Method for screening B cell lymphocytes

文档序号:1539460 发布日期:2020-02-14 浏览:18次 中文

阅读说明:本技术 筛选b细胞淋巴细胞的方法 (Method for screening B cell lymphocytes ) 是由 M·帕克 J·C·布里格斯 J·M·麦克尤恩 R·K·拉梅纳尼 H·基拉蒂纳格尔 K·W· 于 2017-10-23 设计创作,主要内容包括:本文描述了用于在微流体环境中筛选抗体产生细胞的方法。抗体产生细胞可以是B细胞淋巴细胞,其可以是记忆B细胞或浆细胞。可以使目标抗原靠近抗体产生细胞,并且可以监测抗体产生细胞产生的抗体与抗原的结合。还描述了从抗体产生细胞获得测序文库的方法。(Described herein are methods for screening antibody producing cells in a microfluidic environment. The antibody-producing cell may be a B cell lymphocyte, which may be a memory B cell or a plasma cell. The antigen of interest can be brought into proximity with the antibody-producing cell, and binding of the antibody produced by the antibody-producing cell to the antigen can be monitored. Methods of obtaining sequencing libraries from antibody-producing cells are also described.)

1. A method of detecting B cell lymphocytes expressing an antibody that specifically binds an antigen of interest, the method comprising:

introducing a sample comprising B cell lymphocytes into a microfluidic device comprising:

a housing having a flow area and a docking station,

wherein the isolation dock comprises a separation region having a single opening and a connection region providing a fluidic connection between the separation region and the flow region, and wherein the separation region of the support dock is an unswept region of the microfluidic device;

loading B cell lymphocytes from the sample into the isolation region of the isolation dock;

introducing the antigen of interest into the flow region of the housing such that the antigen of interest is in proximity to the B cell lymphocytes; and the number of the first and second electrodes,

monitoring binding of said antigen of interest to said antibody expressed by said B cell lymphocytes,

wherein the separation region of the isolation dock comprises at least one conditioned surface.

2. The method of claim 1, wherein the at least one conditioned surface comprises a layer of covalently linked hydrophilic molecules.

3. The method of claim 2, wherein the hydrophilic molecule comprises a polyethylene glycol (PEG) -containing polymer.

4. The method of any one of claims 1 to 3, wherein the housing of the microfluidic device further comprises a Dielectrophoresis (DEP) configuration.

5. The method of claim 4, wherein the housing of the microfluidic device further comprises a base, a microfluidic conduit structure, and a lid that together define a microfluidic conduit, and wherein the microfluidic conduit comprises the flow region and the isolation dock.

6. The method of claim 5, wherein:

the base includes a first electrode;

the cover comprises a second electrode; and is

The base or the cover comprises an electrode-activated substrate,

wherein the electrode activation substrate has a surface comprising a plurality of DEP electrode regions, and wherein the surface of the electrode activation substrate provides the inner surface of the flow region.

7. The method of claim 1, wherein the width W of the connection regionconFrom about 20 microns to about 60 microns.

8. The method of claim 1, wherein the linking region has a length LconAnd wherein said length L of said connection regionconThe width W of the connection regionconHas a value of at least 1.5.

9. The method of claim 7 or 8, wherein the width W of the separation regionisoIs greater than the width W of the connection regioncon

10. The method of claim 9, wherein the width W of the separation regionisoFrom about 50 microns to about 250 microns.

11. The method of claim 1, wherein the isolating dock comprises a volume of about 0.5nL to about 2.5 nL.

12. The method of claim 1, wherein the isolated area of the isolation dock comprises at least one surface coated with a coating material.

13. The method of claim 12, wherein the coating material comprises a polymer comprising polyethylene glycol (PEG).

14. The method of claim 1, wherein the sample comprising B cell lymphocytes is a sample of peripheral blood, spleen biopsy, bone marrow biopsy, lymph node biopsy, or tumor biopsy.

