may mean 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.



FIG. 1 depicts a perspective view of a hollow fiber bioreactor, in accordance with 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. 5 depicts a schematic of a cell expansion system, in accordance with an embodiment of the present disclosure.



FIG. 6 illustrates a schematic of another embodiment of a cell expansion system.



FIG. 7 depicts the cell expansion system embodiment of FIG. 6 with a waste bag replaced by a media bag, in accordance with embodiments of the present disclosure.



FIG. 8 illustrates the cell expansion system embodiment of FIG. 5 with a waste bag replaced by a media bag, in accordance with embodiments of the present disclosure.



FIG. 9 depicts the cell expansion system embodiment of FIG. 6 with a molecule source included as part of the premounted fluid conveyance device, in accordance with embodiments of the present disclosure.



FIG. 10 illustrates a flow diagram depicting the operational characteristics of a process for passively replacing media in a cell expansion system, in accordance with embodiments of the present disclosure.



FIG. 11 depicts a flow diagram illustrating the operational characteristics of a process for adding a molecule from a molecule source implemented as part of the cell expansion system itself, in accordance with embodiments of the present disclosure.



FIG. 12 illustrates a flow diagram depicting the operational characteristics of another embodiment of a process for passively replacing media in a cell expansion system.



FIG. 13 depicts an example processing system of a cell expansion system upon which embodiments of the present disclosure may be implemented.





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.


Embodiments of the present disclosure are generally directed to systems and methods for passively replacing media in a cell expansion system. Passive replacement of media may be accomplished by interrupting one or more protocol procedures being executed with respect to the system, e.g., cell loading, cell feeding, etc., and replacing a waste or outlet bag(s) used with the system with a media bag(s). By interrupting, or stopping, mechanisms of the cell expansion system from operating according to the protocol being executed, active inlet fluid flow to the system may be halted to reduce or prevent the dilution of chemical signaling used to inhibit the internal signaling pathways that keep a cell population in the lag phase in a bioreactor of the closed system. Reducing or preventing such dilution may thus reduce the lag phase of cell growth. More efficient and increased cell expansion may therefore occur, in which a greater number of cells may be expanded in a shorter amount of time, according to embodiments of the present disclosure.


Dilution of chemical signaling may occur where an inlet fluid flow into a cell expansion system overcompensates for the evaporation of fluid from the system. For example, an oxygenator or gas transfer module may be used in a closed cell expansion system to maintain the media in fibers in the bioreactor with a desired gas concentration, e.g., 5% CO2, 20% O2, 75% N2. As an example, evaporation in the gas transfer module may occur at 14 mL/day. Without any inlet flow, such evaporation could result in either a build-up of air in the system or a back-flow of fluid from the waste or outlet line in embodiments where the waste line is the only source of fluid for the system which is not occluded by a pump, for example. Using an inlet flow, however, to account for such evaporation may result in overcompensating for the actual amount of fluid lost due to evaporation. For example, in an embodiment, the inlet flow rate into the cell expansion system may have a minimum flow rate. As an example, the inlet flow rate may be set at a minimum rate of 0.1 mL/min or 144 mL/day. Where evaporation in the gas transfer module occurs at 14 mL/day, the fluid lost due to evaporation may be overcompensated for by a rate of 130 mL/day in such embodiment. Such excess 130 mL/day dilutes chemical signaling for initiating cell expansion. For example, such dilution may occur in embodiments where chemical signaling molecules are able to cross, or pass through, a hollow fiber membrane from an intracapillary to an extracapillary side. As a result, adding replacement fluid to either the intracapillary or extracapillary side may result in dilution of the chemical signaling molecules by preventing or reducing them from building up by continuously adding fluid into the system. Where such dilution occurs, communication between the chemical signaling cells may be significantly impacted such that the cells may be unable to expand or even survive. Such dilution may have a particularly significant impact with respect to some cell types as compared to others. For example, reducing or preventing the dilution of chemical signaling molecules may have a significant impact on the expansion of Chinese hamster ovary (CHO) cells, according to embodiments.


In embodiments, instead of using an active inlet fluid flow which may unduly overcompensate for the evaporation of fluid from the system, the active inlet fluid flow to the system may be halted to prevent or minimize the dilution of chemical signaling used to inhibit the signaling pathways that maintain the cell population in a bioreactor in the lag phase. Such active inlet fluid flow may be halted, for example, by interrupting, or stopping, system mechanisms from operating according to the protocol(s) being executed. Instead of using an overcompensating active inlet fluid flow, such active inlet fluid flow may therefore be stopped while using a passive replacement of media and, therefore, not result in a build-up of air or back-flow of waste. To accomplish such passive media replacement, fluid, e.g., base media, may be added to the system at a rate equal to the rate of evaporation from the system, e.g., such as the rate of evaporation from a gas transfer module, through the use of one or more media bags used to replace one or more waste or outlet bags normally used with the system. The active inlet fluid flow may therefore be stopped while media from the replacement, or substitute, media bag replaces any fluid lost from the system due to evaporation. Such passive addition of fluid avoids adding an excess amount of fluid, in which an excess amount of fluid may dilute the chemical signaling used to initiate cell expansion. As a result, lost fluid may be replaced by adding media at about the rate of evaporation and without diluting chemical signaling used to inhibit signaling pathways that keep the cell population in the lag phase. The lag phase of cell growth may therefore be significantly reduced. Further, media constituents themselves may ultimately be conserved, resulting in increased system efficiencies and a savings of resources.


In other embodiments, fluid may be passively replaced by interrupting protocol procedures being executed and allowing any fluid in the waste or outlet bag (assuming no constituents toxic to cell growth are present in the waste or outlet bag) to be passively added to the system at the rate of evaporation during conditions of no active inlet fluid flow.


The dilution of chemical signaling may be particularly costly where the cell media includes expensive additives. For example, cell-signaling proteins, e.g., cytokines, may be used in the bioreactor to spur cell growth. Diluting cytokines may thus result in significant costs. Accordingly, saving the excess media, e.g., 130 mL/day, may provide significant cost savings over other cell expansion processes. Instead of using an overcompensating active inlet fluid flow, the passive replacement of media may thus be used, according to embodiments of the present disclosure, to maintain media constituent concentrations and conserve media in general. Further, in embodiments, other types of replacement fluids are used in the media bag(s), such as, for example, a media bag comprising cytokines or other cell-signaling protein molecules.


In embodiments, molecules, such as cell-signaling protein molecules, may be added to the bioreactor from a source of such molecules. For example, tubing or other material connected to a molecule source, such as a cytokine source, may be sterile-welded to a sample coil in the cell expansion system, and cytokines in the bioreactor may be replenished by such direct source of cytokines. In an embodiment, such tubing or other material comprises an additional volume added to the sampling coil. In another embodiment, such tubing or other material comprises a segment of tubing or other material used to replace a corresponding segment, or portion, of the sampling coil. In embodiments, such tubing or other material may be pre-filled with the desired constituents, e.g., cytokines. In another embodiment, such tubing or material may be connected to a container or bag comprising such desired constituents. A source of cytokines conserves the amount of cytokines used because the cytokines are not added to an IC media bag, for example, which could dilute the cytokines and use a larger amount of cytokines to achieve the same replenishment concentrations. Further, the cytokines may be added closer to the expanding cell population to minimize degradation of the cytokines. Degradation of the cytokines increases with exposure time to the media bags and UV light where they may be stored. Where cytokines are added closer to the expanding cell population, such degradation may be reduced because the cytokines reach the expanding cell population in a shorter amount of time in an environment protected from any sources of UV light. Such cytokines may be passively or actively added to the bioreactor, according to embodiments, to enhance chemical signaling capabilities. For example, such passive addition of cytokines may occur where the cytokines are added to the system from a media bag used to replace a waste bag, according to an embodiment, at the rate of evaporation during conditions of no active inlet fluid flow.


In an embodiment, chemical signaling may thus be controlled by the addition of cytokines at the sample coil. In another embodiment, chemical signaling may be controlled through such addition of cytokines at the sample coil coupled with the replacement of a waste bag(s) with a media bag(s). By replacing a waste bag(s) with a media bag(s), dilution in the bioreactor may be significantly reduced, as discussed above. Such dilution may be particularly costly where cytokines are used in the cell population expansion in the bioreactor. Preventing or reducing such dilution through the use of the media bag replacement thus may result in significant savings, according to embodiments.


In embodiments, a method provides for controlling chemical signaling in a bioreactor of a closed cell expansion system that includes a disposable tubing set(s). In such embodiments, the method may include the steps of coating the bioreactor and loading cells from a cell inlet bag into the bioreactor. For example, steps for loading cells with circulating distribution may be performed, according to an embodiment. In another embodiment, steps involving the loading of cells with uniform suspension, for example, may be performed. The cells may then be distributed across a membrane of the bioreactor by activating an intracapillary circulation pump, for example. In embodiments, after the loading and the distributing, a waste bag attached to the cell expansion system may be replaced with a media bag. After the waste bag is replaced, one or more pumps, e.g., an intracapillary circulation pump, extracapillary inlet pump, and intracapillary inlet pump, may be turned “OFF” or otherwise deactivated, according to an embodiment. In another embodiment, one or more pumps may be turned “OFF” or otherwise deactivated before replacing the outlet or waste bag. For example, in an embodiment expanding adherent cells, the intracapillary circulation pump may be deactivated after replacing a waste or outlet bag with a media bag. In another embodiment expanding adherent cells, for example, the intracapillary circulation pump may be deactivated before replacing the waste or outlet bag with a media bag. In yet other embodiments expanding non-adherent cells, for example, the intracapillary circulation pump may stay activated while one or more other pumps are deactivated.


In at least one embodiment, the media from the media bag flows through an extracapillary waste valve to the extracapillary circulation loop to replenish fluid evaporated from a gas transfer module in the extracapillary circulation loop. In embodiments, after replacement of the waste bag with the media bag, the method further includes deactivating an intracapillary inlet pump, deactivating an extracapillary inlet pump, maintaining an extracapillary circulation pump in an activated state, and maintaining the extracapillary waste valve in an open position.


In some embodiments, the cells include adherent cells, and the method may include the additional steps of enabling the adherent cells to attach to the bioreactor membrane and maintaining flow on an extracapillary circulation loop by maintaining an extracapillary circulation pump in an activated state. In some embodiments, the adherent cells are allowed to attach to the bioreactor membrane for a period of time, e.g., a first period of time, of about eighteen (18) hours to about twenty-four (24) hours. In other embodiments, the cells include non-adherent or suspension cells, such as, for example, CHO cells.


The method, in some embodiments, may further include feeding the cells in the bioreactor of the closed cell expansion system while maintaining the media bag in replacement of the waste bag and while reducing an intracapillary inlet rate. In these embodiments, feeding may include activating the intracapillary circulation pump. In embodiments, the feeding of the cells may be stopped after a second period of time of about forty-five (45) hours to about fifty (50) hours of feeding. In yet other embodiments, the feeding may be stopped after a second period of time of about forty-eight (48) hours of feeding.


The method, in embodiments, further involves measuring a concentration of lactate generated from the cells and stopping the feeding of the cells when the concentration of lactate is equal to or greater than about 6 mmol/L. In some embodiments, the method includes removing the media bag, inserting the waste bag, activating the intracapillary inlet pump, activating the intracapillary circulation pump, and maintaining an extracapillary circulation pump in an activated state. The intracapillary inlet pump may operate at an intracapillary inlet rate of about 0.1 mL/min, in some embodiments. The intracapillary circulation pump may operate at an intracapillary circulation rate of about 20 mL/min, in some embodiments. The extracapillary circulation pump may operate at an extracapillary circulation rate of about 30 mL/min, according to embodiments. In an embodiment, the method may additionally involve doubling, or otherwise increasing, according to other embodiments, the intracapillary inlet rate until a desired number of the cells are available for harvest. When the desired number of cells is available for harvest, embodiments include the additional steps of: releasing the cells from the membrane of the bioreactor, suspending the cells in the intracapillary circulation loop, and transferring the cells in suspension to a harvest bag.


