Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SYSTEMS AND METHODS FOR HANDLING AND TRANSFERRING FLUIDS ASEPTICALLY VIA MICROFLUIDIC COMPONENTS
Document Type and Number:
WIPO Patent Application WO/2023/220055
Kind Code:
A1
Abstract:
An Integrated fully automated closed fluid handling system includes a set of fill-finish aseptic cartridges filled at least partially with a liquid or lyophilized and/ or dried, reagents, a microfluidic or fluidic conduit, a reusable or disposable consumable with channels and hollow tubes or syringes used for piercing a septum on the cartridges, integrating an actuator to push cartridges towards hollow tubes to pierce the septum or seal such that the liquids are transferred from the cartridges to the microfluidic or fluidic conduit, a robot arm or tweezer to transfer the cartridges into a consumable, a barcode scanner to confirm the QR code on the cartridges, a consumable handling system, a flow control system to provide automated control of various aspects of the liquid handling process. The integrated and fully automated closed fluid handling system may include a cartridge retraction mechanism to retract the cartridges.

Inventors:
MIANSARIGAVZAN MORTEZA (US)
DATWANI SAMMY (US)
RAZINKOV IVAN (US)
WHITNEY BRANDON (US)
Application Number:
PCT/US2023/021531
Publication Date:
November 16, 2023
Filing Date:
May 09, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SYNTHEGO CORP (US)
International Classes:
B01L3/00; A61M3/02; A61M5/14; B01L9/00
Foreign References:
US20110005932A12011-01-13
US20170105779A12017-04-20
US20130115607A12013-05-09
US20160038942A12016-02-11
US20190168221A12019-06-06
Attorney, Agent or Firm:
KEATING, Thomas, J. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A microfluidic system for transferring fluid, the system comprising: a housing having a first chamber including a first cannula, a second chamber including a second cannula, and a channel in fluid communication with the cannula of the first chamber and the cannula of the second chamber; a source cartridge defining an internal volume, the source cartridge having a barrier that seals the internal volume of the source cartridge from the ambient environment; a recipient cartridge defining an internal volume, the recipient cartridge having a barrier that seals the internal volume of the recipient cartridge from the ambient environment; one or more actuators that are configured to: insert the source cartridge into the first chamber of the housing until the cannula of the first chamber passes through the barrier of the source cartrid ge and enters into the internal volume of the source cartridge; and insert the recipient cartridge into the second chamber of the housing until the cannula of the second chamber passes through the barrier of the recipient cartridge and enters into the internal volume of the recipient cartridge; and a pump system configured to drive liquid from the internal volume of the source cartridge, through the cannula of the first chamber, through the channel, through the cannula of the second chamber and into the internal volume of the recipient cartridge.

2. The microfluidic system of claim 1, wherein the system is configured for transferring a volume of less than about 10 ml, 1000 μL, 100 μL, 10 μL, 1 μL, 0.5 μL, 0.2 , 0.1 μL, 0.01 μL, 0.001 μL, 100 pL, 10 pL, or IpL from the source cartridge to the recipient cartridge.

3. The microfluidic system of claim 1 or 2, wherein the liquid driven from the source cartridge to the recipient cartridge has a volume of less than about 10 mL, 1000 μL, 100 μL, 10 μL, 1 μL, 0.5 μL, 0.2 , 0.1 μL, 0.01 μL, 0.001 μL, 100 pL, 10 pL, or IpL.

4. The microfluidic system of any of the foregoing claims, wherein the pump system includes a pump that drives the liquid from the source cartridge to the recipient cartridge, and wherein the minimum displacement of the pump is less than about 10 mL, 1000 μL, 100 μL, 10 μL, 1 μL, 0.5 μL, 0.2 , 0.1 μL, 0.01 uL, 0.001 μL, 100 pL, 10 pL, or IpL.

5. The microfluidic system of any of the foregoing claims, wherein the pump drives liquid from the internal volume of the source cartridge to the internal volume of the recipient cartridge by at least one of removing gas from the recipient cartridge and adding gas into the source cartridge.

6. The microfluidic system of any of the foregoing claims, wherein the housing includes a second elongated cannula in the first chamber, wherein the second elongated cannula in the first chamber is longer than the first cannula in the first chamber; and the housing includes a second elongated cannula in the second chamber, wherein the second elongated cannula in the second chamber is longer than the first cannula in the second chamber.

7. The microfluidic system of claim 6, wherein a distal end of the elongated cannula of the first chamber is positioned above the top surface of a liquid contained in the source cartridge when the source cartridge is inserted in the first chamber; and wherein a distal end of the elongated cannula of the second chamber is positioned above the top surface of a liquid contained in the recipient cartridge when the recipient cartridge is inserted in the second chamber.

8. The microfluidic system of claim 6, wherein the pump is in fluid communication with the elongated cannula of the first chamber, and wherein the pump is configured to direct gas through the elongated cannula of the first chamber and into the internal volume of the source cartridge thereby driving the liquid from the source cartridge to the second cartridge.

9. The microfluidic system of any of claims 6-8, wherein the pump is in fluid communication with the elongated cannula of the second chamber, and wherein the pump is configured to remove gas through from the internal volume of the second cartridge through the elongated cannula of the second chamber thereby driving the liquid from the source cartridge to the recipient cartridge.

10. The microfluidic system of any of claims 1-9, wherein the source cartridge contains a first liquid, and wherein the recipient cartridge contains a second liquid, and wherein when the first liquid enters the recipient cartridge the first liquid mixes with the second liquid to form a mixed liquid.

11. The microfluidic system of claim 10, wherein the pump is configured to drive at least a portion of the mixed liquid back into the internal volume of the source cartridge thereby further mixing the first and second liquids together.

12. The microfluidic system of any of claims 1-9, wherein the recipient cartridge contains a component that is dehydrated or concentrated, wherein the component dissolves or dilutes in the in the liquid that is driven from the source cartridge to the recipient cartridge.

13. The microfluidic system of any of claims 1-12, wherein the channel contains a component that is dehydrated or concentrated, and wherein the component dissolves or dilutes in the liquid that is driven from the source cartridge, through the channel, and to the recipient cartridge.

14. The microfluidic system of claim 13, wherein the lyophilized component forms a layer in the channel, and wherein the layer does not block fluid communication between the cannula of the first chamber and the cannula of the second chamber.

15. The microfluidic system of any of the foregoing claims, wherein the barrier of the source cartridge is positioned on a top end of the source cartridge, and the barrier of the recipient cartridge is positioned on a top end of the recipient cartridge, wherein the actuator inserts the source cartridge into the first chamber in an inverted position such that the top end of the source cartridge is inserted first into the first chamber, and/or the actuator inserts the recipient cartridge into the second chamber in an inverted position such that the top end of the recipient cartridge is inserted first into the second chamber.

16. The microfluidic system of any of the foregoing claims, wherein the first cartridge is configured to be inserted into the first chamber in an inverted position, and wherein the second cartridge is configured to be inserted into the second chamber in an inverted position.

17. The microfluidic system of any of the foregoing claims, wherein the barrier of the source cartridge forms a seal between the cannula of the first chamber and the internal volume of the source cartridge after the cannula of the first chamber passes through the barrier.of the source cartridge, and wherein the barrier of the source cartridge reforms a seal after the cannula of the first chamber is removed from the source cartridge, thereby isolating the internal volume of the source cartridge from the ambient environment; and/or wherein the barrier of the recipient cartridge forms a seal between the cannula of the second chamber and the internal volume of the recipient cartridge after the cannula of the second chamber passes through the barrier of the recipient cartridge, and wherein the barrier of the recipient cartridge reforms a seal after the cannula of the second chamber is removed from the recipient cartridge, thereby isolating the internal volume of the recipient cartridge from the ambient environment.

18. The microfluidic system of any of the foregoing claims, wherein the barrier of the source cartridge is a resealable septum, and/or wherein the barrier of the recipient cartridge is a resealable septum.

19. The microfluidic system of any of the foregoing claims, wherein the housing comprises an upper part comprising the first chamber and the second chamber, and the housing comprises a lower part comprising the channel in fluid communication with the cannula of the first chamber and the cannula of the second chamber.

20. The microfluidic system of claim 19, wherein the upper part of the housing and the lower part of the housing are coupled aseptically.

21. The microfluidic system of 19 or 20, wherein the upper part of the housing and the lower part of the housing are modular and the lower part of the housing may be replaced with a different lower part having a different channel in order to establish a different fluid communication between a first chamber and a second chamber of the upper part.

22. The microfluidic system of any of the foregoing claims, wherein the housing includes a third chamber including a third cannula, the third cannula being in fluid communication with the channel, and further comprising: a third cartridge defining an internal volume, the third cartridge having a barrier that isolates the internal volume of the third cartridge from the ambient environment, and wherein the pump system is configured to drive a portion of the liquid from the first container, through the first cannula, through the channel, through the third cannula into the internal volume of the third cartridge.

23. The microfluidic system of any of the foregoing claims, wherein the housing comprises multiple chambers for receiving one or more source cartridges and/or one or more recipient cartridges, wherein the one or more source cartridges and the one or more recipient cartridges are placed in fluid communication when the one or more source cartridges and the one or more recipient cartridges are inserted in the multiple chambers.

24. The microfluidic system of claim 23 comprising multiple channels to establish fluid communications between multiple source cartridges and multiple recipient cartridges.

25. The microfluidic system of claim 24, wherein the housing comprises an upper part comprising the multiple chambers, and the housing comprises a lower part comprising the multiple channels that establish fluid communications between the multiple chambers of the upper part.

26. The microfluidic system of claim 25, wherein the upper part of the housing and the lower part of the housing are coupled aseptically.

27. The micro fluidic system of 25 or 26, wherein the upper part of the housing and the lower part of the housing are modular and the lower part of the housing may be replaced with a different lower part having different multiple channels in order to establish different fluid communication between the multiple chambers of the upper part.

28. The microfluidic system of any of the foregoing claims, wherein the pump system includes a pump and a computing device, the pump comprising: a channel defining an inlet and an outlet, the inlet and the outlet defining a flow path for fluid: a first valve positioned within the flow path; a second valve positioned within the flow path; a chamber positioned between the first valve and the second valve within the flow path and defining a chamber volume; and a membrane extending across the chamber and sealing the chamber from the flow' path; wherein the computing device is in communication with a pressure source, and the computing device and pressure source are configured to close and open the first valve, and the computing device and the pressure source are configured to close and open the second valve, thereby controlling the fluid flow through the flow path; and wherein the pump is configured such that when the first valve is open and the second valve is closed, the computing device and pressure source cause the membrane to move and create negative pressure in the chamber such that fluid is drawn from the inlet, through the channel, and into the flow path; and wherein the pump is configured such that when the first valve is closed and the second valve is open, the computing device and pressure source cause the membrane to move and create positive pressure in the chamber such that fluid is driven from the flow path, through the channel, and through the outlet.

29. The microfluidic system of claim 28, wherein the pump system includes one or more pressure sources in pressure communication with the first valve, the membrane, and the second valve, wherein the one or more pressure sources are configured to: close the first valve by moving the first valve into the flow path thereby blocking the flow path; close the second valve by moving the second valve into the flow path thereby blocking the flow path; and move the membrane upwardly within the chamber and downwardly within the chamber and modulate the volume of the chamber volume.

30. The microfluidic system of claim 28 or 29, wherein the one or more pressure sources are configured to: apply pressure to the first valve thereby moving the first valve into the flow path and blocking the flow path and release pressure and/or apply a vacuum to the first valve thereby moving the first valve from the flow path and opening the flow path; apply pressure to the second valve thereby moving the second valve into the flow path and blocking the flow path and release pressure and/or apply a vacuum to the second valve thereby moving the second valve from the flow path and opening the flow path; and apply pressure into the chamber to move the membrane downwardly within the chamber and release pressure and/or apply a vacuum to the chamber to move the membrane upwardly within the chamber.

31 . The microfluidic system of any of claims 28-30, wherein the one or more pressure sources include a first pressure source, a second pressure source, and a third pressure source, the first pressure source configured to adjust the pressure applied to the first valve, the second pressure source configured to adjust the pressure applied to the second valve, and the third pressure source configured to adjust the pressure applied to the chamber.

32. The microfluidic system of claim 31, wherein the pump system includes: a first part that includes the flow path; a second part that includes the inlet, the outlet, the first valve, and the chamber, and wherein the membrane is positioned between the first part and second part and the membrane seals the chamber,

33. The microfluidic system of any of the foregoing claims comprising multiple housings comprising multiple chambers for receiving multiple source cartridges and multiple recipient cartridges.

34. The microfluidic system of any of the foregoing claims, wherein the liquid is at least one of a biological product, cells, a pharmaceutical product, or a biopharmaceutical product.