15. The method of claim 14, wherein the sample comprising B cell lymphocytes is a peripheral blood sample.

16. The method of claim 14, wherein the sample comprising B cell lymphocytes is a bone marrow biopsy.

17. The method of claim 14 or 15, wherein the B cell lymphocyte is a memory B cell.

18. The method of claim 14 or 16, wherein the B cell lymphocyte is a plasma B cell.

19. The method of claim 1, wherein the sample comprising B cell lymphocytes is obtained from a mammal or an avian animal.

20. The method of claim 19, wherein the sample comprising B cell lymphocytes is obtained from a human, mouse, rat, guinea pig, gerbil, hamster, rabbit, goat, sheep, llama, or chicken.

21. The method of claim 19 or 20, wherein the mammal has been immunized against the antigen of interest, wherein the mammal has been exposed to or infected with a pathogen associated with the antigen of interest, wherein the mammal has a cancer and the cancer is associated with the antigen of interest, or wherein the mammal has an autoimmune disease and the autoimmune disease is associated with the antigen of interest.

22. The method of claim 1, wherein the sample comprising B cell lymphocytes has been depleted of cell types other than B cell lymphocytes.

23. The method of claim 1 or 22, wherein the sample comprising B cell lymphocytes is enriched for B cell lymphocytes expressing CD 27.

24. The method of claim 1 or 22, wherein the sample comprising B cell lymphocytes is enriched for CD138 expressing B cell lymphocytes.

25. The method of claim 1, wherein the sample comprising B cell lymphocytes has been contacted with a dnase enzyme prior to introduction into the microfluidic device.

26. The method of claim 1, wherein a single B cell lymphocyte is loaded into the separation region.

27. The method of claim 1, wherein a plurality of B cell lymphocytes are loaded into the separation region.

28. The method of claim 1, further comprising:

contacting the B cell lymphocyte with a stimulating agent that stimulates B cell activation.

29. The method of claim 28, wherein the stimulating agent comprises a CD40 agonist.

30. The method of claim 29, wherein the stimulating agent comprises one or more CD40L+Feeder cells.

31. The method of claim 29 or 30, wherein the stimulating agent further comprises a B Cell Receptor (BCR) -linked molecule, wherein the BCR-linked molecule comprises protein a or protein G.

32. The method of claim 31, wherein the BCR-linked molecule is linked to a micro-object.

33. The method of claim 28, wherein contacting the B cell lymphocyte with a stimulating agent comprises contacting the B cell lymphocyte with CD40L+Contacting feeder cells with a mixture of protein a conjugated to beads, wherein contacting the B-cell lymphocytes with a stimulating agent comprises loading the mixture into the isolated region of the isolated dock.

34. The method of claim 29, wherein the B cell lymphocytes are contacted with the stimulating agent substantially continuously for a period of 3 to 5 days.

35. The method of claim 29, further comprising:

providing a culture medium to the B cell lymphocytes, wherein the culture medium comprises one or more agents that promote B cell expansion and/or activation.

36. The method of claim 35, wherein the culture medium comprises IL-2, IL-4, IL-6, IL-21, and BAFF or April.

37. The method of claim 35, wherein the culture medium comprises a TLR agonist.

38. The method of claim 37, wherein the TLR agonist is a CpG oligonucleotide.

39. The method of claim 35, wherein the B cell lymphocytes are provided with culture medium for a period of 3 to 5 days.

40. The method of claim 35, wherein said contacting with a stimulating agent and said providing a culture medium are performed over a substantially coextensive period.

41. The method of claim 4, wherein loading the B-cell lymphocytes into the separation region of the isolation dock comprises moving the B-cell lymphocytes into the isolation region using DEP forces.

42. The method of claim 41, wherein the B cell lymphocytes move from the flow region to the isolation region.

43. The method of claim 1, wherein providing the antigen of interest comprises flowing a solution comprising a soluble antigen of interest into or through the flow region.