Steps performed, including, for example, coating the bioreactor, loading cells, and distributing the cells, may be performed automatically in some embodiments, such as by a processor executing pre-programmed tasks stored in memory. Replacing a waste bag with a media bag may be performed manually in some embodiments and automatically in others. The automatic replacement of the waste bag may include, in embodiments, receiving, by a processor, a command to execute a task for replacing the waste bag, the task being stored in a memory. In an embodiment, for example, upon receiving such a command for the bag replacement, a processor may send a signal to close a valve(s), for example, for the waste bag and open another valve(s) for an attached media bag. In another embodiment, a single valve, or other type of mechanism, may control the flow of fluid from the waste bag or attached media bag.


The media bag may store base media and, in some embodiments, stores about 500 mL of base media, for example. The base media may include a number of different components, including, for example, glucose to provide an energy source for cells to grow, according to an embodiment. The media bag may comprise other fluids and/or constituents in accordance with embodiments of the present disclosure.


Other embodiments of the method provide for additional steps, some of which include loading cell-signaling protein molecules into a sample coil of an intracapillary circulation loop and activating the intracapillary circulation pump to transfer the cell-signaling protein molecules to the bioreactor. In some embodiments, the sample coil and the intracapillary circulation loop are part of a disposable tubing set.


In embodiments, the method may further include, prior to loading cells into the bioreactor, replacing fluid on an intracapillary circulation loop and on an extracapillary circulation loop with media from an intracapillary media bag, and allowing the media from the intracapillary media bag to reach equilibrium with a gas supply.


Some 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 type, may be grown in a bioreactor in the cell expansion system. According to embodiments, the cell expansion system may include base 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 may be 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 further includes 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, wherein the first fluid flow path comprises an intracapillary portion of the hollow fiber membrane. The system may further include a fluid inlet path fluidly associated with the first fluid flow path, wherein the plurality of cells are introduced into the first fluid flow path through a first fluid inlet path. A first pump for circulating fluid in the first fluid flow path of the bioreactor may also be included. In embodiments, the system includes a controller, e.g., first controller, for controlling operation of the first pump. In an embodiment, the controller may be a computing system, including a processor, for example. The controller may be configured, in embodiments, to control the pump to circulate a fluid at a first rate within the first fluid flow path, and, when a waste bag in the cell expansion system is replaced with a media bag, the controller stops the circulation of the fluid within the first fluid flow path after the plurality of the cells are loaded into the bioreactor. In some embodiments, a second pump for transferring intracapillary inlet fluid from an intracapillary media bag to the first fluid flow path and a controller, e.g., second controller, for controlling operation of the second pump are included. The second controller in embodiments controls the second pump to transfer cells from a cell inlet bag to the first fluid flow path, and when a waste bag in the cell expansion system is replaced with a media bag, stop the transfer of the cells from the cell inlet bag after the plurality of the cells are loaded into the bioreactor. Additional controllers, e.g., third controller, fourth controller, fifth controller, sixth controller, etc., may be used in accordance with embodiments. Further, additional pumps, e.g., third pump, fourth pump, fifth pump, sixth pump, etc., may be used in accordance with embodiments of the present disclosure. In addition, while the present disclosure may refer to a media bag, a waste bag, a cell inlet bag, etc., multiple bags, e.g., a first media bag, a second media bag, a third media bag, a first waste bag, a second waste bag, a third waste bag, a first cell inlet bag, a second cell inlet bag, a third cell inlet bag, etc., or other types of containers, may be used in embodiments. In other embodiments, a single media bag, a single waste bag, a single cell inlet bag, etc., may be used.


In embodiments, the system may be 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 coat the bioreactor, for example. In response to the instruction to coat the bioreactor, the processor may execute a series of steps to coat the bioreactor and may next receive an instruction to load cells into the bioreactor, for example. In response to the instruction to load cells, the processor may execute a series of steps to load the cells from a cell inlet bag into the bioreactor. After loading the cells into the bioreactor, the processor may receive an instruction to stop an intracapillary inlet pump, an intracapillary circulation pump, and an extracapillary inlet pump, for example. The cell expansion system may be operated to allow media to flow from a media bag through an extracapillary waste valve, wherein the extracapillary waste valve is in an open position. The processor may receive an instruction to pump the media in the extracapillary circulation loop to replace fluid evaporated from a gas transfer module located in the extracapillary circulation loop.


Referring to FIG. 1, 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, 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, are typically too large to pass through the hollow fiber membrane, and 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 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 order for the cells to adhere to the surface of the hollow fibers, the surface may be modified in some way, either by coating at least the cell growth surface with a protein such as fibronectin or collagen, or by exposing the surface to radiation. Gamma treating the membrane surface allows for attachment of adherent cells without additionally coating the membrane with fibronectin or the like. Bioreactors made of gamma treated membranes may be reused. Other coatings and/or treatments for cell attachment may be used in accordance with embodiments of the present disclosure.


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 may be 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 includes a bioreactor 100 and an oxygenator or gas transfer module 212. 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 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.


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 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. 5-9, as discussed below. Pump loops 406A and 406B are also 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 the media bags, according to embodiments.



FIG. 5 illustrates a schematic of an embodiment 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. 5 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.



FIG. 5 illustrates a CES 500, which 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 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. Media entering the IC loop 502 may enter through valve 514. As those skilled in the art will appreciate, additional valves 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. This will be described in more detail below.


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. 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. 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 the configuration depicted for CES 500, 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). The CES 500 may also be configured to flow in a counter-current conformation.


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 (from bag 568) or wash solution (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 valve distribution 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 at a predetermined temperature.


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 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. 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 valve distribution 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 at a predetermined temperature. It is further noted that, in embodiments, components of CES 600 and CES 500 (FIG. 5) may be combined. In other embodiments, a CES may include fewer or additional components than those shown in FIGS. 5 and 6 and still be within the scope of the present disclosure.


While FIGS. 5 and 6 illustrate schematics of different embodiments of cell expansion systems, FIGS. 7 and 8 depict these same cell expansion systems with the waste or outlet bags (586 and 686) replaced by media bags in accordance with embodiments of the present disclosure. For example, as depicted in FIG. 7, waste or outlet bag 686 in CES 600 (FIG. 6) has been replaced by media, e.g., base media, bag 700. Further, one or more pumps, e.g., IC Circulation Pump 612, EC Inlet Pump 678, and IC Inlet Pump 654, have been turned “OFF,” according to an embodiment. There is thus no active inlet fluid flow into cell growth chamber 601. To compensate for fluid lost due to evaporation at the oxygenator or gas transfer module 632, the EC Circulation Pump 628 is left “ON” and the EC Waste Valve 692 is left “OPEN.” This configuration allows fluid from media bag 700 to backflow into the CES 600 system at a rate equal to the rate of evaporation from the oxygenator or gas transfer module 632. The fluid lost in the system due to evaporation may thus be replaced without diluting chemical signaling occurring in the bioreactor 601 during cell growth therein. In embodiments, the lag phase of cell growth in the bioreactor 601 may therefore be significantly reduced, and more efficient cell expansion may occur. Further, by turning “OFF,” or otherwise deactivating, the inlet pump(s), system resources may be conserved because there is no active inlet fluid being unnecessarily introduced into the system. While FIG. 7 shows an embodiment in which the IC Circulation Pump 612, EC Inlet Pump 678, and IC Inlet Pump 654 have been turned “OFF,” other embodiments provide for one or more of such pumps, e.g., the IC Circulation Pump 612, for example, to remain “ON” or activated (not shown in FIG. 7). For example, it may be desired in embodiments to continue circulation in the intracapillary side depending on the type of cells, e.g., non-adherent cells, being expanded, according to an embodiment.


In some embodiments, the media bag (e.g., 700) may be positioned at a physically higher level than at least a portion of the EC loop 604 to allow gravity to assist in draining fluid from the media bag into the EC loop 604. In some embodiments, the waste bag 686 (FIG. 6) may be positioned lower than the EC loop 604 to allow gravity to assist in draining waste media into the waste bag 686. According to embodiments, when the media bag 700 replaces the waste bag 686, the substitute or replacement media bag 700 may be positioned physically higher than the original position of the waste bag 686.


Turning to FIG. 8, a similar configuration is shown, in which, for example, waste bag 586 has been replaced by media, e.g., base media, bag 800. Further, one or more pumps, e.g., IC Circulation Pump 512, EC Inlet Pump 578, and IC Inlet Pump 554, have been turned “OFF,” according to an embodiment. There is thus no active inlet fluid flow into the bioreactor 501. To compensate for fluid lost due to evaporation at the gas transfer module or oxygenator 532, the EC Circulation Pump 528 may be left “ON,” and the EC Waste Valve 582 may be left “OPEN.” In embodiments, such configuration allows fluid from the substitute or replacement media bag 800 to backflow into the system at a rate equal to the rate of evaporation from the gas transfer module or oxygenator 532. The fluid lost in the system due to evaporation may thus be replaced without diluting chemical signaling occurring in the bioreactor 501 during cell growth therein. In embodiments, the lag phase of cell growth in the bioreactor 501 may therefore be significantly reduced, and more efficient cell expansion may occur. Further, by turning “OFF” the one or more inlet pumps, system resources may be conserved because there is no active inlet fluid being unnecessarily introduced into the system. While FIG. 8 shows an embodiment in which the IC Circulation Pump 512, EC Inlet Pump 578, and IC Inlet Pump 554 have been turned “OFF,” other embodiments provide for one or more of such pumps, such as the IC Circulation Pump 512, for example, to remain “ON” or activated (not shown in FIG. 8). For example, it may be desired in embodiments to continue circulation in the intracapillary side depending on the type of cells, e.g., non-adherent cells, being expanded, according to an embodiment.


In some embodiments, when the waste bag 586 is replaced by the media bag 800, the substitute or replacement media bag may be positioned physically higher than the original position of the waste bag 586 to allow gravity to assist in draining media into the EC loop 504.


The replacement of the waste bag with a media bag allows passive replacement of fluid lost due to evaporation. Such passive replacement of fluid may provide a significant conservation of fluid in cell expansion processes. In processes involving active media replacement, media may be added and circulated in the IC loop during attachment of cells to replace fluid lost due to evaporation. As described above, if media is added at 0.1 ml/min, which may occur in some processes, according to embodiments, this may result in an excess amount (over the amount that has evaporated) of fluid of up to 130 mL/day in the system, for example. Passive addition of fluid with the replacement of the waste bag with a media bag avoids the addition of an excess amount. As can be appreciated, the media may include expensive additives. Saving about 130 mL/day, for example, may provide significant cost savings over other cell expansion processes.


While FIGS. 7 and 8 allow for the passive replacement of media in a closed cell expansion system through the use of a media bag in replacement of a waste bag, FIG. 9 illustrates an embodiment in which a molecule source, e.g., a cell signaling protein molecule source, may be added to a cell expansion system, such as CES 600 (FIG. 6) (or CES 500 (FIG. 5)), for example. In one embodiment, the molecule source 900 may be a cytokine source welded into the sample coil or sampling coil 618, in which such cytokine source comprises a piece of tubing or other material welded into the sampling coil 618. Through such a source, i.e., direct source, cytokines may be added to the IC loop 602 without diluting such proteins, in which such dilution may occur where the cytokines are added instead at an IC Media bag, for example. In embodiments, the molecules are directly added to the IC loop 602. Such direct addition may also occur at a sample port, for example, according to an embodiment. Cytokines in the cell growth chamber 601 may thus be passively or actively replenished by such cytokine source. In such embodiment, the IC Circulation Pump 612 is turned to the “ON” position to allow the cytokines entering the IC loop 602 at the sampling coil 618 to be pumped to the expanding cell population in the bioreactor 601. Such cell source may ultimately save significant resources where chemical-signaling proteins used in the bioreactor are particularly costly, e.g., cytokines.