35. The microfluidic system of any of the foregoing claims, wherein the liquid that is driven from the source cartridge to the recipient cartridge comprises a dissolved component; and wherein the recipient cartridge contains a second liquid such that the dissolved component is diluted when the first liquid enters the recipient cartridge and mixes with the second liquid.

36. The microfluidic system of claim 35, wherein the dissolved component comprises one or more of a compound, a biomolecule, a biochemical species, a protein, an antibody, a biopharmaceutical, a pharmaceutical, or cells.

37. A microfluidic system comprising: a housing including a chamber, a first cannula positioned within the chamber, and a second cannula that is longer than the first cannula positioned within the chamber; a cartridge defining an internal volume, the cartridge having a barrier that isolates the internal volume from the ambient environment; an actuator configured to drive the cartridge into the chamber of the housing until the first cannula and the second cannula pass through the barrier and enter into the internal volume of the cartridge, wherein the barrier forms a seal between the first cannula and the internal volume of the cartridge and the barrier forms a seal between the second cannula and the internal volume of the cartridge, after the first cannula and the second cannula pass through the barrier and enter into the internal volume of the cartridge, thereby sealing the internal volume of the cartridge from the ambient environment; and a pump system configured to perform at least one of removing liquid from the cartridge by adding gas through the second cannula into the internal volume of the cartridge thereby forcing the liquid out of the cartridge, and adding liquid to the cartridge by removing gas from the internal volume of the cartridge via the second cannula.

38. The microfluidic system of claim 37, wherein the system is configured for transferring a volume of less than about 10 mL, 1000 μL, 100 μL, 10 pl 1 μL, 0.5 μL, 0.2 , 0.1 μL, 0.01 μL, 0.001 μL, 100 pL, 10 pL, or IpL from the cartridge to a recipient cartridge.

39. The microfluidic system of claim 37 or 38, wherein the pump system includes a pump that drives the liquid from the cartridge to a recipient cartridge, and wherein the minimum displacement of the pump is less than about 10 mL, 1000 μL, 100 μL, 10 μL, 1 μL, 0.5 μL, 0,2 , 0.1 μL, 0.01 μL, 0.001 μL, 100 pL, 10 pL, or IpL.

40. The microfluidic system of any of claims 37-39, wherein the pump system includes a pump and a computing device, the pump comprising: a channel defining an inlet and an outlet, the inlet and the outlet defining a flow path for fluid; a first valve positioned within the flow path; a second valve positioned within the flow path; a chamber positioned between the first valve and the second valve within the flow path and defining a chamber volume; and a membrane extending across the chamber and sealing the chamber from the flow path; wherein the computing device is in communication with a pressure source, and the computing device and pressure source are configured to close and open the first valve, and the computing device and the pressure source are configured to close and open the second valve, thereby controlling the fluid flow through the flow path; and wherein the pump is configured such that when the first valve is open and the second valve is closed, the computing device and pressure source cause the membrane to move and create negative pressure in the chamber such that fluid is drawn from the inlet, through the channel, and into the flow path: and wherein the pump is configured such that when the first valve is close and the second valve is open, the computing device and pressure source cause the membrane to move and create positive pressure in the chamber such that fluid is driven from the flow7 path, through the channel and through the outlet.

41. The microfluidic system of claim 40, wherein the pump system includes one or more pressure sources in pressure communication with the first valve, the membrane, and the second valve, wherein the one or more pressure sources are configured to: close the first valve by moving the first valve into the flow' path thereby blocking the flow path; close the second valve by moving the second valve into the flow path thereby blocking the flow path; and move the membrane upwardly within the chamber and downwardly within the chamber and modulate the volume of the chamber volume.

42. The microfluidic system of claim 40 or 41 , wherein the one or more pressure sources are configured to: apply pressure to the first valve thereby moving the first valve into the flow path and blocking the flow path and release pressure and/or apply a vacuum to the first valve thereby moving the first valve from the flow' path and opening the flow' path; apply pressure to the second valve thereby moving the second valve into the flow path and blocking the flow path and release pressure and/or apply a vacuum to the second valve thereby moving the second valve from the flow path and opening the flow' path; and apply pressure into the chamber to move the membrane downwardly within the chamber and release pressure and/or apply a vacuum to the chamber to move the membrane upwardly within the chamber.

43. The microfluidic system of any of claims 40-42, wherein the one or more pressure sources include a first pressure source, a second pressure source, and a third pressure source, the first pressure source configured to adjust the pressure applied to the first valve, the second pressure source configured to adjust the pressure applied to the second valve, and the third pressure source configured to adjust the pressure applied to the chamber.

44. The micro fluidic system of any of claims 37-43, wherein the pump system includes: a first part that includes the flow path; a second part that includes the inlet, the outlet, the first valve, and the chamber, and wherein the membrane is positioned between the first part and second part and the membrane seals the chamber.

45. The microfluidic system of claim 44, wherein the inlet of the pump is in fluid communication with a liquid sample.

46. The microfluidic system of any of the foregoing claims, wherein the pump system includes a pump that is at least one of an electrowetting pump, an acoustic pump, a piezoelectric pump, a capillary pump, a pneumatic pump, a syringe pump, a dosing pump, or a gear pump.

47. The microfluidic system of any of the foregoing claims, wherein the microfluidic system is configured to move liquid from the source cartridge to the recipient cartridge aseptically.

48. The microfluidic system of claim 47, wherein the barrier of the source cartridge is a first barrier, the barrier of the recipient cartridge is a second barrier, and wherein the housing includes: a third barrier extending across the first chamber of the housing, the third barrier positioned above the first cannula and isolating the first chamber from the ambient environment; and a fourth barrier extending across the second chamber of the housing, the fourth barrier positioned above the second cannula and isolating the second chamber from the ambient environment.

49. The microfluidic system of claim 48, wherein when the source cartridge is inserted into the first chamber, the first cannula of the first chamber passes through the third barrier, and wherein when the recipient cartridge is inserted into the second chamber, the second cannula of the second chamber passes through the fourth barrier.

50. A method for moving liquid aseptically, the method comprising moving liquid in any of the microfluidic systems of the foregoing claims from the source cartridge to the recipient cartridge.

51 . The method of claim 50, wherein the method comprises moving a volume of liquid less than about 1000, 100, 10, 1, 0.5, 0.2, or 0.1 μL.

52. The method of claim 50 or 51 wherein the microfluidic system comprises multiple housings and the method comprises moving a liquid from a source cartridge to a recipient cartridge in a first housing, and after the liquid has been moved the method comprises transferring the recipient cartridge to a second housing.

53. The method of claim 52, further comprising moving a liquid from the recipient cartridge to a second recipient cartridge in the second housing

54. The method of any of claims 50-53, wherein the housing comprises an upper part comprising chambers for receiving cartridges and a lower part comprising channels for moving liquid from cartridges inserted in the chambers.

55. An integrated, automated, aseptic, closed fluid handling system, comprising: a consumable with microfluidic or fluidic conduit, which carries cartridges or vials containing reagents; a consumable handling system; a robotic tweezer; a barcode scanner; cartridges or vials; a flow control system; and a cartridge or vial retraction system.

56. The system of claim 55, further comprising software, scripts, and electronic board for controlling and automating the different processes described in the present disclosure.

57. The system of claim 56, wherein the software includes a straightforward, user- friendly interface to run different protocols through a visual representation of upper surface of a consumable that displays the holes, ports, vias, or chambers to be selected.

58. The system of claim 55, further comprising:

(i) a consumable storage area, wherein the consumable handling system can move the consumables from the consumable storage area to a certain position on the belt; and/or

(ii) a system for flushing or decontaminating the fluidic conduit if the consumable tends to be reused; and/or

(iii) an agitation mechanism, wherein external agitator/s can be integrated to the system under the consumable; and/or

(iv) an alignment system to ensure placement of the consumable in a correct position and direction on the belt; and/or

(v) a data output system.

59. The system of claim 58, wherein the consumable storage area is an automated consumable stacker and/or handler to deliver a new consumable to the bel t at a certain time interval.

60. The system of claim 58, wherein the flushing system comprises a few extra holes, ports, vias, or chambers in the consumable to store the cleaning reagents and pump into the fluidic conduit as needed.

61. The system of claim 58, wherein the agitation mechanism is a piezoelectric-based planar ultrasonicator, which is acoustically coupled to the support plate or deck under the belt at the formulation station.

62. The system of claim 58, wherein the data output system is a monitor or touch screen or any other visual display device, an ethernet and/or RS232 and USB, cloud-based connections, a CD-ROM drive, a printer.

63. The system of claim 55, wherein the consumable comprises a microfluidic conduit network plate, and a block comprising holes, ports, vias, or chambers as well as piercing hollow tubes.

64. The system of claim 63, wherein the fluidic conduit comprises channels or grooves that are fabricated either in the upper surface of the bottom plate, or lower surface of the top piece, or in both surfaces.

65. The system of claim 64, wherein the channels or grooves of the fluidic conduit are fabricated in an interlayer material like double-sided adhesive and is sandwiched between the bottom plate and top piece.

66. The system of claim 63, wherein the piercing hollow tubes include one long hollow tube to communicate with the flow control system, and one or more short hollow tubes through which liquids are transferred.

67. The system of claim 55, wherein the fluidic conduit comprises the tree-like network and/or disk-like or the radial network designs for the QC sampling and aliquoting samples.

68. The system of claim 55, wherein the consumable handling system comprises a conveyor to move the consumable along the belt.

69. The system of claim 68, wherein the conveyor includes at least a motor, a belt, two sets of ball bearings, one or two rollers,

70. The system of claim 69, wherein the conveyor comprises spindles, pulleys, gears, or other devices instead of rollers to move a belt.

71. The system of claim 55, wherein the consumable handling system includes a heating and/or cooling mechanism and an integrated thermocouple,

72. The system of claim 71, wherein the heating and/or cooling mechanism comprises a heating or cooling module which is located under the belt and so under the consumable.

73. The system of claim 55, wherein the robotic tweezer comprises at least a robot arm and two fingers to pick and drop cartridges or vials into certain holes or chambers in the consumable,

74. The system of claim 55, wherein the barcode scanner comprises a vision module and QR-code recognition algorithm.

75. The system of claim 74, wherein the vision module and QR-code recognition algorithm are integrated to achieve two functions: first, it recognizes the cartridge or vial information encoded by a QR-code, second, provides the spatial coordinates of cartridges to the robot arm to allocate and capture the targeted cartridges.

76. The system of claim 55, wherein the cartridges or vials are usually made of sterile glass or plastic depending on the samples or contents as well as the storage methods.

77. The system of claim 76, wherein the content of cartridges or vials which are filled at least partially are in liquid or lyophilized or dry powder forms.

78. The system of claim 55, wherein the cartridges or vials include a unique QR-code on the side and/or the bottom.

79. The system of claim 78, wherein the QR-codes contain information about the contents of the cartridges or vials, and about the partial coordinates of the cartridges or vials,

80. The system of claim 55, wherein the cartridges or vials comprise a septum or seal in their cap and/or a stopper or plunger at their rear, which is usually made of silicone rubber.

81. The system of claim 55, wherein the flow control system comprising:

(a), positive or negative pressure and vent manifolds and/or tubes; and/or

(b). mechanical, hydraulic, or pneumatic actuators to push down the cartridges or vials inside the holes or chambers in the consumable; and/or

(c). valves that are pneumatically, hydraulically, and/or electrically actuated; and/or

(d). thin, planar load cells that are integrated with the actuators to control the force on the cartridges or vials and or consumable, which is applied by actuators; and/or

(e). piercing hollow tubes integrated with filters; and

(f). a height adjustment system.

82. The system of claim 81, wherein the pneumatic actuator includes at least a cylinder, two valves, a piston, a spring, and/or a load cell attached to the piston.

83. The system of claim 82, wherein the hydraulic actuator comprises at least a cylinder, two valves, a piston, a spring, and/or a load cell attached to the piston.

84. The system of claim 82, wherein the mechanical actuator comprises at least a stepper motor, a set of gear transition arrangements connected to the motor, a transition shaft, a set of screws, a plunger, and a piston.

85. The system of claim 82, wherein the load cell is a planar force transducer or sensor that converts the force, c.g., compression or pressure between the actuator and the cartridge or vial, into an electrical signal.

86. The system of claim 82, wherein the piercing hollow tube comprises at least a stiff stainless steel perforated closed trocar point hollow' tube, and wherein the piercing hollow tube pierces the cartridge or vial from the top of the consumable, and its other end communicates with positive or negative pressure, or vent manifold through an air filter.

87. The system of claim 82, wherein the height adjustment system includes at least a stepper motor, a lead screw, a nut, a linear shaft, and/or a bail bearing and a flexible coupling.