44. The method of claim 43, wherein the antigen of interest is covalently bound to a first detectable label.

45. The method of claim 43 or 44, further comprising providing a micro-object comprising a first antibody binding agent, wherein the first antibody binding agent binds the antibody expressed by the B-cell lymphocytes without inhibiting binding of a target antigen to the antibody expressed by the B-cell lymphocytes, and wherein monitoring binding of the target antigen to the antibody expressed by the B-cell lymphocytes comprises detecting indirect binding of the target antigen to the micro-object.

46. The method of claim 45, wherein the first antibody binding agent binds to the Fc domain of the antibody expressed by the B cell lymphocytes.

47. The method of claim 45, wherein providing the micro-objects comprises flowing a solution comprising the micro-objects into the flow region and stopping flow while the micro-objects are in proximity to the isolation dock.

48. The method of claim 45, wherein the solution comprising the micro-objects and the solution comprising the soluble target antigen are the same solution.

49. The method of claim 45, further comprising:

providing a second antibody binding agent, wherein the second antibody binding agent comprises a second detectable label; and are

Monitoring indirect binding of the second antibody binding agent to the micro-object,

wherein the first detectable label is different from the second detectable label.

50. The method of claim 49 wherein the second antibody binding agent binds to an IgG antibody.

51. The method of claim 1, wherein providing the antigen of interest comprises providing a micro-object comprising the antigen of interest, wherein the micro-object is a cell, a liposome, a lipid nanoraft, or a bead.

52. The method of claim 51, further comprising:

providing a labelled antibody binding agent prior to or simultaneously with said antigen of interest,

wherein said monitoring binding of said antigen of interest to said antibody expressed by said B cell lymphocytes comprises detecting indirect binding of said labeled antibody binding agent to said antigen of interest.

53. The method of claim 52 wherein the labeled antibody binding agent binds to an anti-IgG antibody.

54. The method of claim 1, wherein monitoring binding of the antigen of interest to the antibody expressed by the B cell lymphocyte comprises imaging all or a portion of the sequestration dock of the microfluidic device.

55. The method of claim 54, wherein said imaging comprises fluorescence imaging.

56. The method of claim 54, wherein said imaging comprises taking a plurality of images.

57. The method of claim 1, wherein the microfluidic device comprises a plurality of the isolated docks, each isolated dock having a separation region and a connection region, each connection region providing a fluidic connection between the separation region and the flow region, the method further comprising:

loading one or more of the plurality of B-cell lymphocytes into the separation region of each of two or more of the plurality of isolation docks;

introducing the antigen of interest into the microfluidic device such that the antigen of interest is proximate to each of the two or more isolated docks loaded with one or more B cell lymphocytes; and is

Monitoring binding of said antigen of interest to said antibody expressed by each of said loaded B cell lymphocytes.

58. The method of claim 57, wherein a single B cell lymphocyte is loaded into the separation region of each of the two or more of the plurality of isolation docks.

59. The method of claim 57, further comprising:

detecting binding of said antigen of interest to said antibody expressed by said loaded B cell lymphocyte or several of said plurality of loaded B cell lymphocytes;

identifying the loaded B-cell lymphocyte or the ones of the plurality of loaded B-cell lymphocytes that express an antibody that specifically binds the antigen of interest.

60. A method of characterizing an antibody that specifically binds to an antigen of interest, the method comprising:

identifying a B cell lymphocyte, or clonal population thereof, that expresses an antibody that specifically binds to said antigen of interest, wherein said identifying is performed according to the method of claim 59;

from said B cell lymphocytes or thereofIsolation of immunoglobulin encoding heavy chain variable region (V) from clonal populationH) And/or immunoglobulin light chain variable region (V)L) The nucleic acid of (1); and are

For encoding the immunoglobulin heavy chain variable region (V)H) At least a part of the nucleic acid of (a) and/or encoding the immunoglobulin light chain variable region (V)L) Sequencing at least a portion of the nucleic acid of (a).