While various example embodiments of a cell expansion system and methods for passively replacing media in conjunction therewith have been described, FIG. 10 illustrates example operational steps 1000 for passively replacing fluid to control chemical signaling in a closed cell expansion system, in accordance with embodiments of the present disclosure. START operation 1002 is initiated, and process 1000 proceeds to load the disposable tubing set 1004 onto the cell expansion system. Next, the system is primed 1006, such as by having a user or operator instruct the system to prime by selecting a task for priming, for example. In another embodiment, the system is primed 1006 automatically without any selection of a task or instruction from a user or operator. After priming the set, process 1000 proceeds to coat the bioreactor 1008, in which the bioreactor is coated with a reagent. For example, a reagent is loaded into the IC loop until the Reagent Bag is empty. The reagent is chased from the air removal chamber into the IC loop, and the reagent is then circulated in the IC loop. Once the bioreactor is coated, the IC/EC Washout task is executed 1010, 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, according to an embodiment. Next, to maintain the proper or desired gas concentration across fibers in the bioreactor membrane, the condition media task 1012 is 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. In an embodiment, the system is then maintained in a proper state until a user or operator is ready to load cells into the bioreactor. In other embodiments, a user or operator may not be needed to perform the noted steps/operations; rather, the steps/operations may be performed automatically by the cell expansion system.


Process 1000 next proceeds to loading cells into the bioreactor from a cell inlet bag with circulating distribution 1014. In an embodiment, cells are loaded into the bioreactor from the cell inlet bag until the bag is empty. Cells are then chased from the air removal chamber to the bioreactor. Larger chase volumes spread the cells and move the cells toward the IC outlet. The distribution of cells is promoted across the membrane via IC circulation, such as through the IC circulation pump, with no IC inlet, for example.


After completion of the load cells with circulating distribution task 1014, the waste or outlet bag is replaced with a media bag 1016. In an embodiment, the media bag comprises about 500 mL of base media. The media bag may comprise other fluids and/or constituents, according to embodiments. In embodiments, the replacement of the outlet or waste bag with a media bag 1016 may be optional, in which fluid may be passively replaced by interrupting protocol procedures being executed and allowing any fluid in the outlet or waste bag (assuming no constituents toxic to cell growth are present in the outlet or waste bag) to be passively added to the system at the rate of evaporation during conditions of no active inlet fluid flow. Such passive addition of fluid avoids adding an excess amount of fluid and, thus, avoids diluting chemical signaling molecules.


Returning to FIG. 10, one or more pumps, e.g., the IC Inlet Pump, the IC Circulation Pump, and the EC Inlet Pump, may then be turned “OFF” or may otherwise be indicated to stop or deactivate 1018. Any adherent cells in the bioreactor are then allowed to attach to the bioreactor membrane 1020 for a period of time, such as for about eighteen (18) to about twenty-four (24) hours, according to an embodiment of the present disclosure. During this timeframe, flow continues on the EC circulation loop, in which the EC circulation rate is maintained at about 30 mL/min, according to an embodiment. A non-zero EC circulation rate helps to maintain the proper or desired gas concentration across the fibers of the bioreactor membrane by continuing to pump fluid in the EC loop through the gas transfer module or oxygenator. While the proper or desired gas concentration is maintained through the use of the gas transfer module, evaporation of fluid also occurs at the gas transfer module. By keeping the EC Waste Valve open, however, media from the media bag (replacing the waste bag) may back-flow into the system and be pumped by the EC Circulation Pump through the EC loop. The media may thus replace fluid lost due to evaporation from the gas transfer module at the rate of evaporation. Thus, membrane fibers in the bioreactor will not be diluted with excess fluid, and the transition of cell growth out of the lag phase will not be inhibited.


After the attaching of any adherent cells for about eighteen (18) to about twenty-four (24) hours, according to an embodiment, a continued cell attachment phase 1022 continues for up to about forty-eight (48) hours. During operation 1022, the IC circulation pump may be activated or turned “ON” to provide even the furthest fibers of the bioreactor membrane with media. For example, the IC circulation pump may be activated to adjust the IC circulation rate to about 20 mL/min, according to an embodiment of the present disclosure. However, during this period of modified feeding through activation of the IC circulation pump 1022, the IC inlet rate remains at 0 mL/min. Rather, the substitute media bag (in replacement of the waste bag) continues to provide any necessary fluid replacement to the system while not diluting the membranes or inhibiting chemical signaling. Operation 1022 with modified feeding of the cells thus allows for cell attachment to continue without disruption of chemical signaling occurring in the bioreactor. This continued cell attachment phase continues, according to embodiments, for up to about forty-eight (48) additional hours and/or, in embodiments, until the lactate generation of the cells is greater than or equal to about 6 mmol/L. In an embodiment, the concentration of lactate is measured. In another embodiment, the lactate generation rate, for example, is measured. In an embodiment, the lactate generation is thus checked at operation 1024 to determine if the concentration of lactate is equal to or exceeds 6 mmol/L. In other embodiments, the lactate generation is checked at operation 1024 to determine the concentration of lactate in relation to another predetermined amount.


Process 1000 next proceeds to query 1026, in which it is determined whether more than forty-eight hours has passed since the IC circulation pump was activated or whether the concentration of lactate is equal to or greater than about 6 mmol/L. If less than forty-eight (48) hours has passed or if the concentration of lactate is not equal to or in excess of about 6 mmol/L, process 1000 proceeds NO to check lactate generation operation 1024 and then to query 1026 again. It is noted that the present disclosure is not limited to determining whether forty-eight (48) hours have passed or whether there is a lactate concentration equal to or in excess of 6 mmol/L. In other embodiments, process 1000 may involve a different predetermined period of time. For example, at query 1026, a determination may be made whether about 12 hours, about 24 hours, about 36 hours, or about 40 hours have passed. In other embodiments, the predetermined period of time may be about 50 hours or about 60 hours. In embodiments, a determination may be made whether more than about 12 hours, more than about 24 hours, more than about 36 hours, or more than about 40 hours have passed. In other embodiments, a determination may be made whether less than about 60 hours or less than about 50 hours have passed. In yet other embodiments, process 1000 may involve determining whether the concentration of lactate is equal to or greater than another predetermined amount, such as about 3 mmol/L, about 4 mmol/L, about 5 mmol/L, about 7 mmol/L, or about 8 mmol/L. In embodiments, a determination may be made whether the concentration of lactate is more than about 3 mmol/L, more than about 4 mmol/L, or more than about 5 mmol/L. In other embodiments, a determination may be made whether the concentration of lactate is less than about 8 mmol/L or less than about 7 mmol/L.


If at query 1026 it is determined that more than about forty-eight (48) hours has passed since the IC circulation pump was activated or that the concentration of lactate is equal to or greater than 6 mmol/L, process 1000 proceeds YES to feed cells operation 1028, in which the IC inlet pump is activated or turned “ON” to maintain an IC Inlet Rate of 0.1 mL/min. Next, process 1000 proceeds to measure the glucose consumption 1030. In an embodiment, the concentration of glucose is measured. In another embodiment, the glucose consumption rate, for example, is measured. At query 1032, it is determined whether the measured glucose consumption is less than about 70 mg/L, in an embodiment. If the glucose consumption is less than about 70 mg/L (or another predetermined amount, according to other embodiments), process 1000 proceeds YES to double the IC Inlet Rate 1034. Process 1000 then proceeds to operation 1030 to continue measuring the glucose consumption of the cells and back to query 1032.


The present disclosure is not limited to determining whether the glucose consumption is less than about 70 mg/L. In other embodiments, process 1000 may involve a different predetermined amount. For example, in embodiments, process 1000 may involve determining whether the glucose consumption is less than another predetermined amount, such as about 65 mg/L, about 60 mg/L, or about 55 mg/L, for example. In other embodiments, the process 1000 may involve determining whether the glucose consumption is less than another predetermined amount, such as about 85 mg/L, about 80 mg/L, or about 75 mg/L, for example. In embodiments, a determination may be made whether the glucose consumption is more than about 55 mg/L, more than about 60 mg/L, or more than about 65 mg/L. In other embodiments, a determination may be made whether the glucose consumption is less than about 85 mg/L, less than about 80 mg/L, or less than about 75 mg/L.


If, at query 1032, the glucose consumption is determined to be greater than 70 mg/L, process 1000 proceeds NO to release the cells operation 1036, in which the cells are released from the membrane of the bioreactor and are suspended in the IC loop. In embodiments, an IC/EC Washout task in preparation for adding a reagent is performed. For example, IC/EC media may be replaced with a phosphate buffered saline (PBS) to remove protein, calcium (Ca2+), and magnesium (Mg2+) in preparation for adding trypsin, or another chemical-releasing agent, to release any adherent cells. A reagent may be loaded into the system until the reagent bag is empty. The reagent may be chased into the IC loop, and the reagent may be mixed within the IC loop. Following the release of any adherent cells, harvest operation 1038 transfers the cells in suspension from the IC circulation loop, including any cells remaining in the bioreactor, to the harvest bag. Process 1000 then terminates at END operation 1040.


Next, FIG. 11 depicts a flow diagram illustrating the operational characteristics of a process 1100 for adding a molecule from a molecule source, implemented as part of a cell expansion system itself, in accordance with embodiments of the present disclosure. While various example embodiments of a cell expansion system and methods for adding a molecule to a cell expansion system have been described, FIG. 11 illustrates example operational steps 1100 for adding a molecule that affects chemical signaling in a closed cell expansion system, in accordance with embodiments of the present disclosure. Some embodiments provide for the passive addition of a molecule from a molecule source. START operation 1102 is initiated, and process 1100 proceeds to load a disposable tubing set 1104 onto the cell expansion system. Next, the system is primed 1106, such as by having an operator or user provide an instruction to the system to prime by selecting a task for priming, for example. In another embodiment, the system is primed 1106 automatically without any selection of a task or instruction from an operator or user.


After priming the set, process 1100 proceeds to coat the bioreactor 1108, in which the bioreactor may be coated with a reagent. For example, in embodiments, a reagent is loaded into the IC loop until a reagent container is empty. The reagent may be chased from the air removal chamber into the IC loop, and the reagent may then be circulated in the IC loop. Once the bioreactor is coated, the IC/EC Washout task may be executed 1110, in which fluid on the IC circulation loop and on the EC circulation loop may be replaced, according to an embodiment. In an embodiment, 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 fibers in the bioreactor membrane, the condition media task 1112 is 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. In an embodiment, the system may then be maintained in a proper or desired state until an operator or user is ready to load cells into the bioreactor. In embodiments, such loading of cells is performed automatically.


Process 1100 next proceeds to loading cells into the bioreactor from a cell inlet bag with circulating distribution 1114. In an embodiment, cells are loaded into the bioreactor from a cell inlet bag until the bag is empty. Cells are then chased from the air removal chamber to the bioreactor. In embodiments that utilize larger chase volumes, cells are spread and move toward the IC outlet. The distribution of cells may be promoted across the membrane via IC circulation, such as through the IC circulation pump, with no IC inlet flow, for example.


After completion of the load cells with circulating distribution task 1114, the waste bag is replaced with a media bag 1116. In an embodiment, the media bag comprises about 500 mL of base media. In another embodiment, the media bag comprises any type of replacement fluid. In a further embodiment, step 1116 is optional, in which the outlet or waste bag stays connected and is not replaced with another bag. In yet a further embodiment, step 1116 is optional, in which the outlet or waste bag stays connected and desired constituents or other fluid(s) are added to the outlet or waste bag for passively adding such constituents/other fluid to the system.