88. The system of claim 55, wherein cartridge or vial retraction system includes a high tensile spring which is loaded into the hole or chamber where cartridge or vial is loaded in the consumable.

89. The system of claim 55, wherein cartridge or vial retraction system includes a high tensile spring and a U-shaped metal pin which is under the consumable and slides back and forth into the cartridges or vials’ holes or chambers via two guide through-holes at the bottom of these holes or chambers.

90. The system of claim 89, wherein the cartridge or vial retraction system comprises an electromagnetic holder attached to the bottom of the high tensile spring to stop or hold the U-shaped metal pin for a certain period.

91. A consumable comprising: at least two valves and a diaphragm which form a series of chambers that can be opened and closed in orchestrated fashion to create an on board fluidic pump, and wherein the size of the diaphragm and the volume of the chamber that contains the diaphragm control the minimum single pump action volume of the fluidic pump, and wherein a pump is operably coupled to one or more reagent vials for metered liquid dispensing.

92. The consumable of claim 91. further comprising multiple on board fluidic pumps with different diaphragm sizes to allow for accurate fluid dispensing at a variety of different volumes.

93. The consumable of claim 91, wherein the valves can be actuated mechanically, electrically, or pneumatically using a flow control system.

Description:
SYSTEMS AND METHODS FOR HANDLING AND TRANSFERRING FLUIDS ASEPTICALLY VIA MICROFLUIDIC COMPONENTS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001} The present application claims the benefit of priority under 35 U.S.C. §119(e) to U.S. Provisional Application No. 63/339,892, as filed May 9, 2022, the contents of which are incorporated herein by reference in its entirety,

BACKGROUND

[0002] Formulation of complex biological reagents, such as those used in cell genome engineering (e.g., CRISPR), often require different reagents to be mixed properly. However, mixing reagents can sometimes take undesirably long periods of time, and in some cases, contaminants can be undesirably introduced into the reagents or the final formulation. Thus, it would be desirable to have improved systems and methods for integration and automation of aseptic cartridges for fluid handling, reconstitution, and formulation.

SUMMARY OF THE DISCLOSURE

[0003] Some embodiments of the disclosure provide a microfluidic system for transferring fluid. The microfluidic system can include a housing having a first chamber including a first cannula, a second chamber including a second cannula, and a channel in fluid communication with the cannula of the first chamber and the cannula of the second chamber, a source cartridge defining an internal volume, the source cartridge having a barrier that seals the internal volume of the source cartridge from the ambient environment, a recipient cartridge defining an internal volume, the recipient cartridge having a barrier that seals the internal volume of the recipient cartridge from the ambient environment, and one or more actuators that are configured to insert the source cartridge into the first chamber of the housing until the cannula of the first chamber passes through the barrier of the source cartridge and enters into the internal volume of the source cartridge; and insert the recipient cartridge into the second chamber of the housing until the cannula of the second chamber passes through the barrier of the recipient cartridge and enters into the internal volume of the recipient cartridge. The microfluidic system can include a pump system configured to drive liquid from the internal volume of the source cartridge, through the cannula of the first chamber, through the channel, through the cannula of the second chamber and into the internal volume of the recipient cartridge.

[0004] Some embodiments of the disclosure provide a microfluidic system. The microfluidic system can include a housing including a chamber, a first cannula positioned within the chamber, and a second cannula that is longer than the first cannula positioned within the chamber, a cartridge defining an internal volume, the cartridge having a barrier that isolates the internal volume from the ambient environment, an actuator configured to drive the cartridge into the chamber of the housing until the first cannula and the second cannula pass through the barrier and enter into the internal volume of the cartridge, wherein the barrier forms a seal between the first cannula and the internal volume of the cartridge and the barrier forms a seal between the second cannula and the internal volume of the cartridge, after the first cannula and the second cannula pass through the barrier and enter into the internal volume of the cartridge, thereby sealing the internal volume of the cartridge from the ambient environment, and a pump system configured to perform at least one of removing liquid from the cartridge by adding gas through the second cannula into the internal volume of the cartridge thereby forcing the liquid out of the cartridge, and adding liquid to the cartridge by removing gas from the internal volume of the cartridge via the second cannula.

[0005] Some embodiments of the disclosure provide a method for moving liquid aseptically. The method can include moving liquid in any of the microfluidic systems described herein from the source cartridge to the recipient cartridge.

BRIEF DESCRIPTION OF THE DRAWINGS

]0006] The following drawings are provided to help illustrate various features of embodiments of the disclosure and are not intended to limit the scope of the disclosure or exclude alternative configurations.

[0007] FIG. 1 shows an isometric view of an automated closed fluid handling system, in accordance with certain aspects of the present disclosure.

[0008] FIG. 2 show's an isometric view of the system of FIG. 1 with a housing, in accordance with certain aspects of the present disclosure.

[0009] FIG. 3 shows a cross-sectional view of the system shown in FIG. 2, in accordance with certain aspects of the present disclosure. [0010] FIG. 4A shows an isometric view' of the flow control system in a position of use, in accordance with certain aspects of the present disclosure.

[0011] FIG. 4B shows an isometric view of the flow control system of FIG. 4A in a position of use, in accordance with certain aspects of the present disclosure.

[0012] FIG. 5 A shows an isometric view of the system of FIG. 1, illustrating an embodiment of a consumable handling system, in accordance with certain aspects of the system of FIG. 1.

[0013] FIG. 5B shows an isometric view of the system of FIG. 1, illustrating an embodiment of a flow control system, in accordance with certain aspects of the system of FIG. 1.

[0014] FIG. 6A shows an exploded isometric view of an exemplary consumable, in accordance with certain aspects of the present disclosure.

[0015] FIG. 6B shows a schematic isometric view of an embodiment of an exemplary' consumable of the present disclosure.

[0016] FIG. 6C show's top and side views of an embodiment of an exemplary controlled liquid transfer and side views of micro valving mechanisms, in accordance with certain aspects of the present disclosure.

[0017] FIG. 7A shows an exploded isometric with a cross-section of a consumable of FIG. 6B, in accordance with certain aspects of the present disclosure.

[0018] FIG. 7B shows another exploded isometric with a cross-section of a consumable of FIG. 6B, in accordance with certain aspects of the present disclosure.

[0019] FIG. 7C shows another exploded isometric with a cross-section of a consumable of FIG. 6B, in accordance with certain aspects of the present disclosure.

[0020] FIG. 8A is an isometric view with a partial cross-section of an exemplary pneumatic cylinder actuation system integrated with a consumable and cartridge(s), in accordance with certain aspects of the present disclosure.

[0021] FIG. 8B shows a side view of an exemplary pneumatic cylinder actuation system for use with a consumable and cartridge(s), in accordance with certain aspects of the present disclosure. [0022] FIG. 9A shows a cross-section view of an exemplary fluid transfer mechanism integrated with a pneumatic cylinder actuation system of FIG. 8A, in accordance with certain aspects of the present disclosure.

[0023] FIG. 9B shows a cross-section view of an alternative embodiment of an exemplary fluid transfer mechanism of the present disclosure.

[0024] FIG. 9C shows a cross-section view of an alternative embodiment of an exemplary fluid transfer mechanism, in accordance with certain aspects of the present disclosure.

[0025 ] FIG. 9D shows a cross-section view of an alternative embodiment of an exemplary fluid transfer mechanism, in accordance with certain aspects of the present disclosure.

[0026] FIG. 10A shows a side view' of an embodiment of a cartridge retraction mechanism, in accordance with certain aspects of the present disclosure.

[0027] FIG. 10B shows a side view of an alternative embodiment of a cartridge retraction mechanism, in accordance with certain aspects of the present disclosure.

[0028] FIG. 1 IA shows a top view of an embodiment of a QC sampling or aliquoting system, in accordance with certain aspects of the present disclosure.

[0029] FIG. 1 IB shows a top view of an alternative embodiment of a QC sampling or aliquoting mechanism, in accordance with certain aspects of the present disclosure,

[0030] FIG. 12A shows a photograph of the prototype of an exemplary incomplete consumable used for the proof-of-concept study, in accordance with certain aspects of the present disclosure.

[0031] FIG. 12B shows a photograph of the prototype of an exemplary complete consumable used for the proof-of-concept study, in accordance with certain aspects of the present disclosure.

[0032] FIG. 13A shows a photograph of the prototype utilized in the experimental setup for the proof-of-concept study, in accordance with certain aspects of the present disclosure.

[0033] FIG. 13B shows a photograph of the prototype of a partial cross-section of an exemplary consumable, used for the proof-of-concept study, in accordance with certain aspects of the present disclosure.

[0034] FIG. 13C shows a photograph of the prototype of a partial cross-section of an exemplary consumable after a liquid transfer, used for the proof-of-concept study, in accordance with certain aspects of the present disclosure. [0035] FIG. 13D shows a photograph of the prototype of a partial cross-section of an exemplary consumable after repeated liquid transfer, used for the proof-of-concept study, in accordance with certain aspects of the present disclosure.

[0036] FIG. 14A shows an isometric view of an embodiment of an exemplar}' consumable of the present disclosure.

[0037] FIG. 14B shows a top view of microfluidic conduits of an embodiment to be integrated with the consumable, in accordance with certain aspects of the present disclosure.

DETAILED DESCRIPTION OF THE PRESENT DISCLOSURE

[0038] The present disclosure relates to systems and methods for liquid transfer, reconstitution, mixing, formulation, and sampling or aliquoting of biological reagents and materials for life science applications, molecular biology, and medical devices. More specifically, the embodiments of the present disclosure relate to an integrated and automated closed system for aseptic transfer of liquids out of one or more process cartridges, vials, and/or chambers, while maintaining the sterility during the liquid transfer process, fill-finish cartridges, methods for manufacturing and microliter (and other volume) liquid handling, and their methods of use.

[0039] The system described is of particular value and importance when accurate and precise microliter, aseptic liquid handling of formulations and/or reconstituted reagents for time-sensitive biological materials’ introduction in an integrated, automated, fully closed system is desired. Standard off-the-shelf liquid handling systems cannot provide the desired capability for a variety of reasons such as but not limited to sterility, cross-contamination, and integration into automated end-to-end processes.

[0040] The embodiments of the present disclosure may be further integrated into automated cell therapy manufacturing processes, which require accurate and precise formulation and/or reconstitution workflow steps using sterile, needle-free aseptic, and closed fluid handling mechanisms. Furthermore, the cartridges or vials of the present disclosure can be pre-filled or pre-dosed with reagents, drugs or medicament, biological materials, dried components (e.g., a reagent), dehydrated components (e.g., lyophilized components), concentrated components, etc., for example but not limited to synthetic guide RNA (gRNA), single-stranded donor templates (ssODNs), or pharmaceutical treatments using standard filling systems and equipment.

[0041] One of the key advantages is that this system maintains the sterility of the fluid pathways by only transferring the liquids between these aseptic cartridges via the integrated microfluidic conduit without getting exposed to the surrounding or the adjacent environment, as discussed in more detail below.

[0042] Although the disclosure is not limited to a particular state, the cartridges or vials of the present disclosure may contain liquids, buffers, reagents, or components in a dehydrated form, a concentrated form, a lyophilized form, a reconstituted form, etc., which may also be referred to as biological materials, drugs, or medicaments. The liquid may be but is not limited to, distilled water and buffers used for reconstitution or formulations. The biological materials may be, but are not limited to, gRNA, single-stranded donor templates (ssODNs), cytokines, antibodies, or peptides.

[0043] Certain formulations require and/or are more effective when the reagents are stored separately in cartridges, vials, or containers and combined and mixed at a specific time avoiding degradation or other environmental effects that reduce potency and efficacy. Therefore, there is a high unmet need for the development of a closed fluid and biologic or drug preparation system that can integrate and automate the reconstitution and/or formulation processes.

[0044] One perceived disadvantage of certain known formulators is the inability to accurately and precisely transfer microliter amounts (or less) of liquids aseptically (e.g., along a closed aseptic fluidic pathway isolated from the ambient environment). Almost all known liquid handlers transfer liquids by pipetting liquids from and into open or closed cartridges and/ or vials using special disposable or reusable pipette tips, syringes, or needles. However, the chance of cross-contamination, spills, and aerosolization during these processes are high when such equipment is used, thereby providing a source of contamination. This concern is addressed in the present disclosure where all fluids can be handled in a fully integrated and automated closed system without any exposure to the surroundings or the adjacent environment, which could lead to contamination, degradation, etc., of the formulation components.

[0045] Various aspects of the integrated, automated, closed, aseptic fluid handling system may be illustrated with reference to the accompanying drawings and one or more exemplary implementations. As used herein, the term “exemplary” means “serving as an example, for instance, or illustration,” and should not necessarily be construed as preferred or advantageous over other variations of the devices, systems, or methods disclosed herein.