61. The method of claim 60, wherein the immunoglobulin heavy chain variable region (V) is mutatedH) Performing sequencing comprises:

lysing said identified B cell lymphocytes or said clonal population of B cell lymphocytes;

reverse transcribing mRNA isolated from said B cell lymphocytes of said B cell lymphocytes or said clonal population thereof, wherein said mRNA encodes said immunoglobulin heavy chain variable region (V)H) Thereby forming VHcDNA; and are

For the VHAt least a portion of the cDNA is sequenced.

62. The method of claim 60, wherein the immunoglobulin light chain variable region (V) is mutatedL) Performing sequencing comprises:

lysing said identified B cell lymphocytes or said clonal population of B cell lymphocytes;

reverse transcribing mRNA isolated from said B cell lymphocytes or said clonal population thereof, wherein said mRNA encodes said immunoglobulin light chain variable region (V)L) Thereby forming VLcDNA; and are

For the VLAt least a portion of the cDNA is sequenced.

63. The method of claim 61 or 62, wherein reverse transcribing the mRNA comprises contacting the mRNA with a capture/priming oligonucleotide.

64. The method of claim 63, wherein the reverse transcription is performed in the presence of a transcript conversion oligonucleotide.

65. The method of claim 61 or 62, wherein said identified B cell lymphocytes or said B cell lymphocytes of said clonal population thereof are output from said microfluidic device prior to lysis.

66. The method of claim 65, wherein outputting the identified B-cell lymphocytes or the clonal population thereof comprises:

moving the identified B-cell lymphocytes or the B-cell lymphocytes of the clonal population thereof from the isolation region of the isolation dock into the flow region of the microfluidic device; and are

Flowing the identified B cell lymphocytes or the B cell lymphocytes of the clonal population thereof through the flow region and out of the microfluidic device.

67. The method of claim 66, wherein moving said identified B cell lymphocytes or said B cell lymphocytes of said clonal population thereof from said isolated region of said isolation dock comprises capturing and moving said identified B cell lymphocytes or said clonal population using DEP forces.

68. The method of claim 61 or 62, wherein said identified B cell lymphocytes or said B cell lymphocytes of said clonal population thereof are lysed within said microfluidic device.

69. The method of claim 68, further comprising:

providing one or more capture beads in close proximity to the identified B cell lymphocytes or the B cell lymphocytes of the clonal population thereof, wherein each of the one or more capture beads comprises a bead capable of binding the VHmRNA and/or the VLAn oligonucleotide of mRNA;

lysing said identified B cell lymphocytes or said clonal population thereof; and is

Allowing the V from the lysed B cell lymphocytes or the lysed B cell lymphocytes from the clonal population thereofHmRNA and/or the VLThe mRNA is bound by the one or more capture beads.

70. The method of claim 69, wherein each capture bead of the one or more capture beads comprises a plurality of capture/priming oligonucleotides.

71. The method of claim 69, wherein the bound VHmRNA and/or said bound VLReverse transcription of mRNA into V upon binding to the one or more capture beadsHcDNA and/or VLcDNA。

72. The method of claim 71, wherein said VHcDNA and/or VLThe cDNA is output from the microfluidic device while bound to the one or more capture beads.

73. The method of claim 61 or 62, further comprising amplifying the V prior to the sequencingHcDNA and/or said VLcDNA。

74. The method of claim 73, wherein said expanding comprises increasing V in reverse transcribed mRNA isolated from said B cell lymphocyteHcDNA and/or VLExpression of cDNA or fragments thereof.

75. The method of claim 74, wherein the amplifying comprises:

a first round of expansion that increases V in reverse transcribed mRNA isolated from the B cell lymphocytesHcDNA and/or VLExpression of cDNA or fragments thereof; and

second round amplification which introduces barcode sequences into V amplified in the first roundHcDNA and/or VLcDNA or a fragment thereof.