In embodiments, one or more pumps, e.g., the IC Inlet Pump, the IC Circulation Pump, and the EC Inlet Pump, may then be turned “OFF” or may otherwise be indicated to stop or deactivate 1118. Any adherent cells in the bioreactor are then allowed to attach to the bioreactor membrane 1120 for a period of time, such as for about eighteen (18) to about twenty-four (24) hours, according to an embodiment of the present disclosure. During this timeframe, flow may continue on the EC circulation loop, in which the EC circulation rate may be maintained at about 30 mL/min, according to an embodiment. A non-zero EC circulation rate helps to maintain the proper or desired gas concentration across the fibers of the bioreactor membrane by continuing to pump fluid in the EC loop through the gas transfer module or oxygenator. While the proper or desired gas concentration is maintained through the use of the gas transfer module, evaporation of fluid also occurs at the gas transfer module. By keeping the EC Waste Valve open, however, media from the substitute media bag (replacing the waste bag) may back-flow into the system and be pumped by the EC Circulation Pump through the EC loop. The media may thus replace fluid lost due to evaporation from the gas transfer module at the rate of evaporation. Thus, membrane fibers in the bioreactor will not be diluted with excess fluid, and the transition of cell growth out of the lag phase will not be inhibited.


After the attaching of any adherent cells, an add molecule phase 1122 is performed. The molecule may be a protein molecule that is added to promote expansion of the cells. For example, the molecule may be a signaling molecule, such as one or more cytokines or growth factors that are involved in intercellular communications. The molecule may signal the cells to expand. In other embodiments, the molecule may not be directly involved in signaling but may help create an environment that is conducive to cell growth, in which examples of such molecules include carrier proteins, buffers, pH modifiers, etc. In embodiments, the molecule is added to the space where the cells are being grown, e.g., the IC or EC space. In embodiments, the molecules are added directly to the IC loop from a direct source of such molecules. Such direct addition may occur at a sampling coil or at a sample port, for example, according to embodiments. Cytokines, or other type of cell-signaling protein molecules, may be added to the bioreactor by, for example, welding a tubing line or other material connected to a cytokine source to a sampling coil or sample coil of the cell expansion system. The cytokines may thus be added to the bioreactor at the sample coil. Such direct addition results in a significant savings of cytokines, which may be costly, because a much higher amount of cytokines would need to be added to a media bag to compensate for dilution of the cytokines by the media than are needed when only the cytokine source itself replenishes the bioreactor, according to an embodiment. Further, cytokines tend to degrade quickly, in which such degradation may be minimized by adding cytokines closer to the expanding cell population, e.g., at the sample coil of the bioreactor itself. In such embodiments, the cytokines in the bioreactor may thus be maintained at a certain level while conserving resources. Through such a source, i.e., direct source, cytokines may be added to the IC loop without diluting such proteins, in which such dilution may occur where the cytokines are added instead at the IC Media bag, for example.


As noted above, the add molecule phase 1122 may be performed after the waste bag is replaced with a media bag 1116, according to an embodiment. In some embodiments, the molecule that is added at operation 1122 may be relatively expensive, and it is desirable to use the minimum amount required to promote growth of the cells. Performing operation 1116 first allows media from the media bag (replacing the waste bag) to back-flow into the system and be pumped by the EC Circulation Pump through the EC loop. According to an embodiment, only the media that is lost due to evaporation from the gas transfer module is replaced and at the rate of evaporation. Thus, the molecule may not be diluted with excess fluid. Accordingly, in an embodiment, only an amount of the molecule that may be effective at promoting growth may be added at operation 1122 since dilution by excess fluid may not be occurring.


After operation 1122, cells are grown at operation 1124. It is noted that, in embodiments, operation 1124 may involve a number of sub-operations. In some embodiments, the sub-operations include operations performed in process 1000 (FIG. 10). For example, in one embodiment, a circulating media operation may be performed to feed the cells. The IC circulation pump may be activated or turned “ON” to provide even the furthest fibers of the bioreactor membrane with media. The IC circulation pump may be activated to adjust the IC circulation rate to about 20 mL/min, according to an embodiment of the present disclosure. In some embodiments, even though the IC circulation pump is turned on, the IC inlet rate remains at 0 mL/min. Rather, the media bag (substitute media bag in replacement of the waste bag) continues to provide any necessary fluid replacement to the system while not diluting the molecule or otherwise inhibiting chemical signaling. In embodiments, operation 1124 allows cell attachment and cell growth to occur without disruption of chemical signaling by dilution of the molecules. This continued cell attachment and growth may continue, according to embodiments, for some predetermined period of time or may be based on a lactate generation of the cells, e.g., 6 mmol/L (in an embodiment). In these embodiments, additional sub-operations, such as determining lactate concentration(s) or that a predetermined period of time has elapsed, may be performed.


Operation 1124 may further involve a sub-operation of activating the IC inlet pump to maintain a predetermined IC inlet rate, e.g., 0.1 mL/min. This sub-operation may be triggered based on a predetermined period of time having elapsed or on a measurement, such as lactate concentration, for example.


In some embodiments, operation 1124 may involve a number of sub-operations to determine when to stop growing cells and begin releasing and harvesting cells. In one embodiment, this may include measuring a parameter, such as glucose consumption. In some embodiments, a predetermined glucose concentration, e.g., greater than 70 mg/L, may trigger subsequent operations, e.g., 1126 and 1128. In other embodiments, other parameters or the passage of a predetermined period of time may trigger subsequent operations.


At operation 1126, any adherent cells are released from the membrane of the bioreactor and are suspended, e.g., in the IC loop. In embodiments, an IC/EC washout task in preparation for adding a reagent to release the cells may be performed as part of operation 1126. For example, IC/EC media may be replaced with PBS to remove protein, calcium (Ca2+), and magnesium (Mg2+) in preparation for adding trypsin, or other chemical-releasing agent, to release any adherent cells. A reagent may be loaded into the system until the reagent bag is empty. The reagent may be chased into the IC loop, and the reagent may be mixed within the IC loop. Following the release of any adherent cells, harvest operation 1128 transfers the cells in suspension from the IC circulation loop, including any cells remaining in the bioreactor, to a harvest bag(s). Process 1100 then terminates at END operation 1130.


Turning to FIG. 12, example operational steps 1200 for passively replacing fluid to control chemical signaling in a closed cell expansion system are shown, in accordance with embodiments of the present disclosure. START operation 1202 is initiated, and process 1200 proceeds to load the disposable tubing set 1204 onto a cell expansion system. Next, the system is primed 1206, such as by having a user or operator instruct the system to prime by selecting a task for priming, for example. In another embodiment, the system is primed 1206 automatically without any selection of a task or instruction from a user or operator. After priming the set, process 1200 proceeds to IC/EC washout 1208, in which fluid on the IC and EC circulation loops may be replaced in preparation for cell culturing. The replacement volume may be specified by the number of IC Volumes and EC Volumes exchanged, according to embodiments. Next, to allow media to reach equilibrium with the gas supply prior to the loading of cells, process 1200 proceeds to condition media task 1210. For example, rapid contact between the media and the gas supply may be provided by using a high EC circulation rate. In an embodiment, the system may then be maintained in a proper state until the user or operator is ready to load cells into the bioreactor. In embodiments, a user or operator may not be needed to perform the noted steps/operations; rather, the steps/operations may be performed automatically by the cell expansion system.


Process 1200 next proceeds to loading cells with uniform suspension 1212. In an embodiment, cells may be loaded from a cell inlet bag. IC circulation may be used to distribute the cells. In an embodiment, cells are loaded into the bioreactor from a cell inlet bag. Cells are then chased from the air removal chamber to the IC loop. The distribution of cells is promoted across the membrane via IC circulation with no IC inlet, for example, and thus no ultrafiltration, according to embodiments.


Next, process 1200 proceeds to the optional (shown in a dashed-line format) step of replacing an outlet or waste bag with a media bag (e.g., a substitute media bag) 1214. In an embodiment, the substitute media bag comprises about 0.2 L of media without protein. Other volumes and types of replacement fluid in the substitute media bag may be used in accordance with embodiments of the present disclosure. Process 1200 next proceeds to turning “OFF” or otherwise deactivating one or more pumps 1216. In an embodiment, the IC inlet pump and the EC inlet pump are turned “OFF” or otherwise indicated to stop or deactivate 1216. Such pump deactivation allows chemical signals, such as CCK, to increase in concentration by turning the inlet media flow rate “OFF” to the IC circulation loop and the EC circulation loop. In such embodiments, fluid from the substitute bag may be passively added to the system at the rate of evaporation during conditions of no active inlet fluid flow. In embodiments where the outlet or waste bag is not replaced, fluid may be passively replaced in the system by interrupting protocol procedures being executed and allowing any fluid in the outlet or waste bag (assuming no constituents toxic to cell growth are present in the outlet or waste bag) to be passively added to the system at the rate of evaporation during conditions of no active inlet fluid flow. Such passive addition of fluid avoids adding an excess amount of fluid and, thus, avoids diluting chemical signaling. In an embodiment, the EC circulation pump may remain “ON.” In further embodiments, both the IC circulation pump and the EC circulation pump remain activated or “ON.”


Next, process 1200 proceeds to feeding the cells 1218. In an embodiment, the cell culture may be sampled for cell counts as well by excising a length of tubing to provide a representative cell concentration sample of the IC loop. In other embodiments, cells may be counted by withdrawing a sample from the sampling coil or sample port, for example.


Process 1200 next proceeds to measuring the glucose and/or lactate concentration(s) 1220. At query 1222, it is determined whether the cell culture conditions have reached a minimum tolerance glucose concentration or a maximum tolerance lactate concentration. Such tolerance concentrations may occur earlier or later than day 4, according to embodiments. If the tolerance concentration(s) have not been reached, process 1200 proceeds NO to continue to measure the glucose/lactate concentration(s) 1220. If the tolerance concentration(s) have been reached, process 1200 proceeds YES to the optional (shown in a dashed-line format) step of replacing the substitute media bag (from optional step 1214) with the waste or outlet bag 1224. In an embodiment, the original waste or outlet bag removed at optional step 1214 is used to replace the substitute media bag at optional step 1224. In another embodiment, a different waste or media bag is used to replace the substitute media bag at optional step 1224.


Following optional step 1224, process 1200 proceeds to feed the cells by adding a controlled flow rate to the IC circulation loop and/or the EC circulation loop 1226 once the cell culture conditions have reached a minimum tolerance glucose concentration or a maximum tolerance lactate concentration, for example. In an embodiment, a low flow rate is continuously added to the IC circulation loop and/or the EC circulation loop. Such feeding with the continuous addition of a low flow rate, for example, may occur earlier or later than day 4, according to embodiments.


Harvest operation 1228 next transfers cells in suspension from the IC circulation loop, including cells in the bioreactor, to a harvest bag. Process 1200 then terminates at END operation 1230.


With respect to the processes illustrated in FIGS. 10, 11, and 12, the operational steps depicted 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. Thus, although the processes have been described with steps listed in a particular order, the present disclosure is not limited to such order. In other embodiments, steps may be performed in a different order, in parallel, or any different number of times, e.g., before and after another step. Further, fewer or additional steps may be used in embodiments without departing from the spirit and scope of the present disclosure. For example, where only suspension or non-adherent cells are present, some steps may not be used as they may be used with adherent cells, such as coating the bioreactor 1008 and 1108, allowing cells to attach 1020 and 1120, and releasing cells 1036 and 1126, for example. Even without such steps, FIGS. 10 and 11, for example, may still apply to the expansion of suspension or non-adherent cells, for example, according to embodiments. As a further example, although not shown in FIGS. 10 and 11, an additional step(s) may include replacing the substitute media bag (previously used to replace the outlet or waste bag) with an outlet or waste bag. Such outlet or waste bag may be the original outlet or waste bag used with the system, according to an embodiment. In another embodiment, a different outlet or waste bag may be used to replace the substitute media bag. Also, the parameters, such as lapse of a predetermined period of time, lactate concentration, glucose consumption, and circulation rates, for example, may also be different than those described above, which are provided merely for illustrative purposes. In addition, as indicated above, process 1200 includes some optional steps/sub-steps shown with dashed-line format. However any steps listed above (in any of processes 1000, 1100, and/or 1200) that are not indicated as optional should not be considered as essential to the one or more present inventions, but may be performed in some embodiments of the one or more present inventions and not in others. Further, while some steps, operations and/or sub-operations are described with reference to an operator or user, such steps, operations and/or sub-operations may be performed automatically, according to embodiments.