[0046] FIG. 1 shows an isometric view' of the components of an exemplary fluid handling system 100, which can be automated, aseptic, and closed. For example, the fluid handling system 100 can facilitate the distribution of one or more fluids (e.g., a liquid) aseptically. In some cases, this can include the fluid handling system 100 moving one or more fluids from one or more containers (including a cartridge) to one or more other containers along one or more flow' paths that are isolated from the ambient environment (e.g., that surrounds the fluid handling system 100). The fluid handling system 100 can include a consumable 602, which can include a housing having one or more chambers 124 (e.g., each of which can alternatively be a hole, a port, a vial, a channel, a recess, etc.) that are configured to receive a respective cartridge 128 (e.g., each cartridge 128 is configured to be inserted into a respective chamber 124). The consumable 602 can include one or more channels 122 (e.g., each of which can alternatively be a hole, a port, a vial, a channel, a recess, etc.) that are configured to be placed into (and out of) fluid communication with a pressure source (e.g., a negative pressure source, a positive pressure source, etc.). For example, each channel 122, which can have a smaller diameter than a chamber 124, can be coupled to a respective conduit 134, which can provide positive or negative pressure to the corresponding channel 122.

[0047] The consumable 602 can include a top piece 106, and a bottom piece 126 that can be aligned with and coupled to a bottom of the top piece 106. As shown in FIG. 1, the top piece 106 can include the one or more chambers 124, and the one or more channels 122, and the bottom piece 126 can include one or more fluidic conduits (e.g., that can be aligned with the one or more channels 122, the one or more chambers 124, etc., of the top piece 106), each of which can be a microfluidic conduit. In some embodiments, a height of the top piece 106 can be larger than a height of the bottom piece 126, which can include the height of the top piece 106 being substantially (i.e., deviating by less than 10 percent from) larger than a height of the bottom piece 126. In some embodiments, the height of the top piece 106 can be at least twice, at least three times, etc., the height of the bottom piece 126. While the top piece 106 is illustrated in FIG. 1 as being separate from the bottom piece 126 (but coupled together), in alternative configurations, the consumable 602 can be a single monolithic component (e.g., with the top piece 106 being integrally formed with the bottom piece 12.6).

[0(M8] In some embodiments, the fluid handling system 100 can include a robot arm 102 that can include a gripper (e.g., tweezers, fingers, etc.) to access the cartridges 128 and pick and place them into the respective chambers 124, a symbol scanner 108 (e.g., a barcode scanner), a consumable handling system that can include a conveyor 115 to move the consumable 602 across a deck 116 (e.g., which can include one or more plates that can be formed out of metal), and a flow control system 142. The flow control system 142 can be adjustable and can include one or more actuators 144 (e.g., pneumatic cylinders), a manifold 148 (e.g., that is in fluid communication with one or more pressure sources, such as, for example, a positive pressure source, a negative pressure source, etc,), and a manifold 146 (e.g., that is in fluid communication with a vent, a pressure source, the ambient environment, etc.). In some cases, the flow control system 142 can be enclosed in a housing 202 (shown in FIG. 2).

[0049] As shown in FIG. 1, the consumable 602 can be positioned on a belt 118, which can be supported by the deck 116. First, an empty consumable 602 can be placed on belt 118. Then, the conveyor 115 can move the empty consumable 602 to a second position in front of the symbol scanner 108 where the prefilled and/or empty cartridges 128 are scanned and loaded into the each respective chamber 124. After, the conveyor 115 moves the cartridge-loaded consumable 602 to a third position where the flow control system 142 is brought into engagement with the consumable 602 that is loaded with the empty cartridges 128. For example, the flow control system 142 can be moved (e.g., lowered) until each conduit 134 is brought into fluid communication with a respective channel 122 of the consumable 602 (e.g., each conduit 134 being in sealing engagement, or in other words, in gas-tight contact with the respective channel 122 of the consumable 602) to perform the formulation process. At this time, the actuators 144 are caused to extend (e.g., the pistons 304) to push the cartridges 128 until the cartridges 128 move further into the respective chamber 124. In some cases, the actuators 144 can push the cartridges 128 into the respective chamber 124 until a long cannula and a short cannula are positioned within a respective chamber 124 (e.g., the cannulas at the bottom of the chambers 124 pierce a septum 806 (e.g., or a barrier) of the cartridge 128),

[0050] The fluid handling system 100 can include an upper flow control system 142. In the example shown in FIG. 1 , the flow control system 142 can include the one or more actuators 144 (e.g., a pneumatic actuator, an electrical actuator, a hydraulic actuator, etc.), the manifolds 146, 148, and one or more valves 302. As shown in FIG. 1, the flow control system 142 can be configured to be moved upwardly (and downwardly), using, for example, a motor (e.g., an integrated stepper motor 502), a lead screw 138, and a nut mechanism 136. In this way. the flow r control system 142 can be lowered to bring the system 142 into contact with the consumable 602 (e.g., during a formulation process), and can be raised to bring the system 142 out of contact with the consumable 602 (e.g., when the formulation process is completed). In some configurations, then, the flow control system 142 only operates on a consumable 602 when the consumable 602 is pressed against the flow control system 142 (e.g., a plate of the flow control system 142), the actuators 144 are advanced to force the cartridges 128 into the chambers 124 of consumable 602, and the conduits 134 (e.g,, a tube can define a conduit) are fluidically coupled to the channels 122.

[0051] In some configurations, the symbol scanner 108 can scan a symbol (e.g., a QR code 130) that is attached to a cartridge 128 (e.g., a side surface of the cartridge 128). In some cases, when the symbol scanner 108 scans a symbol of the cartridge 128, the symbol scanner 108 can extract symbol information encoded therein. In some cases, the symbol scanner 108 can recognize (or in other words identify) the cartridge information or vial information encoded by a symbol on the cartridge 128 (e.g., QR-code 130), and can provide the spatial coordinates of the cartridge 128 (and others) to the robot arm 102 to allocate and capture the targeted cartridges or vials. For example, the symbol scanner 108 can identify the spatial coordinates of a cartridge 128, which can be used by the robot arm 102 to move to and grasp the cartridge 128. [0052] FIG. 2 shows an isometric view of the fluid handling system 100. As shown in FIG. 2, the flow control system 142 and the symbol scanner 108 can be located within a housing 202 (or in other words an enclosure). The robot arm 102 of the fluid handling system 100 can transfer the cartridges 128 into the chambers 124 of a consumable 602, which is placed onto, supported, and moved by the conveyor belt 118. The consumable 602 can be positioned on the conveyor 115 as the conveyor 115 moves, which can move to deliver the consumable 602 to a position in front of the symbol scanner 108. Then, the consumable 602 can be moved to a third position, which is under the flow control system 142 and inside the housing 202. In some cases, once the consumable 602 reaches the third position on the conveyor 115, the conveyor 115 can stop (temporarily), during, for example, the formulation process. After the consumable 602 has been processed (e.g., the formulation has been completed), the conveyor 115 can move the consumable 602 out of the housing 202, so that the consumable 602 may be picked up for other downstream analyses (e.g., by a different robot arm, the same robot arm, etc.). Accordingly, the consumable 602 can be disposable or reusable. For example, in the case in which the consumable 602 is reusable, after the consumable 602 has been sanitized (e.g., by autoclaving the consumable 602, exposing the consumable 602 to a sanitizing agent, such as ethylene oxide, periodide, alcohol, etc.), the consumable 602 can be placed back on the conveyor 115 and can be loaded with new cartridges. Correspondingly, the consumable 602 with the new cartridges placed therein, can be moved back into the housing 202 (e.g., under the flow control system 142) where a new formulation can be processed by the flow control system 142. In an alternative embodiment, the fluid handling system 100 can include a belt or a deck that can slide into and out of the housing 202 (e.g., similarly to a CD sliding in and out of a CD player). In this case, for example, the flow control system 142 can be positioned within a recess of the housing 202, in which the conveyor 115 is positioned outside of the recess. In this way, the consumable 602 (e.g., after the consumable 602 is loaded with one or more cartridges) can be moved off of the conveyor 115 and placed into the recess of the housing 202 so that the conveyor 115 does not have to be stopped while the flow control system 142 implements a formulation process.

[0053] FIG. 3 shows a side view cross-section of the fluid handling system 100, illustrating the placement of the consumable 602 within the housing 202 at three distinct positions on a conveyor belt 118 that is supported by the deck 116 (e.g., that can be metal) located under the belt 118. Three distinct positions, which are determined by the motion of conveyor 115 can include a first position (e.g., an initial placement) of the consumable 602 on belt 118, a second position once the cons umable 602 is in front of the symbol scanner 108 , and a third position in which the consumable 602 is positioned under a flow control system 142 (e.g., where the formulation process occurs). The symbol scanner 108 can read a QR code 130 attached on the surface of a cartridge 128, which is unique to the content and/or location of each cartridge.

[0054] FIG. 4A shows an isometric view of the flow control system 142. which can be used to facilitate the integrated, automated, aseptic, and closed aspects of the fluid handling system 100. After the consumable 602 (e.g., that is loaded with one or more cartridges 128) is positioned under the flow' control system 142 and the flow control system 142 is engaged with the consumable 602 (e.g., the consumable 602 is in contact with the flow' control system 142), the actuators 144 can be extended to force the cartridges 128 further down into the chambers 124 toward the hollow tubes 606, 608. In some cases, at this point, the conduits 134 (e.g., each of which can be defined by a tube) can sealingly engage the channels 122 to provide either positive pressure, negative pressure, or to vent gasses from (or to) the hollow tube 606 inside the cartridges 128 through one or more fluidic conduits 604 of the bottom piece 126 of the consumable 602. In some cases, an end of each conduit 134 can include a special fitting, such as an O-ring, a Luer lock connection, etc., for a secure and gas-tight engagement between each conduit 134 and each channel 122, In some cases, this can include each conduit 134 being positioned within each channel 122 to fluidically seal each conduit 134 to each channel 122, while in other cases, this can include each channel 122 being positioned within each conduit 134 to fluidically seal each conduit 134 to each channel 122,

[6055] FIG. 4B shows another isometric view of the flow control system 142 in which each actuator 144 can be in selective fluid communication with a first manifold tube 148 that is in pressure communication with a pressure source, and can be in selective fluid communication with a second fluid manifold tube 146 that is pressure communication with the ambient environment, or a different pressure source similarly to the pressure source. Correspondingly, each channel 12.2 can be in selective fluid communication with the first manifold 148 (and thus the pressure source), and can be in selective fluid communication with the manifold 146 (and thus a vent). For example, each actuator 144 and each conduit 134 can be in fluid communication with a respective valve 302 that can block or allow fluid communication (e.g., each valve 302, which can be a solenoid valve, can be actuated between multiple positions, with one position blocking fluid communication and a different position allowing fluid communication). For example, when the valve 302 that is positioned between the manifold 148 and the actuator 144 is opened, a piston 304 of the actuator 144 is forced to extend downwardly (e.g., to push the cartridge 128 into the chamber 124 of the consumable 602. Correspondingly, when this valve 302 closes, and a different valve 302 that is positioned between the manifold 146 and the actuator 144 is opened, the pressure within the actuator 144 is released and vented to atmosphere, via the manifold 146 thereby returning the piston 304 to a neutral position. As yet another example, when a valve 302 that is positioned between the manifold 148 and the conduit 134 is opened, fluid from the manifold 148 is directed through the conduit 134 (and to a cannula as described below). Correspondingly, when the valve 302 closes, and a different valve 302 that is positioned between the manifold 146 and the conduit 134 opens, fluid from the conduit 134 (and originating from the cannula) can be directed out through the manifold 146 (e.g., to atmosphere).

[0056] In some embodiments, each valve 302 may be pneumatically, hydraulically, or electrically actuated to move between different positions. In some configurations, the valves 302 coupled to conduits 134 may be actuated between different positions, which can include opening a valve 302 coupled to the manifold 148, thereby directing gas out of the cartridge 128, via the hollow tube 606. In some embodiments, a valve 302 that is coupled to the manifold 148 can be opened, which can allow (filtered) gas to be pulled into a second cartridge 128, which is fluidicaliy coupled to the first cartridge by one or more fluid channels (e.g., one or more fluidic conduits 604 of the consumable 602). Thus, a fluid can flow' between any two cartridges 128 accordingly.

[0057] FIG. 5 A shows an isometric view of the fluid handling system 100, which includes the conveyor 115. The conveyor 115 can include a belt 118, one or more rollers 114, a motor 110, a belt 112, and a ball bearing coupling 150. In some cases, a roller 114 can be rotated by the motor 110 via the belt 112, which can be coupled between the motor 11 and the ball bearing coupling 150. In some configurations, the conveyor 115 may use spindles, pulleys, gears, or other devices instead of rollers 114 to move a belt 118 and thus a consumable 602. In some cases, a belt 1 18 can slide on the deck 116, which can prevent the belt 118 from bending when a consumable is placed on the belt 118, and when the flow control system 142 comes into contact with the consumable 602.