Background

It is of interest to screen and identify cells that produce antibodies capable of specifically binding to an antigen of interest, including in the field of hybridoma development. Furthermore, it is of interest to recognize highly expressed antibody producing cells. Providing a suitable environment suitable for the growth environment of the antibody producing cells and providing an environment in which binding/expression assays can be easily monitored has been a formidable challenge. Furthermore, it is desirable to provide a correlation of the assay results with a particular cell that demonstrates the desired expression/binding characteristics of the antibody it secretes. Improvements to these aspects of the field of antibody development are provided herein.

Disclosure of Invention

The present invention is based in part on the following findings: b cell lymphocytes, including primary B cells, can be screened within a microfluidic device to determine whether the B cell lymphocytes express antibodies that specifically bind to an antigen of interest. Thus, in one aspect, a method of detecting expression of an antibody that specifically binds to an antigen of interest by an antibody-producing cell is provided. The method includes the step of introducing antibody-producing cells into a microfluidic device. The antibody-producing cell can be, for example, a B cell lymphocyte, such as a memory B cell or a plasma cell.

For example, a microfluidic device may include a flow region, which may include a microfluidic channel, and at least one microfluidic isolation dock (e.g., a plurality of isolation docks). Each isolation dock may include a separation region and a connection region that fluidly connects the separation region to a flow region (e.g., a microfluidic channel).

Some of the methods of the present disclosure include the following additional steps: loading antibody-producing cells into the isolated region of the sequestration dock; introducing a target antigen into the microfluidic device such that the target antigen is in proximity to the antibody-producing cells; and monitoring binding of the antigen of interest to the antibody expressed by the antibody-producing cell. The loaded cell may be one of a population of cells (e.g., B cells) loaded into a microfluidic device having a plurality of isolated docks. In such embodiments, one or more antibody-producing cells can be loaded into the isolated region of each of the plurality of isolated docks. In some embodiments, a single antibody-producing cell is loaded into each isolation dock. When provided in the vicinity of an antibody-producing cell, the antigen of interest may be solubilized or attached to a micro-object, such as a cell, a liposome, a lipid nanoraft (lipid nanoraft), or a synthetic bead (e.g., a microbead or nanobead). These micro objects can be visualized under a microscope. Monitoring binding between the antigen of interest and the antibody produced by the antibody-producing cell may comprise: providing a labeled target antigen and detecting direct binding of the target antigen (e.g., labeled target antigen); providing a labeled antibody binding agent, and detecting indirect binding of the labeled antibody binding agent to the antigen of interest (e.g., to a micro-object presenting the antigen of interest); and providing an antibody binding agent and detecting indirect binding of the labeled target antigen to the antibody binding agent (e.g., a micro-object attached to a plurality of antibody binding agents). The antibody binding agent can be isotype specific (e.g., an anti-IgG antibody or IgG-binding fragment thereof). The label on the antigen or target or antibody binding agent may be a fluorescent label.

For antibody-producing cells that are recognized as expressing an antigen-binding antibody, the disclosed methods can further comprise the steps of: lysing the identified cells (e.g., B cells); reverse transcription of V from lysed cellsHmRNA and/or VLmRNA to form V respectivelyHcDNA and/or VLcDNA; and for the VHcDNA and/or VLAt least a portion of the cDNA is sequenced. The lysis and reverse transcription steps may be performed within the microfluidic device or external to the microfluidic device. For example, the identified cells may be exported (e.g., as individual cells) for cell lysis and further processing. Alternatively, the recognized cells are lysed in the isolation dock in which they are loaded, the V released after lysisHmRNA and/or VLThe mRNA can be captured on beads (i.e., beads having oligonucleotides attached to their surfaces, wherein the oligonucleotides are capable of specifically binding to VHmRNA and/or VLmRNA) capture. The capture beads may be at the captured VHmRNA and/or captured VLThe mRNA is output from the microfluidic device either before or after reverse transcription.

These and other features and advantages of the method of the present invention will be set forth or will become more fully apparent in the description that follows and in the appended claims. The features and advantages may be realized and obtained by means of the instruments and combinations particularly pointed out in the appended embodiments and claims. Furthermore, the features and advantages of the described systems and methods may be learned by the practice or will be obvious from the description, as set forth hereinafter.