Finally, FIG. 13 illustrates example components of a computing system 1300 upon which embodiments of the present disclosure may be implemented. Computing system 1300 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 1000, 1100, and 1200 described above. For example, a pre-programmed task may include, “Feed Cells.”


The computing system 1300 may include a user interface 1302, a processing system 1304, and/or storage 1306. The user interface 1302 may include output device(s) 1308, and/or input device(s) 1310 as understood by a person of skill in the art. Output device(s) 1308 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 1310 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 1304 to deduce the location(s) of touch event(s), as understood by those of skill in the art. The processing system 1304 may then map the location of touch events to user interface (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 1308 may include a printer, speaker, etc. Other input devices 1310 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 1304 may include a processing unit 1312 and/or a memory 1314, according to embodiments of the present disclosure. The processing unit 1312 may be a general purpose processor operable to execute instructions stored in memory 1314. Processing unit 1312 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 1314 may include any short-term or long-term storage for data and/or processor executable instructions, according to embodiments. The memory 1314 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 1306 may be any long-term data storage device or component. Storage 1306 may include one or more of the systems described in conjunction with the memory 1314, according to embodiments. The storage 1306 may be permanent or removable. In embodiments, storage 1306 stores data generated or provided by the processing system 1304.


EXAMPLES

Below are examples of protocols that may be used with a cell expansion system, such as CES 500 (FIG. 5), CES 600 (FIG. 6), CES 700 (FIG. 7), CES 800 (FIG. 8), or CES 900 (FIG. 9), for example, that implements features of this disclosure. It is noted that the example protocols below are provided for illustrative purposes and are not intended to limit other embodiments, which may include different steps, parameters, or other features. The example protocols, including the steps (and any sub-steps) of loading cells and distributing cells, for example, 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) are performed through the combination of automated and manual execution of operations. In further embodiments, the steps (and any sub-steps) are performed by an operator(s) or user(s) or through other manual means.


Example 1: Protocol 1
Day: −1 Coat Bioreactor

This part of the example protocol coats a bioreactor with a reagent. The bioreactor may include a hollow fiber membrane.

    • Step 1: loads a reagent into the IC loop until the bag is empty.
    • Step 2: chases the reagent from the ARC into the IC loop.
    • Step 3: circulates the reagent in the IC loop.


Before starting this task, the following preconditions may be satisfied:

    • Include a minimum of 40 mL of air in the cell inlet bag.


Table 1 describes the bags of solution that are attached to each line when performing the Coat Bioreactor portion of the protocol. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 1







Solutions for Coat Bioreactor













Volume



Bag
Solution in Bag
(estimation)






Cell Inlet
None
N/A



Reagent
Fibronectin
about 5 mg Fibronectin in about





100 mL PBS



IC Media
None
N/A



Wash
PBS
about 0.1 L + 6 mL/hr (overnight)



EC Media
None
N/A









The values for each setting for step 1 may be used as shown in Table 2.









TABLE 2







Step 1 for Coat Bioreactor











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
Reagent




IC Inlet Rate
about 10 mL/min




IC Circulation Rate
about 100 mL/min




EC Inlet
None




EC Inlet Rate
about 0 mL/min




EC Circulation Rate
about 30 mL/min




Outlet
EC Waste




Rocker Control
Stationary,





approximately (0°)




Stop Condition
Empty Bag









Values for each setting for step 2 shown in Table 3 may be used.









TABLE 3







Step 2 Settings for Coat Bioreactor











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
Wash




IC Inlet Rate
about 10 mL/min




IC Circulation Rate
about 100 mL/min




EC Inlet
None




EC Inlet Rate
about 0 mL/min




EC Circulation Rate
about 30 mL/min




Outlet
EC Waste




Rocker Control
Stationary,





approximately (0°)




Stop Condition
about IC Volume





(22 mL)









Values for each setting for step 3 shown in Table 4 may be used.









TABLE 4







Step 3 Settings for Coat Bioreactor











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
about 0 mL/min




IC Circulation Rate
about 20 mL/min




EC Inlet
Wash




EC Inlet Rate
about 0.1 mL/min




EC Circulation Rate
about 30 mL/min




Outlet
EC Waste




Rocker Control
Stationary,





approximately (0°)




Stop Condition
Manual









Day: 0 IC EC Washout

This part of the example protocol is performed to replace the fluid on both the IC circulation loop and the EC circulation loop. The replacement volume may be specified by the number of IC Volumes and EC Volumes exchanged.


Table 5 describes the bags of solution that are attached to each line when performing IC EC Washout of this example protocol. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 5







Solutions for IC EC Washout











Volume


Bag
Solution in Bag
(estimation)





Cell Inlet
None
N/A


Reagent
None
N/A


IC Media
Media with Protein
about 1.4 L


Wash
None
N/A


EC Media
None
N/A









The values for IC EC Washout shown in Table 6 may be used.









TABLE 6







Task Settings for IC EC Washout











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
IC Media




IC Inlet Rate
about 100 mL/min




IC Circulation Rate
about −17 mL/min




EC Inlet
EC Media
IC Media



EC Inlet Rate
about 148 mL/min




EC Circulation Rate
about −1.7 mL/min




Outlet
IC and EC Waste




Rocker Control
In Motion





approximately





(−90°, 180°, 1 sec)




Stop Condition
Exchange





(about 2.5 IC





Volumes)





(about 2.5 EC





Volumes)









Day: 0 Condition Media

This part of the example protocol is performed to allow the media to reach equilibrium with the provided gas supply before loading the cells. This task may include two separate steps:

    • Step 1: provides rapid contact between the media and the gas supply by using a high EC circulation rate.
    • Step 2: maintains the system in a proper state until the operator is ready to load the cells.


Table 7 describes the bags of solution that are attached to each line when performing Condition Media. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 7







Solutions for Condition Media











Volume


Line
Solution in Bag
(estimation)





Cell Inlet
None
N/A


Reagent
None
N/A


IC Media
None
N/A


Wash
None
N/A


EC Media
Media without Protein
about 0.1 L plus 6 mL/hour









The values for step 1 shown in Table 8 may be used.









TABLE 8







Step 1 Settings for Condition Media











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
about 0 mL/min




IC Circulation Rate
about 100 mL/min




EC Inlet
EC Media
IC Media



EC Inlet Rate
about 0.1 mL/min




EC Circulation Rate
about 250 mL/min




Outlet
EC Waste




Rocker Control
Stationary,





approximately (0°)




Stop Condition
Time (about 10





min)









The values for step 2 shown in Table 9 may be used.









TABLE 9







Step 2 Settings for Condition Media











Example
Example




Factory
Laboratory



Setting
Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
0




IC Circulation Rate
about 100 mL/min




EC Inlet
EC Media
IC Media



EC Inlet Rate
about 0.1 mL/min




EC Circulation Rate
about 30 mL/min




Outlet
EC Waste




Rocker Control
Stationary,





approximately (0°')




Stop Condition
Manual









Day: 0 Load Cells with Circulating Distribution

This part of the example protocol is performed to loads cells into the bioreactor from a cell inlet bag. IC circulation may be used to distribute the cells and may not attempt to chase the cells from the line into the bioreactor. This task may include three separate steps.

    • Step 1: loads the cells from the cell inlet bag into the bioreactor.
    • Step 2: chases the cells from the ARC to the bioreactor. Larger chase volumes spread the cells and move them towards the IC outlet.
    • Step 3: promotes distribution of cells across membrane via IC circulation and no IC inlet thus no ultrafiltration.


Before starting this task, the following preconditions may be satisfied:

    • Include a minimum of 40 mL of air in the cell inlet bag.


Table 10 describes the bags of solution attached to each line when performing Load Cells with Circulating Distribution. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 10







Solutions for Load Cells With Circulating Distribution











Volume


Line
Solution in Bag
(estimation)





Cell Inlet
Cells
Cells in about 100 mL




complete media


Reagent
None
N/A


IC Media
Media with Protein
about 0.1 L


Wash
None
N/A


EC Media
None
N/A









The values for step 1 shown in Table 11 may be used.









TABLE 11







Step 1 Settings for Load Cells With Circulating Distribution












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
None

Cells


IC Inlet Rate
about 0 mL/min

about 25 mL/min


IC Circulation
about 0 mL/min.

about 150 mL/min


Rate


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation
about 0 mL/min

about 30 mL/min


Rate


Outlet
EC Waste


Rocker Control
Stationary,

In Motion,



approximately

approximately





(−90°, 180°, 1 sec)


Stop Condition
Manual

Empty Bag









The values for step 2 shown in Table 12 may be used.









TABLE 12







Step 2 Settings for Load Cells with Circulating Distribution












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
None

IC Media


IC Inlet Rate
about 0 mL/min

about 25 mL/min


IC Circulation
about 0 mL/min.

about 150 mL/min


Rate


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation
about 0 mL/min

about 30 mL/min


Rate


Outlet
EC Waste


Rocker Control
Stationary,

In Motion,



approximately

approximately





(−90°, 180°, 1 sec)


Stop Condition
Manual

IC Volume (about





47 mL)









The values for step 3 shown in Table 13 may be used.









TABLE 13







Step 3 Settings for Load Cells with Circulating Distribution












Laboratory



Setting
Factory Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
about 0 mL/min


IC Circulation
about 0 mL/min.

about 200 mL/min


Rate


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation
about 0 mL/min

about 30 mL/min


Rate


Outlet
EC Waste


Rocker Control
Stationary,

In Motion,



approximately

approximately





(−90°, 180°, 1 sec)


Stop Condition
Manual

Time (about 2.0 min)









Day: 0 Attach Cells

This part of the example protocol is performed to enable adherent cells to attach to the bioreactor membrane while allowing flow on the EC circulation loop. The pump flow rate to the IC loop is set to approximately zero.


Table 14 describes the bags of solution attached to each line when performing Attach Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 14







Solutions for Attach Cells













Volume (estimation based



Bag
Solution in Bag
on factory default values)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
None
N/A



Wash
None
N/A



EC Media
None
N/A



Waste
Base Media
500 mL










The values for Attach Cells shown in Table 15 may be used.









TABLE 15







Task Settings for Attach Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
about 0 mL/min


IC Circulation Rate
about 0 mL/min


EC Inlet
EC Media
None


EC Inlet Rate
about 0.1 mL/min
0


EC Circulation Rate
about 30 mL/min


Outlet
EC Waste


Rocker Control
Stationary,



approximately (0°)


Stop Condition
Manual









Day: 1 Feed Cells

This part of the example protocol is performed to continuously add a low flow rate to the IC circulation loop and/or the EC circulation loop. There are several outlet settings that may used to remove the fluid added to the system.


Table 16 describes the bags of solution attached to each line when performing Feed Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 16







Solutions for Feed Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
Media with Protein
about 6 mL/hour



Wash
None
N/A



EC Media
None
N/A



Waste
Base Media
about 500 mL










The values for step 1 shown in Table 17 may be used.









TABLE 17







Task Settings for Feed Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
IC Media




IC Inlet Rate
about 0.1 mL/min
0 mL/min


IC Circulation Rate
about 20 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
IC Waste


Rocker Control
Stationary,



approximately (0°)


Stop Condition
Manual









The IC Inlet rate may be increased as needed. As one example, the IC inlet rate may be increased as follows: Day 1-Day 2: 0.0 mL/min; Day 2-Day 3: 0.1 mL/min; Day 3-Day 4: 0.2 mL/min; Day 4-Day 5: 0.4 mL/min; and Day 5-Day 6: 0.8 mL/min.