[0058] FIG. 5B shows an isometric view' of the fluid handling system of FIG. 1. As shown in FIG. 5B, the fluid handling system 142 can move in a vertical direction, which can include moving the fluid handling system 142 downwardly when the cartridge-loaded consumable 602 is moved to the position under the flow control system 142 (e.g., to conduct a formulation process). Correspondingly, the fluid handling system 142 can be raised upwardly, after, for example, the formulation process has been completed. In some cases, the fluid handling system 142 can include a pair of stepper motors 502, a lead screw' 138, a nut 136, and a linear shaft 140 on two sides of the deck 116, which can facilitate the vertical movement of the fluid handling system 142.

[0059] FIG. 6A shows an exploded isometric view of the consumable 602, which can be used for an automated and aseptic reconstitution and/or formulation thereof. As shown in FIG. 6A, the consumable 602 can include a bottom piece 126, which can be a plate. In some cases, the bottom piece 126 can be made of a solid substrate that can be structurally planar (or can have a planar, or substantially planar region). Correspondingly, the bottom piece 126 can have at least one substantially flat upper surface. The bottom piece 126 may be fabricated from a variety of materials such as plastic, glass, or fused silica. In some embodiments, the bottom piece 126 can include one or more fluidic conduits 604 that can extend through the bottom piece 126. For example, the one or more fluidic conduits can be grooves, channels, etc., that are directed into (e.g., etched, machined, etc.) into the upper surface of bottom piece 12.6. In some cases, these fluidic conduits 604 can be enclosed (and isolated from the ambient environment) when the bottom piece 126 is coupled to a bottom surface of the top piece 106 (e.g., which can be a block of material).

[0060] The one or more fluidic conduits 604 can be formed in the upper surface of the bottom piece 126 using any fabrication method capable of creating features in the material forming the bottom piece 126, For example, when the bottom piece 126 is made of a polymer (e.g., plastic), the one or more fluidic conduits 604 can be created by injection molding or by hot embossing methods. When the bottom piece 126 is made of a silica based substrate (e.g., a glass substrate), the one or more fluidic, conduits 604 can be fabricated by a known photolithographically and subsequent etching steps into the upper surface of the bottom piece 126. In alternative embodiments, the one or more fluidic conduits 604 can be fabricated in the lower surface of the top piece 106 (e.g., in which case the bottom piece 126 can seal the one or more fluidic conduits 604), or can be fabricated in both the upper surface of the bottom piece 126 and the lower surface of the top piece 106 (e.g., in which case the top piece 106 and the bottom piece 126 can collectively define each fluidic conduit of the one or more fluidic conduits 604). In some cases, the top piece 106 can include a substantially planar bottom surface, which can engage with the substantially planar surface of the bottom piece 126. The top piece 106 can be made of different solid materials, such as, but not limited to, plastic, silicon, quartz, metal, etc.

[0061] FIG. 6B show's a schematic isometric view of the consumable 602, in which the bottom piece 126 is coupled to a botom surface of the top piece 106. As shown in FIG. 6B, the top piece 106 can include the one or more chambers 124, the one or more channels 122, and one or more chambers 132 (e.g., which can be structured in a similar manner as the chambers 124). The channels 122 can each be through holes that can extend entirely through the top piece 106, which can start from the upper surface to the lower surface of the top piece 106. The chambers 132 can each be blind holes, and can each include a hollow' tube 606, and a hollow' tube 608. In some cases, the hollow tubes 606, 608 can each be cannulas, and can each have one or more sharp tips on a distal end thereof to facilitate puncturing of a septum (or a barrier). In some cases, the hollow tube 606 can be longer than the hollow tube 608, and thus the follow'ing description will reference the hollow' tube 606 as being a long hollow' tube 606, and the hollow tube 608 as being the short hollow' tube 608. Similarly, the term cannula can be substituted as appropriate for the term hollow tube, particularly with reference to the hollow tubes 606, 608.

[0062] As shown in FIG. 6B, the long hollow tube 606 and the one or more short hollow tubes 608 can be positioned at a bottom of a chamber 124. For example, an end of each long hollo w tube 606, and an end of each short hollow tube 608 can be coupled to a bottom surface of each chamber 124. Correspondingly, each long hollow tube 606, and each short hollow tube 608 can extend from the botom surface of each chamber 124 and towards a top surface of the top piece 106. zAs shown in FIG. 6B, each long hollow tube 606, and each short hollow tube 608 can be coaxially positioned relative to each chamber 124. In addition, each chamber 124 can be longer than each of the hollow tubes 606, 608.

[0063] FIG. 6C shows an alternative embodiment of a consumable 602. in some cases, active mechanical microvalves can be integrated within the consumable 602. For example, a mechanical microvalve can be a diaphragm valve, which uses a flexible sheet, thin membrane, or diaphragm 622 (e.g., including but not limited to, a TPU diaphragm or a thin PDMS membrane) pressed close to the edge of a solid dam to narrow' (or close) the fluid path or conduit 604 and the ports 626 for fluid. In some cases, as shown in FIG. 6C-III, the fluidic conduit 604 and ports 626 can be fabricated in the upper surface of the bottom piece 126 of a consumable 602, and the conduit 632, 616, and different defined- volume pumping chambers 614 that may be required for actuating the microvalves can be fabricated in the lower surface of the top piece 106, which can be separated from upper surface of the bottom plate 126 with an intermediate flexible sheet or membrane 622. These mechanical microvalves can be controlled by positive or negative pressures via communicating with the one or more channels 122 through a set of fluidic conduits 632, 616. Thus, on-demand opening and closing of the valves as well as pumping a certain volume of a liquid, which can be defined and controlled by the volumes of the pumping chambers 614, is possible.

[0064] In an alternative embodiment, as depicted in FIG. 6C-II, the flexible sheet, thin membrane, or diaphragm 622 can be replaced by a piston rod 618 that is coupled to a blocking component 620 at the end, which can include a sealant ring and a blocking pin that fits into the fluidic conduit 604 and/or ports 626 which can provide on-demand control of the fluid flow's.

[0065] In another alternative embodiment shown in FIG. 6C-IV, an actuator 634 (e.g., that can be reusable) can be integrated to the bottom of a consumable 602 to not only provide on- demand control of closing and opening of the microvalves, but can also provide versatile, on- demand, tunable volumetric fluid pumping or fluid movement via sliding the piston up and down, thus forming a re-sizable pumping chamber 614 with a certain volume of interest. An actuator 634 can include a substrate 628 made of plastics, metals, etc., a piston 624 that has a through hole 630 at its center line which provides positive and/or negative pressures via communicating with holes, ports, vias, or chambers 122. The movement of the piston 624 may be controlled by using a precision stepper motors. This microvalving and pumping mechanism can be of great importance since it reduces the cost and complexity of a consumable 602 by providing a reusable actuator that externally acts on a flexible sheet, thin membrane, or diaphragm 622 which may be attached on the lower surface of the bottom plate 126.

[0066] In some embodiments, the pump can include a membrane (e.g., that is flexible) and that is sandwiched between two substrates (e.g., that are rigid). The bottom substrate can contain a fluidic channel, and the membrane can be positioned on top of the bottom substrate, which can include holes for the input and output of the fluidic channel . Ths top substrate can include at least 3 pneumatic connections and at least 2 liquid connections. Furthermore, the top substrate can have a cavity that allows the membrane to stretch into the cavity. A positive pressure or a vacuum can be applied to the pneumatic connections to control the fluid flow through the channel. In some cases, at ambient pressure, the pneumatic connections fluid can allow fluid to flow freely through the channel. Once the channel is filled, pressure can be applied to the left pneumatic connection, which can stretch the membrane and block the channel, creating a valve to stop or allow fluid flow. By applying a vacuum to the central pneumatic connection the flexible membrane can be expanded into the cavity in the top substrate. Next pressure can be applied to the right pneumatic connection, blocking the flow therethrough. Then, a vacuum or ambient pressure can be applied to the left pneumatic connection and by applied pressure to the central pneumatic connection the fluid is expelled out through the channel. The valves can then be reset to the initial state. Repeating the process many times can create a pumping action with a set volume. By controlling the void of the cutout in the top substrate the volume of a single displacement of the pump can be controlled.

[0067] In some embodiments, the chamber 132 can be implemented in a similar manner as the one or more chambers 124. For example, the chamber 132 can be a blind hole, and can be used as a collection chamber where the final product of the formulation is transferred to and temporarily stored. In some cases, when the lower surface of the top piece 106 is placed into contact with and coupled to the upper surface of the bottom piece 126, the one or more fluidic conduits 604 can be enclosed and can be formed. In addition, the channels 122, the chambers 124, and the chamber 132 in the top piece 106 of the consumable 602 can be aligned with and can be in fluid communication with the one or more fluidic conduits 604. For example, each hollow tube 606, 608 can be in fluid communication with a fluidic conduit 604, and each channel 122 can be in fluid communication with a fluidic conduit 604. In this way, the fluidic conduits 604 can provide external access to the chambers 124, 132 (e.g., via the hollow tubes 606, 608) to facilitate fluid transfer between the chambers 124, 132. In other words, the chambers 124, 132 can direct fluid or material introduction into (or out of) a fluidic conduit 604. Correspondingly, then, cartridges 128 positioned within respective chambers 124, 132 can provide reagents (e.g., which may be required for a formulation process) to another cartridge 128 positioned within one of the other chambers 124, 132 (and vice versa), via the application of pressure from the manifold 148, which is described in more detail below.

[0068] In some embodiments, the chambers 124, 132 can be wider than the channels 122 and can have a width (e.g., a diameter) of substantially 5 mm to 10 mm. In some cases, the channels 122 can have a width of about 1 mm to 2 mm. The one or more fluidic conduits 604 can each have a square cross-section, and can have a length between substantially 100 pm to 1000 pm. In some cases, the one or more fluidic conduits 604 can be microfluidic channels. In this way, and advantageously, smaller volumes of fluid (e.g., a liquid) can be transferred between chambers (e.g., the chambers 124) more accurately, and thus liquid can be transferred between cartridges more accurately. In some cases, the final product of the formulation can be transferred from the chamber 132 to a cartridge 128 that is loaded in the chamber 124, such that a final product of a formulation can be collected in the cartridge 128 (e.g., that is an aseptic cartridge or vial), which can then be retracted and moved around in a sterile condition.

[0069] In some configurations, active mechanical microvalves, for example, diaphragm valves, which use a flexible sheet or membrane pressed close to the edge of a solid dam to narrow or close the flow path for fluid, may be integrated with the one or more fluidic conduits 604. For example, the fluidic conduit 604 may be fabricated in the lower surface of the top block 106 of a consumable 602, and the conduit required for actuating the microvalves may be fabricated in the upper surface of the bottom piece 126, which is separated from lower surface of top piece 106 with an intermediate flexible sheet or membrane. These mechanical microvalves may be controlled by positive or negative pressures via communicating with the channels 122 through a set of fluidic conduits.

[0070] In some embodiments, the flow' control system 142 can perform a self-cleaning process. In other words, the flow control system 142 may be capable of flushing the contaminated surfaces in a consumable 602 to avoid cross-contamination. Thus, a consumable 602 can be able to handle a variety of materials at a time considering appropriate washing between each material or processing steps.

[0071] FIG. 7A, 7B, and 7C show isometric cross-section views of the consumable 602. As shown in FIG. 7 A, each chamber 124 in the top piece 106 of the consumable 602 may include a long hollow tube 606 close to the lower surface of the block 106, which communicates with a pressure source, via a channel 122 and one or more fluidic conduits 604. In some embodiments, each chamber 132 can include one or more short hollow tubes 608, through which liquids can be moved between any two cartridges 128 that are connected through the one or more fluidic conduits 604. In other words, the long hollow tubes 606 can introduce (or remove) gas from the cartridges 128 by communicating with the channels 122 through the fluidic conduits 604, while short hollow tubes 608 can be used to move liquids between cartridges 128 loaded in the chambers 124 or between a cartridge 128 (or a chamber 132), for example, to move the final product of a formulation to the chamber 132. In some embodiments, the chamber 132 can include a chimney structure 610, through which the final product may be pulled into the chamber, and a drain hole 612, through which a final product of a formulation may be pushed out for any other downstream use (e.g., for further processing in another liquid path).