Brief description of the drawings

Fig. 1A illustrates an example of a microfluidic device and system for use with the microfluidic device, including associated control apparatus according to some embodiments disclosed herein.

Fig. 1B and 1C illustrate vertical and horizontal cross-sectional views, respectively, of a microfluidic device according to some embodiments disclosed herein.

Fig. 2A and 2B show vertical and horizontal cross-sectional views, respectively, of a microfluidic device with isolated docks, according to some embodiments of the present invention.

Fig. 2C illustrates a detailed horizontal cross-sectional view of an isolation dock according to some embodiments disclosed herein.

Fig. 2D illustrates a partial horizontal cross-sectional view of a microfluidic device with isolated docks according to some embodiments disclosed herein.

Fig. 2E and 2F show detailed horizontal cross-sectional views of an isolation dock according to some embodiments disclosed herein.

Fig. 2G illustrates a microfluidic device having a flow region comprising a plurality of flow channels, each flow channel fluidically connected to a plurality of isolation docks, according to embodiments disclosed herein.

Fig. 2H illustrates a partial vertical cross-sectional view of a microfluidic device according to embodiments disclosed herein, wherein the inward-facing surface of the base and the inward-facing surface of the lid are conditioned surfaces.

Fig. 3A illustrates a specific example of a system nest configured to be operably connected with a microfluidic device and associated control apparatus, according to some embodiments disclosed herein.

Fig. 3B illustrates an optical train of a system for controlling a microfluidic device according to some embodiments disclosed herein.

Fig. 4 illustrates steps in an exemplary workflow for detecting B cell lymphocytes expressing antibodies that specifically bind to an antigen of interest, according to some embodiments disclosed herein.

Fig. 5A-5C are photographic illustrations of a microfluidic device comprising a plurality of microfluidic channels, each microfluidic channel being fluidically connected to a plurality of sequestration docks, and showing a method of screening for B-cell lymphocytes according to some embodiments disclosed herein.

Fig. 6A is a schematic illustration of a method for activating and screening memory B cells, according to embodiments disclosed herein.

Fig. 6B is an image of individual memory B-cells moved into an isolated dock according to embodiments disclosed herein.

Fig. 6C is a flow diagram of a multiplex assay according to some embodiments disclosed herein.

Fig. 6D is a fluorescence image of memory B cells determined according to embodiments disclosed herein.

Fig. 6E is a schematic diagram of steps in a method for screening memory B cells, according to embodiments disclosed herein, that begins with assaying a polyclonal group of memory B cells, and then separating the memory B cell group into individual sequestration docks for subsequent assays.

Fig. 7A is a schematic illustration of a method for screening plasma cells according to embodiments disclosed herein.

Fig. 7B is a set of bright field and corresponding fluorescence images of plasma cells determined according to embodiments disclosed herein.

Figure 8 is a schematic of a method of generating a BCR sequencing library.

Figure 9A is a graphical representation of electropherographic analysis of the size distribution of cDNA generated by single cell export and mRNA capture.

FIG. 9B is a photographic illustration of an electropherogram generated from single-cell amplicons produced by one embodiment of the methods described herein.

FIGS. 10A-10C are pictorial illustrations of amplicon electropherograms generated from 19 single cell captured mRNA, according to one embodiment of the methods described herein.