Release Adherent Cells

This part of the example protocol is performed to release cells from the membrane, leaving the cells in the IC loop.

    • Step 1: performs the IC/EC Washout task in preparation for adding a reagent. For example, the system replaces IC/EC media with PBS to remove protein, Ca++, and Mg++ in preparation for adding trypsin.
    • Step 2: loads a reagent into the system until the bag is empty.
    • Step 3: chases the reagent into the IC loop.
    • Step 4: mixes the reagent within the IC loop.


Before starting this task, the following preconditions may be satisfied:

    • Include a minimum of 40 mL of air in the cell inlet bag.


Table 18 describes the bags of solution attached to each line when performing Release Adherent Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 18







Solutions for Release Adherent Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
Trypsin
about 180 mL



IC Media
None
N/A



Wash
PBS
about 1.4 L



EC Media
None
N/A










The values for step 1 shown in Table 19 may be used.









TABLE 19







Step 1 Settings for Release Adherent Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
Wash




IC Inlet Rate
about 100 mL/min


IC Circulation Rate
about −17 mL/min


EC Inlet
Wash


EC Inlet Rate
about 148 mL/min


EC Circulation Rate
about −1.7 mL/min


Outlet
IC and EC Waste


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
Exchange



(about 2.5 IC



Volumes)



(about 2.5 EC



Volumes)









The values for step 2 shown in Table 20 may be used.









TABLE 20







Step 2 Settings for Release Adherent Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
Reagent




IC Inlet Rate
about 50 mL/min


IC Circulation Rate
about 300 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Waste


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
Empty Bag









The values for step 3 shown in Table 21 may be used.









TABLE 21







Step 3 Settings for Release Adherent Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
Wash




IC Inlet Rate
about 50 mL/min


IC Circulation Rate
about 300 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Waste


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
IC Volume (22 mL)









The values for step 4 shown in Table 22 may be used.









TABLE 22







Step 4 Settings for Release Adherent Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
None




IC Inlet Rate
about 0 mL/min


IC Circulation Rate
about 300 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Waste


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
Time (about 4 min)









Samples may be taken from a sample coil and/or a sample port for a trypsin assay.


Harvest Cells

This part of the example protocol is performed to transfer cells in suspension from the IC circulation loop, including cells in the bioreactor, to the harvest bag.


Table 23 describes the bags of solution attached to each line when performing Harvest Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 23







Solutions for Harvest Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
Harvest Media
about 0.6 L



Wash
None
N/A



EC Media
None
N/A










The values for Harvest Cells shown in Table 24 may be used.









TABLE 24







Task Settings for Harvest Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications





IC Inlet
IC Media




IC Inlet Rate
about 400 mL/min


IC Circulation Rate
about −69 mL/min


EC Inlet
EC Media
IC Media


EC Inlet Rate
about 60 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
Harvest


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
IC Volume (about



378 mL)









Example 2: Protocol 2

Cholecystokinin (CCK) is a regulatory hormone secreted by cells and, in many cases, may in part be responsible for cell culture maintenance and proliferation via chemical signaling. If CCK concentration in the culture media does not reach a threshold, the cell population can be compromised. Example 2 provides an example of a cell-secreted chemical signal used to maintain and proliferate a population of cells in vitro; in this case, CHO cells. According to an embodiment, the molecular weight of CCK of approximately 4,000 Daltons makes it small enough to readily pass through the microporous membrane of a hollow-fiber bioreactor. In an embodiment, regardless of inlet media addition to the IC circulation loop or EC circulation loop, dilution of the chemical signal may occur due to the freedom to pass through the membrane. However, through the passive replacement of media, according to embodiments, such dilution of chemical signaling can be minimized or eliminated altogether. The following protocol provides for the passive replacement of media during the cell expansion of non-adherent or suspension cells, such as CHO cells, for example, according to embodiments.


Day: 0 IC EC Washout

This part of the example protocol is performed to replace the fluid on both the IC circulation loop and the EC circulation loop in preparation for cell culturing. The replacement volume may be specified by the number of IC Volumes and EC Volumes exchanged.


Table 25 describes the bags of solution that are attached to each line when performing IC EC Washout of this example protocol. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 25







Solutions for IC EC Washout













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
Media without Protein
about 1.4 L



Wash
None
N/A



EC Media
None
N/A










The values for IC EC Washout shown in Table 26 may be used.









TABLE 26







Task Settings for IC EC Washout












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
IC Media



IC Inlet Rate
about 100 mL/min


IC Circulation Rate
about −17 mL/min


EC Inlet
EC Media
IC Media


EC Inlet Rate
about 148 mL/min


EC Circulation Rate
about −1.7 mL/min


Outlet
IC and EC Waste


Rocker Control
In Motion



approximately



(−90°, 180°, 1 sec)


Stop Condition
Exchange



(about 2.5 IC



Volumes)



(about 2.5 EC



Volumes)









Day: 0 Condition Media

This part of the example protocol is performed to allow the media to reach equilibrium with the provided gas supply before loading the cells. This task may include two separate steps:

    • Step 1: provides rapid contact between the media and the gas supply by using a high EC circulation rate.
    • Step 2: maintains the system in a proper state until the operator is ready to load the cells.


Table 27 describes the bags of solution that are attached to each line when performing Condition Media. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 27







Solutions for Condition Media











Volume


Line
Solution in Bag
(estimation)





Cell Inlet
None
N/A


Reagent
None
N/A


IC Media
Media without Protein
about 0.1 L plus 6 mL/hour


Wash
None
N/A


EC Media
None
N/A









The values for step 1 shown in Table 28 may be used.









TABLE 28







Step 1 Settings for Condition Media












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
None



IC Inlet Rate
about 0 mL/min


IC Circulation Rate
about 100 mL/min


EC Inlet
EC Media
IC Media


EC Inlet Rate
about 0.1 mL/min


EC Circulation Rate
about 250 mL/min


Outlet
EC Waste


Rocker Control
Stationary,



approximately (0°)


Stop Condition
Time (about 10



min)









The values for step 2 shown in Table 29 may be used.









TABLE 29







Step 2 Settings for Condition Media












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
None



IC Inlet Rate
0


IC Circulation Rate
about 100 mL/min


EC Inlet
EC Media
IC Media


EC Inlet Rate
about 0.1 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Waste


Rocker Control
Stationary,



approximately (0°)


Stop Condition
Manual









Day: 0 Load Cells with Uniform Suspension

This part of the example protocol is performed to load cells into the bioreactor from a cell inlet bag. For example, in an embodiment, such cells comprise CHO cells. IC circulation may be used to distribute the cells and may not attempt to chase the cells from the line into the bioreactor. This task may include three separate steps.

    • Step 1: loads the cells from the cell inlet bag into the bioreactor.
    • Step 2: chases the cells from the ARC to the IC Loop.
    • Step 3: promotes distribution of cells across membrane via IC circulation and no IC inlet, thus no ultrafiltration.


Before starting this task, the following preconditions may be satisfied:

    • Include a minimum of 40 mL of air in the cell inlet bag.


Table 30 describes the bags of solution attached to each line when performing Load Cells with Uniform Suspension. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 30







Solutions for Load Cells with Uniform Suspension











Volume


Line
Solution in Bag
(estimation)





Cell Inlet
Cells
9.45E+07 Cells in about 100 mL




media


Reagent
None
N/A


IC Media
Media without Protein
about 0.1 L


Wash
None
N/A


EC Media
None
N/A









The values for step 1 shown in Table 31 may be used.









TABLE 31







Step 1 Settings for Load Cells with Uniform Suspension












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications











IC Inlet
Cell Inlet


IC Inlet Rate
about 50 mL/min


IC Circulation Rate
about 200 mL/min.


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Outlet


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
Empty Bag









The values for step 2 shown in Table 32 may be used.









TABLE 32







Step 2 Settings for Load Cells with Uniform Suspension












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications











IC Inlet
IC Media


IC Inlet Rate
about 50 mL/min


IC Circulation Rate
about 200 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Outlet


Rocker Control
In Motion



(−90°, 180°, 1 sec)


Stop Condition
IC Volume (about



22 mL)









The values for step 3 shown in Table 33 may be used.









TABLE 33







Step 3 Settings for Load Cells with Uniform Suspension












Laboratory



Setting
Factory Default
Default
Modifications











IC Inlet
None


IC Inlet Rate
about 0 mL/min


IC Circulation Rate
about 200 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
EC Outlet


Rocker Control
In Motion



(−90°, 180°, 1 sec)


Stop Condition
Time (about 2.0



min)









Day: 0 Feed Cells

This part of the example protocol is performed to allow chemical signals, such as CCK, to increase in concentration by turning the inlet media flow rate “OFF” to the IC circulation loop and the EC circulation loop. IC or EC Outlet can be used in this configuration.


Table 34 describes the bags of solution attached to each line when performing Feed Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 34







Solutions for Feed Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
None
N/A



Wash
None
N/A



EC Media
None
N/A



Outlet
Media without Protein
0.2 L










The values for step 1 shown in Table 35 may be used.









TABLE 35







Task Settings for Feed Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
IC Media
None


IC Inlet Rate
about 0.1 mL/min
about 0 mL/min


IC Circulation Rate
about 20 mL/min
about 50 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
IC Outlet
EC Outlet


Rocker Control
Stationary,
In Motion



approximately
(0°, 180°,



(0°)
about 60 sec)


Stop Condition
Manual









In an embodiment, each day, the cell culture is sampled for cell counts using the following settings:









TABLE 36







Task Settings for Counting Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
None



IC Inlet Rate
about 0 mL/min


IC Circulation Rate
about 200 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
IC Outlet
EC Outlet


Rocker Control
In Motion



(0°, 180°, 1 sec)


Stop Condition
Time (about 5.0



min)









In an embodiment, immediately following the stop condition, a length of tubing of about six (6) inches long (1 mL) is excised. The volume in this sample provides a representative cell concentration sample of the entire IC loop. This allows the user(s) to monitor the cells throughout the duration of culturing.


Day: 4 Feed Cells

This part of the example protocol is performed to continuously add a low flow rate to the IC circulation loop and/or the EC circulation loop once the cell culture conditions have reached a minimum tolerance glucose concentration or maximum tolerance lactate concentration. This may occur earlier or later than day 4, in embodiments. There are several outlet settings that may used to remove the fluid added to the system.


Table 37 describes the bags of solution attached to each line when performing Feed Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 37







Solutions for Feed Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
Media without Protein
about 6 mL/hour



Wash
None
N/A



EC Media
None
N/A










The values for step 1 shown in Table 38 may be used.









TABLE 38







Task Settings for Feed Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
IC Media



IC Inlet Rate
about 0.1 mL/min


IC Circulation Rate
about 20 mL/min
about 50 mL/min


EC Inlet
None


EC Inlet Rate
about 0 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
IC Outlet
EC Outlet


Rocker Control
Stationary,
In Motion



approximately
(0°, 180°,



(0°)
about 60 sec)


Stop Condition
Manual









In an embodiment, each day, the cell culture is sampled for cell counts (see Table 36, for example).


Day: 7 Harvest Cells

This part of the example protocol is performed to transfer cells in suspension from the IC circulation loop, including cells in the bioreactor, to the harvest bag.


Table 39 describes the bags of solution attached to each line when performing Harvest Cells. These solutions and corresponding volumes are provided as one example of default settings that may be used.









TABLE 39







Solutions for Harvest Cells













Volume



Bag
Solution in Bag
(estimation)







Cell Inlet
None
N/A



Reagent
None
N/A



IC Media
Harvest Media
about 0.6 L



Wash
None
N/A



EC Media
None
N/A










The values for Harvest Cells shown in Table 40 may be used.