[0072] In some embodiments, the consumable 602 can be aseptic, and can facilitate the aseptic transfer of fluids between respective cartridges 128. For example, although not shown in FIG. 7A-7C, the consumable 602 can include one or more barriers (e.g., a septum), each of which can extend across a respective chamber 124, 132. In this way, the barrier can isolate the chamber from the ambient environment to prevent contamination therein by the ambient environment. In some cases, each barrier can be positioned above the hollow tubes 606, 608. In some cases, when a cartridge 128 is inserted into a chamber 124, 132, the barrier can rupture to allow the cartridge 128 to continue moving further into the chamber 124, 132.

[0073] In some embodiments, the consumable 602 being aseptic can include a septum 133 extending across the chamber 132 to isolate the chamber 132, including the needle 610, and the port 612 from the ambient environment (e.g., to maintain an aseptic environment within the chamber 132). In this way, a needle (e.g., of a vacuum tube) can penetrate the septum 133 to enter into the chamber 132 to collect a liquid (e.g., a final formulation) positioned within the chamber 132. In other cases, the septum 133 can be ruptured when a cartridge is loaded into the chamber 132 (e.g., until the barrier of the cartridge is pierced by the needle 610). In some configurations, the liquid positioned within the chamber 132 can be transferred out of the chamber 132 through the port 612.

[0074] In some embodiments, while the fluid handling system 100 has been described as having multiple actuators 144, with each actuator 144 being individually moveable to insert a cartridge 128 into a chamber 124, 132, in other configurations, one actuator (e.g., an actuator 144) can drive multiple cartridges 128 into multiple respective chambers 124, 132, For example, an actuator can include multiple pistons that can be actuated together, in which case, the actuator can extend the multiple pistons to contact and drive each cartridge 128 into each chamber 124, 132. In some cases, a piston (e.g., or extender) of an actuator having can drive multiple cartridges into the respective chambers 124, 132 simultaneously.

[0075] FIG. 8A shows an isometric partial cross-section view of an exemplary embodiment of the present disclosure, illustrating the mechanism of the flow control system 142, which includes actuation of, for example, actuators 144 as well as engagement of cartridges 128 and hollow lubes 606 and 608 in the respective chambers 124. An actuator 144 may be extended (e.g., by opening a valve 302 to introduce pressurized gas from the manifold 148 to the actuator 144). Consequently, when the actuator 144 extends, a piston 304 of the actuator 144 moves downwardly to push the cartridge 128 further into the chamber 124, and compresses a spring 808 (e.g., a high tensile spring). As the cartridge 128 is pushed further into the chamber 124 towards the hollow tubes 606, 608, a spring 810 (e.g., a high tensile spring) that is positioned within the chamber 124 (e.g., coaxially surrounding the hollow tubes 606, 608) compresses, and each hollow tube 606, 608 pierces the septum 806 of the cartridge 128 until each hollow tube 606, 608 enters an interior volume of the cartridge 128 (e.g. that is isolated from the ambient environment). In some cases, to achieve maximum liquid transfer from or to a cartridge 128, the length of a short hollow 7 tube 608 may be equivalent to the length or thickness of a fully compressed spring 810 plus the length or thickness of the septum 806. In some configurations, the short hollow 7 tubes 608 may have perforated tips, making sure that almost all the liquid is pulled out of a cartridge 128 even if the hollow tube length is not exactly equivalent to the length or thickness of a fully compressed spring 810 plus the length or thickness of a septum 806, thus minimizing any dead volume left inside a cartridge 128.

[0076] FIG. 8B shows a schematic cross-section view of an exemplary embodiment of the present disclosure. For example, FIG. 8B shows that the system can allow for and facilitate controlled and sequential actuation of each actuator 144, and thus the controlled and sequential advancement of each cartridge 128 into engagement with each hollow 7 tube 606, 608 (and the one or more fluidic conduits 604). As shown in FIG. 8B, each actuator 144 can be independently actuated by opening a first valve 302, which is coupled between a manifold 148 (e.g., that is in fluid communication with a pressure source) and the target actuator 144. Once the actuator 144 is pressurized, the piston 304 begins to be forced down into and along the chamber 124 while compressing the spring 802. As the piston 304 moves the cartridge 128 down into the chamber 124, a spring 804 (e.g., a high tensile spring) compresses the spring 804 until the spring 804 is fully compressed and the hollow 7 tubes 606, 608 pierce the septum 806 of the cartridge 128. After a formulation process is complete, a second valve 302, which is connected to the same target actuator 144 and is positioned between the same target actuator 144 and the manifold 146 (e.g., in fluid communication with the ambient environment), can be opened while the first valve 302 is closed to release the pressure from the actuator 144, so that the piston 304 is retreated back to an initial position by unloading of the spring 808. In some cases, a piston 304 can include a load cell 812 (e.g., that can be thin, planar, etc.) that can be coupled to a bottom surface of the piston 304. In some cases, the load cell 812 can control the amount of pressure or force required to safely push a cartridge 128 down, for example, until the load or pressure reaches a threshold set value, preventing damage to a cartridge 128 as well as a consumable 602. In some configurations, all the actuators 144 can be actuated simultaneously (e.g., with each actuator 144 driving a respective cartridge 128 into a respective chamber 124).

[0077] FIG. 9 A shows a schematic cross-section view of an exemplary embodiment of the present disclosure, illustrating the mechanisms of liquid transfer between cartridges. A formulation and/or reconstitution process can include the movement of material in a controlled manner between cartridges 128 that are positioned within respective chambers 124, 132 through one or more fluidic conduits 604 arranged in the consumable 602. In some cases, the material can be in liquid (e.g., concentrated), lyophilized, dehydrated, dry powder, etc., forms. Liquids can be moved within the one or more fluidic conduits 604 in different ways. For example, as shown in FIG. 9 A, fluid movement may be controlled by introducing positive or negative partial pressure to certain cartridges 128A and 128B in a consumable 602, so that fluid moves from a high-pressure location to a low-pressure location through a fluidic conduit 604 that connects those two target locations. As another example, after hollow tubes 606, 608 pierce the septum 806, a negative pressure can be applied to a long hollow tube 606 inside the cartridge 128 B as shown with arrow 904, by connecting the end of the long hollow tube 606 to a channel 122 that provides a negative pressure through a fluidic conduit 604. Similarly, the long hollow tube 606 inside the cartridge 128 A can be connected to a channel 122 that provides a vent through another fluidic conduit 604, so that filtered air can enter the cartridge 128 A as shown with arrow 902. As a result of the pressure difference between these two cartridges, fluid is moved from cartridge 128 A to cartridge 128 B in a direction shown with arrow 906.

[0078] In other configurations, the same pressure difference can be produced to move the fluid by applying positive pressure to the long hollow tube 606 inside the cartridge 128 A through a fluidic conduit 604 and a channel 122, while connecting the long hollow tube 606 inside the cartridge 128 B to a vent source through another fluidic conduit communicating with a channel 122, so that trapped gas (e.g., air) inside cartridge 128 B can be pushed out.

[0079] In some configurations, the driving force (e.g., the pressure difference) can be enhanced by simultaneously applying positive pressure to the long hollow tube 606 inside the cartridge 128 A, and a negative pressure to the long hollow tube 606 inside the cartridge 128 B to move the liquid from cartridge 128 A to 128B. In other configurations, fluids can be moved from cartridges 128 to chamber 132, vice versa.

[0080] An alternative embodiment of a fluid transfer mechanism of the present disclosure is shown in FIG. 9B and 9C. This alternative design can work with cartridges 128 that can be pierceable (e.g., made of plastic) with a septum 806 as a cap and/or with cartridges 128 made of glass, however, with a septum 806 as a cap together with a rear pierceable plunger or stopper 914 (or another septum) as shown in FIG. 9C, which may be made of, but not limited to, silicone rubber. This alternative fluid transfer mechanism can eliminate the need for long hollow tubes 606 in a consumable 602, which may limit the use of cartridges with arbitrary heights. In other words, the long hollow tubes 606 and their functionalities can be replaced by stiff stainless steel perforated closed trocar point hollow' tube 912, which may be connected to filters, such as, but not limited to, 0.2 pm PTFE filters, and then to the positive/negative or vent sources. Therefore, the flow control system 142 can include the capabilities for providing and controlling forces like pressure gradient between any two cartridges 128 and/or between a cartridge 128 and chamber 132 to move fluids through fluidic conduit 604.

[0081] In another alternative embodiment depicted in FIG. 9D, liquid between any two cartridges 128A and 128B can be transferred in a consumable 602 which can include only one long needle 606 placed at the center of a guide cannula 916. A guide cannula 916 can have more than one cuts or openings 918 on its surface, which provide a path for the liquids to move in or out of cartridges 128 that are connected through fluidic conduits 604 and 920. In this embodiment, more importantly, the use of two short needles 608 is eliminated and they are replaced by fluidic conduits 920 directly formed in the top piece 106 of a consumable 602. For example, by applying negative pressure as shown by arrow 904 to the long needle 606 inside cartridge 128 B, and applying positive pressure or vent as depicted by arrow 902 to the long needle 606 inside cartridge 128A, liquid is transferred from the cartridge 1.28A to the cartridge 128B flowing through the gap between the long needle 606 and the guide cannula 916 and the fluidic conduits 920 and 604.

[0082] As shown in FIG. 9D, each chamber of the consumable of FIG. 9D can include a first needle and a second needle, with the second needle coaxially surrounding the first, needle. The first needle can be longer than the second needle, and the second needle including the central lumen can include one or more holes. In this way, gas can be introduced into (or vacated from) the first needle to introduce gas (or remove gas from) the interior volume f the cartridge 128 A. Correspondingly, liquid can be introduced into (or removed from) the interior volume of the cartridge 128A, via the central lumen or the one or more holes of the second needle.

[0083 ] In general, the redissolution process of dry, lyophilized, or dehydrated reagents and compounds can be limited by two main factors. First, the physical properties and chemical structure of the reagents and compounds, and second, the long redissolution time required in most used, passive, diffusion- based mixing methods. The first limitation, which is a concern shared between any other reconstitution and or formulation methods and systems, and in fact depends on the methods and processes producing the reagents and compounds, may be beyond the scope of the present disclosure. Accordingly, the long diffusion time is one of the main factors addressed by the present disclosure.

[0084] Consider the example of small molecule adsorbing from an infinite solution to a freshly formed planar fluid-solid interface. There is an implicit length scale in this problem, which is characterized by an adsorption depth h, which is the depth depleted to supply the interface with the diffusing small molecule. From a simple mass balance, h can be shown to be the ratio of the equilibrium surface concentration T eq to the bulk concentration C°°. The characteristic time scale for diffusion to the interface is thus: tD ::: h2/D, where D is the diffusivity of the small molecule in solution. The significance of this timescale is demonstrated by solving the equations governing diffusion-controlled adsorption to a planar interface (from the Navier-Stokes equation). The timescale for equilibrium is in fact the same timescale for mixing as a consequence of the diffusion driving the mixing of the small molecules in solution. Of interest to note here is that the timescale is proportional to the square of the adsorption depth, h. Thus, since the diffusion coefficient is a non-changing quantity, the fluidic system can be mixed faster (due to diffusion) when this length scale, h is small. When h is larger, since the timescale is proportional to the square of this length scale - it can slow down the progress of mixing the fluidic system. [0(385] Using exi sting methods, if one were to use reagents in microtiter plates, larger vials, etc., the mixing process would be significantly more time-consuming. For example, if it is assumed that one could use a similar excipient combination and the only limitation in resolubilization was passive diffusional mixing and a typical volume used in a 384 well assay plate is 20 μL, which occupies approximately 4 mm (4000 pm) of well fluid height, for the reagent to mix through this volume, approximately 8 hours would be required in the absence of an external mixing mechanism such as stirring (which provides convective mixing) or incubation (which provides heat induced mixing). Although, in a microplate-based system some alternative external plate mixing technique and/or device could be employed, however, complications could arise from having to seal the plate to avoid cross-contamination, transfer the plate to a suitable device and then transfer the plate again to an unsealing device, a possible centrifugation step, followed by an incubation step and then to subsequently unseal the plate to access the mixed reagents. In other words, these types of methods and/or processes may clinically be considered as open-system processing, which is typically not acceptable for, but not limited to, automated cell therapy manufacturing processes (e.g., in which the product is cells).

[0086] In some embodiments, the fluid handling system 100 can provide much faster mixing of reagents. For example, the fluid handling system 100 can provide a truly miniaturized and fully integrated system with chemistry and biology at the micro and nano- liter scale, in which the time required for mixing is almost instantaneous once dried, dehydrated, or lyophilized reagents are reconstituted in liquid, dissolved in liquid, or diluted in liquid. In some cases, these compounds or reagents can be positioned within a cartridge 128, or one of the fluidic conduits 604. In some configurations, these compounds or reagents can reside in a layer (e.g., a. “pancake” layer) that can be only tens to hundreds of microns in height. Given the assumption that diffusion alone is the driving mechanism for mixing - the reagent or compound can be solubilized in only a few seconds. As an example, if the dried/ lyophilized reagent occupied an area of 400 x 400 pm and was 1 pm in height creating a “pancake” layer on a surface, the time required to resolubilize the dissolved reagent is instantaneous. [0087] In this some embodiments, and importantly, the fluid handling system 100 has the added capability to internally and aseptically move the reagents to be mixed between cartridges 128, and chambers 132, back and forth, to drive convective mixing without using any external agitators, simulating a pipetting-like mixing mechanism, which differentiates the present disclosure from any other existing liquid handlers or formuiators.