TABLE 40







Task Settings for Harvest Cells












Example




Example Factory
Laboratory


Setting
Default
Default
Modifications












IC Inlet
IC Media



IC Inlet Rate
about 400 mL/min


IC Circulation Rate
about −69 mL/min


EC Inlet
EC Media
IC Media


EC Inlet Rate
about 60 mL/min


EC Circulation Rate
about 30 mL/min


Outlet
Harvest


Rocker Control
In Motion,



approximately



(−90°, 180°, 1 sec)


Stop Condition
IC Volume (about



378 mL)









It will be apparent to those skilled in the art that various modifications and variations may be made to the apparatus, systems, structure, and methods described herein. Thus, it should be understood that the embodiments are not limited to the subject matter discussed in the present disclosure. Rather, the present disclosure is intended to cover modifications, variations, and/or equivalents. The acts, features, structures, and/or media are disclosed as illustrative embodiments for implementation of the claims.

Claims
  • 1. A cell expansion system, comprising: a bioreactor comprising 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, wherein the first fluid flow path comprises an intracapillary portion of the hollow fiber membrane;a fluid inlet path fluidly associated with the first fluid flow path, wherein a plurality of cells are introduced into the first fluid flow path through the first fluid inlet path;a first pump that circulates fluid in the first fluid flow path of the bioreactor;an outlet line in fluid communication with the bioreactor, wherein the outlet line is attached, at a first time, to an outlet bag in the cell expansion system; anda controller that controls operation of the first pump, wherein the controller is configured to control the first pump to: circulate a fluid at a first rate within the first fluid flow path; andstop, when the outlet bag is replaced with a media bag at a second time, the circulation of the fluid within the first fluid flow path after the plurality of the cells are loaded into the bioreactor, wherein the outlet line is attached to the media bag at the second time; andwherein media from the media bag is added to the bioreactor while the circulation of the fluid within the first fluid flow path is stopped.
  • 2. The cell expansion system of claim 1, wherein the media bag comprises base media.
  • 3. The cell expansion system of claim 1, wherein the cell expansion system is closed.
  • 4. The cell expansion system of claim 1, wherein the plurality of cells comprises adherent cells.
  • 5. The cell expansion system of claim 1, wherein the plurality of cells comprises non-adherent cells.
  • 6. The cell expansion system of claim 1, further comprising: a second pump that transfers intracapillary inlet fluid from an intracapillary media bag to the first fluid flow path;a second controller that controls operation of the second pump, wherein the second controller is configured to control the second pump to: transfer the plurality of cells from a cell inlet bag to the first fluid flow path; andstop, at the second time, the transfer of the plurality of cells from the cell inlet bag after the plurality of the cells are loaded into the bioreactor.
  • 7. The cell expansion system of claim 1, wherein the bioreactor further comprises: a second fluid flow path comprising an extracapillary portion of the hollow fiber membrane, wherein the outlet line is in fluid communication with the bioreactor via the second fluid flow path.
  • 8. The cell expansion system of claim 7, wherein the media from the media bag is added to the bioreactor by pumping the media through the second fluid flow path.
  • 9. The cell expansion system of claim 8, further comprising: an extracapillary pump that pumps the media from the media bag along the outlet line and through the second fluid flow path.
  • 10. A cell expansion system, comprising: a bioreactor comprising: 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, wherein the first fluid flow path comprises an intracapillary portion of the hollow fiber membrane; anda second fluid flow path comprising an extracapillary portion of the hollow fiber membrane;a fluid inlet path fluidly associated with the first fluid flow path, wherein a plurality of cells are introduced into the first fluid flow path through the fluid inlet path;a first pump that circulates fluid in the first fluid flow path of the bioreactor;a second pump that circulates fluid in the second fluid flow path of the bioreactor;an outlet line in fluid communication with the bioreactor via the second fluid flow path wherein the outlet line comprises an attachment to one of an outlet bag or a media bag;a processor coupled with the first pump; anda memory coupled with and readable by the processor and storing therein instructions that, when executed by the processor, cause the processor to: circulate, via operating the first pump, a fluid at a first rate within the first fluid flow path while the outlet line is attached to the outlet bag;stop, via turning off the first pump, the circulation of the fluid within the first fluid flow path after the plurality of the cells are loaded into the bioreactor and when the outlet line is attached to the media bag instead of the outlet bag; andadd, via operating the second pump while the first pump is turned off, media from the media bag to the bioreactor when the outlet line is attached to the media bag instead of the outlet bag.
  • 11. The cell expansion system of claim 10, wherein the media bag comprises base media, and wherein the cell expansion system is closed.
  • 12. The cell expansion system of claim 10, wherein at least one of the first pump and the second pump are peristaltic pumps.
  • 13. The cell expansion system of claim 10, wherein an amount of the media from the media bag added to the bioreactor replaces an amount of fluid evaporated from the bioreactor.
  • 14. The cell expansion system of claim 10, further comprising: a valve fluidly interconnected to the outlet line, wherein the valve selectively controls a flow of the media from the media bag.
  • 15. The cell expansion system of claim 14, wherein, prior to adding the media from the media bag to the bioreactor, the instructions further cause the processor to maintain the valve in an open state.
  • 16. The cell expansion system of claim 10, further comprising: an intracapillary media bag comprising intracapillary inlet fluid;a cell inlet bag comprising the plurality of cells; anda third pump that transfers the intracapillary inlet fluid from the intracapillary media bag to the first fluid flow path.
  • 17. The cell expansion system of claim 16, wherein the instructions further cause the processor to: transfer, via operating the third pump, the plurality of cells from the cell inlet bag and the intracapillary inlet fluid from the intracapillary media bag to the first fluid flow path; andstop, via turning off the first pump, the transfer of the plurality of cells from the cell inlet bag after the plurality of the cells are loaded into the bioreactor.
  • 18. A cell expansion method, comprising: loading a disposable tubing set onto a cell expansion system, the disposable tubing set comprising: a bioreactor comprising 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, wherein the first fluid flow path comprises an intracapillary portion of the hollow fiber membrane;a fluid inlet path fluidly associated with the first fluid flow path; andan outlet bag; andan outlet line attached to the outlet bag and in fluid communication with the bioreactor;pumping, by a first pump of the cell expansion system, a plurality of cells into the first fluid flow path through the fluid inlet path;circulating, by the first pump, a fluid at a first rate within the first fluid flow path;stopping, by a processor when the outlet bag is replaced with a media bag at a second time, the circulation of the fluid within the first fluid flow path after the plurality of the cells are loaded into the bioreactor, wherein the outlet line is attached to the media bag at the second time; andpumping, by a second pump of the cell expansion system, media from the media bag through the outlet line to the bioreactor while the circulation of the fluid within the first fluid flow path is stopped.
  • 19. The method of claim 18, wherein stopping the circulation of the fluid within the first fluid flow path after the plurality of the cells are loaded into the bioreactor comprises: deactivating, by the processor, the first pump causing the first pump to turn off.
  • 20. The method of claim 18, wherein pumping the plurality of cells into the first fluid flow path through the fluid inlet path further comprises: pumping, via a third pump of the cell expansion system, the plurality of cells from a cell inlet bag and intracapillary inlet fluid from an intracapillary media bag to the first fluid flow path.
CROSS-REFERENCE TO RELATED APPLICATION

This application is a divisional application of, and claims priority to, U.S. patent application Ser. No. 14/668,659, entitled, “Passive Replacement of Media,” filed on Mar. 25, 2015, which claims the benefit of U.S. Provisional Application Ser. No. 61/970,274, filed on Mar. 25, 2014, and entitled, “Passive Replacement of Media.” 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 (1050)
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
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
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
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
5126238 Gebhard et al. Jun 1992 A
5130141 Law et al. Jul 1992 A
5149544 Gentile et al. Sep 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 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
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 Grandics et al. Nov 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
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
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
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
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
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
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
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
7534601 Wikswo 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
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
8895291 DiLorenzo et al. Nov 2014 B2
9057045 Gibbons et al. Jun 2015 B2
9109193 Galliher et al. Aug 2015 B2
9175259 Nankervis Nov 2015 B2
9220810 Ma et al. Dec 2015 B2
9441195 Wojciechowski et al. Sep 2016 B2
9534198 Page et al. Jan 2017 B2
9732313 Hirschel et al. Aug 2017 B2
10093956 Hirschel et al. Oct 2018 B2
10494421 Castillo Dec 2019 B2
11008547 Nankervis May 2021 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
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
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
20040027914 Vrane 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 Vacanti 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
20060019388 Hutmacher 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
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
20070231305 Noll et al. Oct 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
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
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
20100092536 Hunter et al. Apr 2010 A1
20100093607 Dickneite Apr 2010 A1
20100105138 Dodd et al. 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
20100144037 Antwiler 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
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
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
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
20150104431 Pittenger et al. Apr 2015 A1
20150111252 Hirschel et al. Apr 2015 A1
20150125138 Karnieli 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
20160362650 Wojciechowski et al. Dec 2016 A1
20160362652 Page et al. Dec 2016 A1
20180010082 Jacques 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 (319)
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
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
2027247 Feb 2009 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
1758985 Jun 2015 EP
1414671 Nov 1975 GB
2297980 Aug 1996 GB
2360789 Oct 2001 GB
3285 May 2007 HU
H02245177 Sep 1990 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
2019-516029 Jun 2019 JP
2019-525765 Sep 2019 JP
101228026 Jan 2013 KR
10-2015-0002762 Jan 2015 KR
101504392 Mar 2015 KR
101548790 Aug 2015 KR
101553040 Sep 2015 KR
10-2017-0076679 Jul 2017 KR
10-2018-0027501 Mar 2018 KR
102027596 Oct 2019 KR
10-2020-0034790 Mar 2020 KR
10-2020-0058433 May 2020 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
9210564 Jun 1992 WO
WO-9425571 Nov 1994 WO
9504813 Feb 1995 WO
9521911 Aug 1995 WO
WO 9524468 Sep 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
1997-040137 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-0016420 Mar 2000 WO
WO-0017326 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-2001000783 Jan 2001 WO
WO-2001011011 Feb 2001 WO
WO-2001018174 Mar 2001 WO
WO-2001021766 Mar 2001 WO
0123520 Apr 2001 WO
WO-2001025402 Apr 2001 WO
WO-2001029189 Apr 2001 WO
WO-0122810 Apr 2001 WO
WO-2001034167 May 2001 WO
WO-2001049851 Jul 2001 WO
WO-2001054706 Aug 2001 WO
2001-094541 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
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 Aug 2003 WO
WO-2003068961 Sep 2003 WO
WO-2003072064 Sep 2003 WO
WO-2003078609 Sep 2003 WO
WO-2003078967 Oct 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
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-2006121445 Nov 2006 WO
WO-06124021 Nov 2006 WO
WO-06129312 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-2008017128 Feb 2008 WO
WO-2008028241 Mar 2008 WO
WO-08040812 Apr 2008 WO
WO 2008073635 Jun 2008 WO
2008109674 Sep 2008 WO
WO-2008116261 Oct 2008 WO
WO-2008149129 Dec 2008 WO
2009034186 Mar 2009 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-2010019886 Feb 2010 WO
WO-10014253 Feb 2010 WO
WO-10019997 Feb 2010 WO
WO-2010026573 Mar 2010 WO
WO-2010026574 Mar 2010 WO
WO-2010026575 Mar 2010 WO
WO 2010036760 Apr 2010 WO
WO-2010036760 Apr 2010 WO
WO-2010059487 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-10119036 Oct 2010 WO
WO-10123594 Oct 2010 WO
WO-2011025445 Mar 2011 WO
WO 2011098592 Aug 2011 WO
WO 2011130617 Oct 2011 WO
WO-2011132087 Oct 2011 WO
WO-2011147967 Dec 2011 WO
WO-2012072924 Jun 2012 WO
WO-2012127320 Sep 2012 WO
WO-2012138968 Oct 2012 WO
WO-2012140519 Oct 2012 WO
2012171026 Dec 2012 WO
2012171030 Dec 2012 WO
WO-2012171026 Dec 2012 WO
WO-2012171030 Dec 2012 WO
WO 2013085682 Jun 2013 WO
WO-2013110651 Aug 2013 WO
WO-2014037862 Mar 2014 WO
WO-2014037863 Mar 2014 WO
WO-2014068508 May 2014 WO
WO-2014128306 Aug 2014 WO
WO-2014128634 Aug 2014 WO
WO-2014131846 Sep 2014 WO
WO-2014141111 Sep 2014 WO
WO-2015004609 Jan 2015 WO
WO 2015059714 Apr 2015 WO
2015073913 May 2015 WO
WO 2015069943 May 2015 WO
WO 2015118148 Aug 2015 WO
WO 2015118149 Aug 2015 WO
WO-2015131143 Sep 2015 WO
WO 2016130940 Aug 2016 WO
WO 2017072201 May 2017 WO
WO 2017158611 Sep 2017 WO
WO 2017207822 Dec 2017 WO
WO 2018183426 Oct 2018 WO
WO 2019155032 Aug 2019 WO
WO 2019238919 Dec 2019 WO
WO 2020020569 Jan 2020 WO
WO 2020079274 Apr 2020 WO
Non-Patent Literature Citations (293)
Entry
Communication pursuant to Article 94(3) EPC, European Patent Application No. 15718657.8, dated Jul. 21, 2017.
Communication pursuant to Article 94(3) EPC, European Patent Application No. 15718657.8, dated Mar. 22, 2018.
First Office Action, Chinese Patent Application No. 201580020869.5, dated Apr. 27, 2018 (English language translation included).
International Search Report and Written Opinion, PCT/US2015/022541, dated Jul. 17, 2015.
Rejection of the Application, Japanese Patent Application No. 2016-558755, dated Feb. 5, 2019 (English language translation included).
Rejection of the Application, Japanese Patent Application No. 2016-558755, dated Jan. 28, 2020 (English language translation included).
Second Office Action, Chinese Patent Application No. 201580020869.5, dated May 21, 2019 (English language translation included).
Third Office Action, Chinese Patent Application No. 201580020869.5, dated Nov. 6, 2019 (English language translation included).
Chang et al., “Membrane Bioreactors: Present and Prospects”, Advances in Biochemical Engineering, 1991, pp. 27-64, vol. 44.
Chang, Ho Nam, “Membrane Bioreactors: Engineering Aspects”, Biotech. Adv., 1987, pp. 129-145, vol. 5.
Edgington, Stephen M., “New Horizons for Stem-Cell Bioreactors”, Biotechnology, Oct. 1992, pp. 1099-1106, vol. 10.
Gastens et al., “Good Manufacturing Practice-Compliant Expansion of Marrow-Derived Stem and Progenitor Cells for Cell Therapy”, Cell Transplantation, 2007, pp. 685-696, vol. 16.
Gramer et al., “Screening Tool for Hollow-Fiber Bioreactor Process Development”, Biotechnol. Prog., 1998, pp. 203-209, vol. 14.
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, 1987, pp. 113-144, Hanser Publishers.
Infanger et al., “Simulated weightlessness changes the cytoskeleton and extracellular matrix proteins in papillary thyroid carcinoma cells”, Cell and Tissue Research, 2006, 324(2): 267-277.
Jones et al., “Genetic stability of bone marrow-derived human mesenchymal stromal cells in the Quantum System”, Cytotherapy, 2013; 15: 1323-1339.
Liu et al., “Ex vivo Expansion of Hematopoietic Stem Cells Derived from Umbilical Cord Blood in Rotating Wall Vessel”, Journal of Biotechnology, 2006, 124:592-601.
Nankervis et al., “Shear Stress Conditions in the Quantum Cell Expansion System”, Poster Session—TERMIS AM Annual Conference 2013, Nov. 12, 2013.
Nguyen et al., “QUANTUM® Cell Expansion System: Automated Expansion of Human Mesenchymal Stem Cells from Precultured Cells Using the Quantum Cell Expansion System”, Terumo BCT, Inc., 2012.
Nielsen, Lars Keld, “Bioreactors for Hematopoietic Cell Culture”, Annu. Rev. Biomed. Eng., 1999, vol. 1, pp. 129-152.
Office Action, Chinese Patent Application No. 201580020869.5, dated Apr. 27, 2018. (English language translation included).
Office Action, Chinese Patent Application No. 201580020869.5, dated May 21, 2019. (English language translation included).
Official Communication, European Patent Application No. 15718657.8, dated Jul. 21, 2017.
Official Communication, European Patent Application No. 15718657.8, dated Mar. 22, 2018.
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.
The Extended European Search Report, European Patent Application No. 19202519.5, dated Nov. 15, 2019.
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.
“The Effect of Rocking Rate and Angle on T Cell Cultures Grown in XuriTM Cell Expansion Systems,” GE Healthcare UK Limited, Cell therapy bioreactor systems, Application note 29-1166-55 AA, Aug. 2014, www.gelifesciences.com/xuri.
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.
Alenazi et al., “Modified Polyether-sulfone Membrane: a Mini Review,” Designed Monomers And Polymers, vol. 20, No. 1, 2017, pp. 532-546.
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.
Baecher-Allan et al., “CD4+CD25high Regulatory Cells in Human Peripheral Blood,” The Journal of Immunology, vol. 167, 2001, pp. 1245-1253.
Bai et al., “Expansion of Primitive Human Hematopoietic Stem Cells by Culture in a Zwitterionic Hydrogel,” Nature Medicine, vol. 25, Oct. 2019, pp. 1566-1575.
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, Apr. 23, 2019, pp. 1267-1271.
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 Cells 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.
Garlie et al., “T Cells Coactivated with Immobilized Anti-CD3 and Anti-CD28 as Potential Immunotherapy for Cancer,” Journal of immunotherapy, vol. 22, No. 4, 1999, pp. 336-345.
Gloeckner et al., “New Miniaturized Hollow-Fiber Bioreactor for in Vivo Like Cell Culture, Cell Expansion, and Production of Cell-Derived Products,” Biotechnology Progress, vol. 17, Aug. 21, 2001, pp. 828-831.
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.
Horwitz et al., “Phase I/II Study of Stem-Cell Transplantation Using a Single Cord Blood Unit Expanded Ex Vivo with Nicotinamide,” Journal of Clinical Oncology, vol. 37, No. 5, Dec. 4, 2018, pp. 367-376.
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 II2rγnull 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.
Rahmahwati et al., “The Synthesis of Polyethersuifone (PES) Derivatives for the Immobilization of Lipase Enzyme,” Key Engineering Materials, vol. 811, Jul. 8, 2019, pp. 14-21.
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.
Streltsova et al., “Recurrent Stimulation of Natural Killer Cell Clones with K562 Expressing Membrane-Bound Interleukin-21 Affects Their Phenotype, Interferon-γ Production, and Lifespan,” International Journal of Molecular Sciences, vol. 20, No. 443, 2019, pp. 1-18.
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 Leukemia Cell lines,” PLoS ONE, vol. 4, Issue 1, Jan. 22, 2009, e4251.
Ueda et al., “Interaction of Natural Killer Cells with Neutrophils Exerts a Significant Antitumor Immunity in Hematopoietic Stem Cell Transplantation Recipients,” Cancer Medicine, vol. 5, No. 1, 2016 pp. 49-60.
Urbich et al., “Fluid Shear Stress-induced Transcriptional Activation of the Vascular Endothelial Growth Factor Receptor-2 Gene Requires Sp1-Dependent DNA Binding,” FEBS Letters, 535, 2003, pp. 87-93.
Von Laer, “Loss of CD38 Antigen on CD34 CD38 Cells during Short-term Culture,” Leukemia, Correspondence, 1999 pp. 947-948.
Wagner et al., “Phase I/II Trial of StemRegenin-1 Expanded Umbilical Cord Blood Hematopoietic Stem Cells Supports Testing as a Stand-alone Graft,” Cell Stem Cell, Jan. 7, 2016, vol. 18, pp. 144-155.
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, vol. 86, No. 10, Nov. 15, 1995, pp. 3691-3969.
Yang et al., “Suspension Culture of Mammalian Cells Using Thermosensitive Microcarrier that Allows Cell Detachment without Proteolytic Enzyme Treatment,” Cell Transplantation, vol. 19, Aug. 18, 2010, pp. 1123-1132.
Yi et al., “A Readily Modified Polyethersuifone with Amino-Substituted Groups: Its Amphiphilic Copolymer Synthesis and Membrane Application,” Polymer, vol. 53, Dec. 2, 2011, pp. 350-358.
Zheng et al., “Differential Effects of Cyclic and Static Stretch on Coronary Microvascular Endothelial Cell Receptors and Vasculogenic/Angiogenic Responses,” American Journal of Physiology—Heart and Circulatory Physiology, vol. 295, Aug. 2008, H794-H800.
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.
Almeida L, Lochner M, Berod L, Sparwasser T. 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.
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, Lom H, Thirugnanabalan B, Wedderburn 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”, John Wiley & Sons. Current Protocols in Protein Science (1995) 5.3.1-5.3.18.
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 O. 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.
Bluestone, Jeffrey A., et al. “Type 1 diabetes immunotherapy using polyclonal regulatory T cells.” Science translational medicine 7.315 (2015): 315ra189-315ra189.
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.
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.
Chapman NM, Chi H. mTOR signaling, Tregs and immune modulation. Immunotherapy. 2014;6(12):1295-311.
Chaudhry A, Samstein RM, Treuting 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 I/II study of intravenous allogeneic mesenchymal stromal cells in acute myocardial infarction. Cytotherapy 17(3): 250-261.
Claudio G. Brunstein, Jeffrey S. Miller, Qing Cao, Daivd H. McKenna, Keli L. Hippen, Julie Curtsinger, Todd Defor, Bruce L. Levine, Carl H. June, Pablo Rubinstein, Philip B. McGlave, 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.
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, Tessler 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, Tournier-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, Gugger M, Gazdhar A, 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):210-25.
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):769-77.
Fuchs A, Gliwinski M, Grageda N, Spiering R, Abbas AK, Appel S, Bacchetta R, Battaglia M, Berglund D, Blazar B, Bluestone JA, Bornhauser 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):2567-75.
Gedaly R, De Stefano F, Turcios L, Hill M, Hidalgo G, Mitov MI, 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):807-26.
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.
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.
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-beta1 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.
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.
Horwitz, Mitchell E., et al. “Phase I/II 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.
http://www.ucdenver.edu/academics/colleges/medicalschool/centers/cancercenter/Research/sharedresources/AnimalImaging/smallanimalimaging/Pages/MRI.aspx.
ISCT Webinar “vol. 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.
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.
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 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.
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):1392-9.
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.
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.
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.
Klysz D, Tai X, Robert PA, Craveiro M, Cretenet G, Oburoglu L, Mongellaz C, Floess S, Fritz V, Matias MI, 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.
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 OX40 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.
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.
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. coli 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.
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.
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.
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.
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 | 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-1alpha 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, Wing 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.
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.
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 U S A. 2008;105(29):10113-8.
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 U S A. 2018;115(28):E6546-E6555.
Parhi, Purnendu, Avantika Golas, 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.
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.
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 non-human 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 100.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.
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.
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):546-58.
Rudensky, Alexander Y. “Regulatory T cells and Foxp3.” Immunological reviews 241.1 (2011): 260-268.
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.
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.
Stuart, Martien A. Cohen, et al. “Emerging applications of stimuli-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 U S A. 2016;113(41):E6192-E6198.
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.
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):139-47.
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):237-40.
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.
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: Single-cell 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):454-63.
Related Publications (1)
Number Date Country
20180119094 A1 May 2018 US
Provisional Applications (1)
Number Date Country
61970274 Mar 2014 US
Divisions (1)
Number Date Country
Parent 14668659 Mar 2015 US
Child 15849309 US