[(1088] In some embodiments, the fluid handling system 100 may also be integrated with one or more external agitators to further enhance the mixing such that the mixing time is significantly reduced. For example, an ultrasonic piezoelectric transducer can be coupled to the bottom of the consumable 602 to drive mixing-based processes utilizing ultrasound or other frequencies that can generate mixing pressure waves. This type of “pressure driver” can be ideal as it can be integrated with no physical contact with the sample, is automated, and is also uniquely positioned for aseptic operations.

[0089] FIG. 10 A show's a schematic side view of a cartridge retraction mechanism of the present disclosure, The retraction mechanism may only be required if either ah or some of the cartridges or vials 128 need to be retracted from the hollow' tubes 606 and/or 608 during or after the fluid transfer or formulation process, or a consumable 602 is reused after going through a decontaminating process. As shown in FIG. 10A, a retraction system can include the spring 804 w'hich is positioned into a chambers 124 in the top piece 106 of the consumable 602 before loading cartridges 128. After actuation of an actuator 144, a piston 304 can push down a cartridge 128 inside a chamber 124 that in turn fully (or partially) compresses the spring 804, and that forces the hollow tubes 606, 608 to pierce the septum 806 (see e.g., step 1 to II of FIG. 10A). To retract the cartridge 128, the actuator 144 can be first vented, so the piston 304 is pulled back into the cylinder, and correspondingly the spring 804 that is in a compressed configuration 810 is unloaded to force the cartridge 128 out of the chamber 124. Thus, the cartridge can be retracted by unl oading of the compressed spring 804 that is compressed in the longitudinal or vertical direction (see e.g., step II to III of FIG. 10A).

[0090] In some configurations, the height of the consumable 602 may be short enough, for example, slightly shorter than the height of cartridges 128 when the septum 806 of the cartridge 128 contacts the bottom surface of the chamber 124, such that there is no need for the spring 804 inside the chamber 124 for retracting the cartridge 128 since a portion, for example, one- fifth of the height of the cartridge 124 is outside of the chambers 124 in a consumable 602. Thus, cartridges 128 may be pulled out, for example, by grabbing the exposed part of them using a robotic arm (e.g., fingers of the robot ami).

[9091] In some configurations, such as the configuration of FIG. 10B, a cartridge retraction system may include a pin 1002 (e.g., a u-shaped pin that can be metal), which may be initially pushed into the chamber 124 through two through-holes 1004 in the consumable 602 when the consumable 602 reaches the position under the flow control system 142 (e.g., for formulation processing). When an actuator 144 is actuated or pressurized, the piston 304 can move down the cartridge 124 and the pin 1002 against the spring 804 until the hollow tubes 606, 608 pierce the septum 806 and the septum 806 comes into contact with the bottom surface of the chamber 124 (see, e.g., step I to II of FIG. 10B). To retract the cartridge 128, the actuator 144 can be first vented thereby removing the force on the cartridge 128, thereby pulling the piston 304 back into the cylinder of the actuator 144. At this point, the spring 804 in a compressed state 810 unloads, thereby pushing the pin 1002 upward, resulting in retraction of the cartridge 128 (see, e.g., step II to HI of FIG. 10B). In some cases, one of the key advantages of the embodiment shown in FIG. 10B (e.g. over the one sho wn in FIG. 10A) is the location of the springs 804. For example, in the embodiment shown in FIG. 10B, the cartridge retraction system that incorporates the springs 804 can advantageously be part, of the instrument or fluid handling system 100, but is not a part of the consumable 602, which prevents the need for the consumables having springs 804, which can save cost, manufacturing time and resources (e.g., prevents needing to load each spring 804 into each chamber 124 for every consumable 602), etc., for the consumable 602.

[0092] In some embodiments, the part 1006 (e.g., a base) may be electromagnetically actuated, such that the pin 1002 is magnetically attracted and coupled to the part 1006 (or magnetically repelled and forced away from the part 1006), when the pin 1002 comes in a certain proximity to part 1006. Thus, the release of the pin 1002 can be controlled and activated either manually by users or in an automated way, for example, after completion of a formulation process at a different time than the time when the force applied by the piston 304 on the cartridge 128 is removed. In other words, this electromagnetic activation of cartridge retraction may provide a manual or automated control on the retraction of cartridges in addition to the prior control mechanism on cartridge retraction that was described in FIG. 10A and 10B (e.g., the capability of the system that allows actuation of the actuators 144 and pistons 304 independently, thus retraction of cartridges 128 could occur in a controlled manner).

[0093] FIG. 1 1A and 1 IB show schematic top views of an exemplary sampling or aliquoting mechanism (e.g., a QC sampling or aliquoting mechanism). As shown in FIG. 11 A and 1 IB, the consumable 602 may include quality control (QC) and/or aliquoting capabilities for the final product of a formulation process. FIG. 11 A shows a tree-like network design for the one or more fluidic conduits 604. The mechanism of the fluid transfer through the fluidic conduits 604 between cartridges 128 and/or chambers 132 is similar to what has been described above. For example, the final formulation product may be stored in chamber 132 in a consumable 602. A certain volume of the product can be transferred from the chamber 132 to cartridges 128 by applying negative pressure to long hollow tubes 606 inside the QC sampling or aliquoting cartridges 128 through a manifold 1102, one or more fluidic conduits 604, then one or more channels 122, and by simultaneously either venting or applying positive pressure to the chamber 132 via conduits 134 inserted into the chamber 132 from the top surface of the consumable 602.

[0094] Similarly, in an alternative embodiment of a sampling or aliquoting mechanism shown in FIG, 1 IB (e.g., another QC sampling or aliquoting mechanism), the mechanism 1100B may include a disk-like or radial network design for fluidic conduit 604 to equally split or aliquot the final product of a formulation process into aseptic cartridges 128. For example, a certain volume of the product can be transferred from the chamber 132 to QC or aliquot cartridges 128 by applying negative pressure to long hollow tubes 606 inside the cartridges 128 through a manifold 1102, the one or more fluidic conduits 604, then channels 122, and by simultaneously either venting or applying positive pressure to the chamber 132 via conduits 134 inserted into the chamber 132 from the top surface of the consumable 602.

[0095] In some embodiments, the barriers and the septums described herein can be resealable. For example, after a hollow tube 608 pierces the septum 806 of a cartridge 128 (e.g., and creates a hole in the septum 806), after the hollow' tube 608 is retreated out from the septum, the septum can reseal (e.g., the edges that define the hole created by the hollow tube 608 can be forced into contact with each other). In this way, the septum 806 can reseal the interior volume of the cartridge 128 from the ambient environment, to, for example, maintain an aseptic contents therein.

[0096] In some embodiments, any of the fluid handling devices herein (e.g., the fluid handling system 100) can include a computing device (e.g., a controller, processor, etc.), which can implement some or all of the processes herein as appropriate. For example, the computing device can cause the actuators 144 to extend (and retract), can cause the symbol scanner 108 to acquire symbol information from a symbol on a cartridge 128, etc.

EXAMPLES

[0097] The following examples have been presented in order to further illustrate aspects of the disclosure and are not meant to limit the scope of the disclosure in any way. The examples below' are intended to be examples of the present disclosure and these (and other aspects of the disclosure) are not to be bounded by theory.

EXAMPLE 1

[0098] One example, as a proof-of-concept study, will be specified below, showing how fluid transfer and dissolution of a dry powder can be carried out using the exemplary embodiment of a consumable and mechanisms of fluid transfer shown in FIGS. 6, 7, and 9.

[0099] Reference for an exemplary consumable is made to FIG. 6. As shown in FIG. 12A, for simplicity, an exemplary consumable was designed and fabricated such that the top piece 106 of a consumable 602 was replaced with a flat plate 1202 made of polycarbonate such that both short 608 and long 606 hollow tubes with 0.5 mm inner and 1 mm outer diameters were fitted in designated through-holes, which were later carefully aligned with fluidic conduit 604. For the same reason, the channels 122 were replaced with Luer lock connectors 1204, which communicate with negative and or positive pressure sources to control the fluid flow through fluidic conduit 608.

[00190] Enclosed fluidic conduit 604 is shown in FIG. 12A was fabricated by sandwiching about 75 pm thick double-sided adhesive as a channel layer between a bottom polycarbonate plate 1206 and a top polycarbonate plate 1202. Channels of the fluidic conduit were fabricated by cutting out the fluidic conduit design in the double-sided adhesive using a CO2 laser cutter (e.g. Universal Laser VLS 3,5). As shown in FIG. 12B, plastic cartridges 128 with 300 μL internal volumes were pressed on the hollow tubes until the tips of short hollow tubes 608 were exposed into the cartridges. To achieve this, the lengths of the short hollow tubes exposed out of the upper surface of the top plate 1202 were equivalent to the thickness of the septum in caps 1210 of the cartridges. A ll the Luer lock connectors 1204 were initially closed using Luer lock blockers 1208.

[00101 ] In the present working example as shown in FIG. 13A, only the fluid transfer between two cartridges 128A and 128B are illustrated. Cartridge 128 A was filled with 200 μL of distil led water, and about Img of Bromophenol Blue powder was placed in cartridge 128B, which turns to red color when it is dissolved in water. First, distilled water was moved from cartridge 128A to cartridge 128B by pulling out the air from cartridge 128B via connecting the long hollow tube 606 inside the cartridge 128B to a Luer lock connector 1204B through fluidic conduit 604. The Luer lock connector 120413 was connected to a vacuum source (e.g., syringe pump 1216) through the integration of a silicon tube 1212, a syringe tip 1214, and a I mL syringe 1216. The Luer lock connector 1204A was open to air such that air could be pulled into cartridge 128 A by connecting the long hollow tube 606 inside cartridge 128zA to lock connector 1204A through the fluidic conduit.

[00102] FIGS. 13B and 13C show the two cartridges, respectively, before and after water transfer and dissolution of the powder. As is evident, water was moved from cartridge 128A to 128B, which was confirmed by the accumulation of the red color liquid in cartridge 128B as shown in the inset of FIG. 13C.

[00103 [ To further enhance the dissolution of the powder, similar to the repetitive pipetting effect, liquid can be moved back and forth between the two cartridges only by switching the vacuum and vent sources on the Luer lock connectors 1204A and 1204B. For example, after the first water transfer, the new red solution was transferred back from cartridge 128B to 128A by applying vacuum source to Luer lock connector 1204 A, which is connected to the long hollow tube 606 inside cartridge 128A, and venting the Luer lock connector 1204B, which is connected to long hollow' tube 606 inside cartridge 128B through fluidic conduit 604. As illustrated in FIG. 13D, for better visualization of the contents of each cartridge, both cartridges were separated from their caps. It is shown in FIG. 13D that the red solution was moved back to cartridge 128 A, which was spread in the cap 1210A after removing the cartridge. This process may be repeated multiple times depending on the dissolution rate of different powders in different liquids or buffers to achieve efficient mixing.

[00104] In an alternative embodiment, an active mixing mechanism may be designed and integrated with the present disclosure to further enhance the dissolution and/or reconstitution of lyophilized materials or dry powders. For example, an ultrasonic agitation system may be integrated to system 100 under belt 118 or under the deck 116 where a consumable 602 is positioned under the flow control system 142.

[00105] In some- embodiments, the systems herein (e.g., the fluid handling system 100) including components of the systems (e.g., the consumable 602) can operate under aseptic conditions (e.g., positioned in an ISO-level sterile environment) during parts or the entire formulation process. For example, the systems herein and the components of the systems herein can be operate in a sterile environment (e.g., a sterile enclosure, an aseptic enclosure, etc,). For example, as described above, each chamber of the top piece 106 of the consumable 602 can include a frangible barrier (e.g., a sheet of a polymer, a sheet of a plastic, etc.) that can temporarily isolate the chamber and the one or more needles positioned therein from the ambient environment. In this way, when the cartridge is loaded in a chamber of the top piece 106, the frangible barrier can be broken to ensure that the chamber is aseptic and free of contaminants (e.g.. that may otherwise undesirably leach into the one or more channels of the bottom piece 126 or into a cartridge loaded in a chamber). As another example, each needle of each chamber of the top piece 106 of the consumable 602 can include a barrier (e.g., a sleeve) that is positioned over the needle and that isolates the corresponding lumen of the needle from the ambient environment (e.g., to ensure an aseptic environment within the needle). In this way, when the cartridge is positioned in the chamber with the needle having a barrier and the cartridge is advanced towards the needle with the actuator, the needle pierces through the barrier coupled to the needle (e.g., surrounding the needle) and pierces the barrier of the cartridge to extend into the interior volume of the cartridge. In this way, the needles can prevent contamination and maintain aseptic connections (e.g., liquid is only transferred after the barrier coupled to the needle is pierced). Correspondingly, the processes herein can be implemented under aseptic conditions. EXAMPLE 2

[00106] Another example will be specified below, demonstrating how reagents’ formulation for CRISPR genome editing can be carried out using the exemplary embodiments of the fluid handling system 100 shown in FIGS. 1 and 2 of the present disclosure.

[00107] Reference is made to FIG. 14. FIG. 14 shows the same exemplary embodiment for the consumable 602 as FIG. 6B, just with different names for each chamber 124 in which 8 cartridges with different contents and QR codes can be loaded. In the present case, materials or reagents in cartridges that can be loaded in channels 122a to 124h are Tris EDTA (TE) buffer, lyophilized guide RNA (gRNA), no content (e.g., empty), TE buffer, lyophilized donor templates (ssODNs), no content, Cas9 protein, and distilled water (dH2O), respectively.

[00108] In summary, the formulation process includes first reconstitution of both lyophilized gRNA and ssODNs, second, efficient mixing of the reconstituted gRNA, ssODNs, and Cas9, third, complex setting/formation (about 10 min), and finally mixing of the gRNA- ssODNs-Cas9 complex with dH2O, which forms the final product of the formulation.

[00109] As described in embodiments of the present disclosure, first a consumable 602 shown in FIG. 14A can be placed in the first position on belt 118. Then, conveyor 115 can move the consumable to the second position in front of the symbol scanner where cartridges with unique QR codes, specifying their content and or location, can be loaded into chambers 124a to 124h using a robot arm 102 and fingers 104. After that, conveyor 115 can move the cartridge-loaded consumable to the third position (e.g., for a formulation process) under the flow control system 142. The flow control system 142 can come into hard contact with the cartridge-loaded consumable where the pistons 304 of the actuators 144 are aligned with a respective chamber 124a to 124h, and a pressure source or a vent engages with the channels 122.

[00110] The formulation can be started by simultaneous actuation of actuators 144 such that all pistons 304 push down the cartridges until hollow tubes 606, 608 pierce the septum 806 on each of the cartridges. When all the cartridges are engaged, the system is ready to transfer fluids between any pair of cartridges, or between a cartridge and the chamber 132 in the consumable 602, via the one or more fluidic conduits 604. A top view of the fluidic conduit design used for the present example is shown in FIG. 14B. [00111 ] Each long hollow tubes 606 can communicate with a channels 122 through certain fluidic conduits, thus controlling flows by generating pressure differences between cartridges, or chambers. For a better understanding of the formulation process and methods of operations, the rest of the descriptions are presented in the form of steps below”

[601121 Step 1 : A pressure gradient or pressure difference can be generated between the cartridges to move the TE buffer from a first cartridge to a second cartridge where reconstitution of lyophilized gRNA occurs. As described in relation to working example 1, the reconstituted solution can be repeatedly and gently transferred between these two cartridges to achieve an efficient mixing or dissolution of the lyophilized gRNA;

[00113] Step 2: The well-mixed reconstituted gRNA can be moved to a third cartridge that is empty, similarly by generating a pressure gradient between the second cartridge and the third cartridge.

[00114] Step 3: TE buffer can be moved from a fourth cartridge to a fifth cartridge to reconstitute the lyophilized ssODNs. The reconstituted solution can be repeatedly transferred between these two cartridges to achieve sufficient mixing:

[00115] Step 4: The well-mixed reconstituted ssODNs can be moved to the sixth cartridge that is empty by generating a pressure gradient between the fifth cartridge and the sixth cartridge;

[00116] Step 5: Both reconstituted gRNA and ssODNs can be transferred to the Cas9 cartridge either one at a time or together simultaneously depending on how the pressure gradient is generated between Cas9 cartridge and the other two, e.g., the third cartridge and the sixth cartridge;

[00117] Step 6: Incubate at room temperature for about 10 min for Cas9-gRNA-ssODNs complex formation;

[00118] Step 7 : The Cas9-gRNA-ssODNs complex can be transferred to the dH2O cartridge by creating a pressure gradient between the seventh cartridge and the eighth cartridge (e.g., the dH2O cartridge).;

[00119] Step 8: Lastly, the final mixture can pass through a serpentine (zig-zag, curved, etc.) fluidic conduit to ensure efficient mixing of the complex and dH2O while transferring from the eighth cartridge to the final destination, which can be the chamber 132 in the consumable 602 or a larger aseptic cartridge. The formulation can now be ready for use. [00120] The present disclosure has described one or more preferred claims, and it should be appreciated that many equivalents, alternatives, variations, and modifications, aside from those expressly stated, are possible and within the scope of the disclosure.

[00121] It is to be understood that the disclosure, is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the following drawings. The disclosure is capable of other claims and of being practiced or of being carried out in various ways. Also, it is to be understood that the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of “including,” “comprising,” or “having” and variations thereof herein is meant to encompass the items listed thereafter and equivalents thereof as well as additional items. Unless specified or limited otherwise, the terms “mounted,” “connected,” “supported,” and “coupled” and variations thereof are used broadly and encompass both direct and indirect mountings, connections, supports, and couplings. Further, “connected” and “coupled” are not restricted to physical or mechanical connections or couplings.

[00122] As used herein, unless otherwise limited or defined, discussion of particular directions is provided by example only, with regard to particular claims or relevant illustrations. For example, discussion of “top,” “front,” or “back” features is generally intended as a description only of the orientation of such features relative to a reference frame of a particular example or illustration. Correspondingly, for example, a “top” feature may sometimes be disposed below a “bottom” feature (and so on), in some arrangements or claims. Further, references to particular rotational or other movements (e.g., counterclockwise rotation) is generally intended as a description only of movement relative a reference frame of a particular example of illustration.

[00123] In some claims, aspects of the disclosure, including computerized configurations of methods according to the disclosure, can be implemented as a system, method, apparatus, or article of manufacture using standard programming or engineering techniques to produce software, firmware, hardware, or any combination thereof to control a processor device (e.g., a serial or parallel general purpose or specialized processor chip, a single- or multi-core chip, a microprocessor, a field programmable gate array, any variety of combinations of a control unit, arithmetic logic unit, and processor register, and so on), a computer (e.g., a processor device operatively coupled to a memory), or another electronically operated controller to implement aspects detailed herein. Accordingly, for example, claims of the disclosure can be implemented as a set of instructions, tangibly embodied on a non-transitory computer-readable media, such that a processor device can implement the instructions based upon reading the instructions from the computer-readable media. Some claims of the disclosure can include (or utilize) a control device such as an automation device, a special purpose or general purpose computer including various computer hardware, software, firmware, and so on, consistent with the discussion below. As specific examples, a control device can include a processor, a microcontroller, a field-programmable gate array, a programmable logic controller, logic gates etc., and other typical components that are known in the art for configuration of appropriate functionality (e.g., memory, communication systems, power sources, user interfaces and other inputs, etc.).

[00124] fhe term “article of manufacture” as used herein is intended to encompass a computer program accessible from any computer-readable device, carrier (e.g., non-transitory signals), or media (e.g., non-transitory' media). For example, computer-readable media can include but are not limited to magnetic storage devices (e.g., hard disk, floppy disk, magnetic strips, and so on), optical disks (e.g., compact disk (CD), digital versatile disk (DVD), and so on), smart cards, and flash memory devices (e.g., card, stick, and so on). Additionally it should be appreciated that a carrier wave can be employed to carry computer-readable electronic data such as those used in transmitting and receiving electronic mail or in accessing a network such as the Internet or a local area network (LAN). Those skilled in the art will recognize that many modifications may be made to these configurations without departing from the scope or spirit of the claimed subject matter.

[00125] Certain operations of methods according to the disclosure, or of systems executing those methods, may be represented schematically in the FIGS, or otherwise discussed herein. Unless otherwise specified or limited, representation in the FIGS, of particular operations in particular spatial order may not necessarily require those operations to be executed in a particular sequence corresponding to the particular spatial order. Correspondingly, certain operations represented in the FIGS., or otherwise disclosed herein, can be executed in different orders than are expressly illustrated or described, as appropriate for particular claims of the disclosure. Further, in some claims, certain operations can be executed in parallel, including by dedicated parallel processing devices, or separate computing devices configured to interoperate as part of a large system.

[00126] As used herein in the context of computer configuration, unless otherwise specified or limited, the terms “component,” “system,” “module,” and the like are intended to encompass part, or all of computer-related systems that include hardware, software, a combination of hardware and software, or software in execution. For example, a component may be, but is not limited to being, a processor device, a process being executed (or executable) by a processor device, an object, an executable, a thread of execution, a computer program, or a computer. By way of illustration, both an application running on a computer and the computer can be a component. One or more components (or system, module, and so on) may reside within a process or thread of execution, may be localized on one computer, may be distributed between two or more computers or other processor devices, or may be included within another component (or system, module, and so on).

[00127] In some configurations, devices or systems disclosed herein can be utilized or installed using methods embodying aspects of the disclosure. Correspondingly, description herein of particular features, capabilities, or intended purposes of a device or system is generally intended to inherently include disclosure of a method of using such features for the intended purposes, a method of implementing such capabilities, and a method of installing disclosed (or otherwise known) components to support these purposes or capabilities. Similarly, unless otherwise indicated or limited, discussion herein of any method of manufacturing or using a particular device or system, including installing the device or system, is intended to inherently include disclosure, as claims of the disclosure, of the utilized features and implemented capabilities of such device or system.

[00128] As used herein, unless otherwise defined or limited, ordinal numbers are used herein for convenience of reference based generally on the order in which particular components are presented for the relevant part of the disclosure. In this regard, for example, designations such as “first,” “second,” etc., generally indicate only the order in which the relevant component is introduced for discussion and generally do not indicate or require a particular spatial arrangement, functional or structural primacy or order.

[00129] As used herein, unless otherwise defined or limited, directional terms are used for convenience of reference for discussion of particular figures or examples. For example, references to downward (or other) directions or top (or other) positions may be used to discuss aspects of a particular example or figure, but do not necessarily require similar orientation or geometry in ah installations or configurations.

[00130] Also as used herein, unless otherwise limited or defined, “or” indicates a non- exclusive li st of components or operations that can be present in any variety of combinations, rather than an exclusive list of components that can be present only as alternatives to each other. For example, a list of “A, B, or C” indicates options of: A; B; C; A and B: A and C; B and C; and A, B, and C. Correspondingly, the term “or” as used herein is intended to indicate exclusive alternatives only when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” For example, a list of “one of A, B, or C” indicates options of: A, but not B and C; B, but not A and C; and C, but not A and B. A list preceded by “one or more” (and variations thereon) and including “or” to separate listed elements indicates options of one or more of any or all of the listed elements. For example, the phrases “one or more of A, B, or C” and “at least one of A, B, or C” indicate options of: one or more A; one or more B; one or more C; one or more A and one or more B; one or more B and one or more C: one or more A and one or more C: and one or more of A, one or more of B, and one or more of C. Similarly, a list preceded by “a plurality of” (and variations thereon) and including “or” to separate listed elements indicates options of multiple instances of any or all of the listed elements. For example, the phrases “a plurality of A, B, or C” and “two or more of A, B, or C” indicate options of: A and B; B and C; A and C; and A, B, and C.

[00131] Furthermore, in those instances where a convention analogous to “at least one of A, B and C, etc.” is used, in general such a construction is intended in the sense of one having ordinary' skill in the art would understand the convention (e.g., “a system having at least one of A, B and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description or figures, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase “A or B” will be understood to include the possibilities of “A” or ‘B or “A and B.” [00132] This discussion is presented to enable a person skilled in the art to make and use claims of the disclosure. Various modifications to the illustrated examples will be readily apparent to those skilled in the art, and the generic principles herein can be applied to other examples and applications without departing from the principles disclosed herein. Thus, claims of the disclosure are not intended to be limited to claims shown, but are to be accorded the widest scope consistent with the principles and features disclosed herein and the claims below. The following detailed description is to be read with reference to the figures, in which like elements in different figures have like reference numerals. The figures, which are not necessarily to scale, depict selected examples and are not intended to limit the scope of the disclosure. Skilled artisans will recognize the examples provided herein have many useful alternatives and fall within the scope of the disclosure.

[00133] Various features and advantages of the disclosure are set forth in the following claims.