Detailed description of the invention

This specification describes exemplary embodiments and applications of the invention. However, the invention is not limited to these exemplary embodiments and applications, nor to the manner in which the exemplary embodiments and applications operate or are described herein. Further, the figures may show simplified or partial views, and the sizes of elements in the figures may be exaggerated or not in proportion. In addition, as the terms "on," "attached to," "connected to," "coupled to" or the like are used herein, an element (e.g., a material, a layer, a substrate, etc.) may be "on," "attached to," "connected to" or "coupled to" another element, whether the element is directly on, attached to, connected to or coupled to the other element, or whether there are one or more intervening elements between the element and the other element. Further, unless the context dictates otherwise, directions (e.g., above, below, top, bottom, side, up, down, below … …, above … …, above, below, horizontal, vertical, "x", "y", "z", etc.) if provided are relative and provided by way of example only and for ease of illustration and discussion and not by way of limitation. In addition, where a list of elements (e.g., elements a, b, c) is referred to, such reference is intended to include any one of the listed elements themselves, any combination of fewer than all of the listed elements, and/or combinations of all of the listed elements. The division of sections in the specification is for ease of review only and does not limit any combination of the elements discussed.

As used herein, "substantially" means sufficient for the intended purpose. Thus, the term "substantially" allows for minor, insignificant variations from absolute or perfect states, dimensions, measurements, results, etc., such as would be expected by one of ordinary skill in the art without significantly affecting overall performance. "substantially" when used in relation to a numerical value or a parameter or characteristic that may be expressed as a numerical value means within ten percent.

The terms "a" and "an" mean more than one.

As used herein, the term "plurality" can be 2,3, 4,5, 6, 7, 8, 9, 10, or more.

As used herein, the term "disposed" includes within its meaning "located".

As used herein, a "microfluidic device" or "microfluidic apparatus" is a device that: comprising one or more separate microfluidic conduits configured to contain a fluid, each microfluidic conduit comprising fluidly interconnected conduit elements including, but not limited to, regions, flow paths, channels, chambers, and/or docks; and at least one port configured to allow fluid (and optionally micro-objects suspended in the fluid) to flow into and/or out of the microfluidic device. Typically, the microfluidic circuit of a microfluidic device will include a flow region (which flow path may include a microfluidic channel) and at least one chamber, and will accommodate a fluid volume of less than about 1mL (e.g., less than about 750, 500, 250, 200, 150, 100, 75, 50, 25, 20, 15, 10, 9, 8, 7,6, 5, 4, 3, or 2 μ Ι _). In certain embodiments, the microfluidic circuit contains about 1-2, 1-3, 1-4, 1-5, 2-8, 2-10, 2-12, 2-15, 2-20, 5-30, 5-40, 5-50, 10-75, 10-100, 20-150, 20-200, 50-250, or 50-300 μ L. The microfluidic circuit may be configured to have a first end in fluid connection with a first port (e.g., inlet) in the microfluidic device and a second end in fluid connection with a second port (e.g., outlet) in the microfluidic device.

As used herein, a "nanofluidic device" or "nanofluidic apparatus" is a microfluidic device having microfluidic tubing containing at least one tubing element configured to accommodate a fluid volume of less than about 1 μ L, e.g., less than about 750, 500, 250, 200, 150, 100, 75, 50, 25, 20, 15, 10, 9, 8, 7,6, 5, 4, 3, 2, 1nL or less. The nanofluidic device may include a plurality of piping elements (e.g., at least 2,3, 4,5, 6, 7, 8, 9, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 700, 800, 900, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 6000, 7000, 8000, 9000, 10,000, or more). In certain embodiments, one or more (e.g., all) of the at least one piping element is configured to hold a fluid volume of about 100pL to 1nL, 100pL to 2nL, 100pL to 5nL, 250pL to 2nL, 250pL to 5nL, 250pL to 10nL, 500pL to 5nL, 500pL to 10nL, 500pL to 15nL, 750pL to 10nL, 750pL to 15nL, 750pL to 20nL, 1 to 10nL, 1 to 15nL, 1 to 20nL, 1 to 25nL, or 1 to 50 nL. In other embodiments, one or more (e.g., all) of the at least one piping element is configured to hold a fluid volume of about 20nL to 200nL, 100 to 300nL, 100 to 400nL, 100 to 500nL, 200 to 300nL, 200 to 400nL, 200 to 500nL, 200 to 600nL, 200 to 700nL, 250 to 400nL, 250 to 500nL, 250 to 600nL, or 250 to 750 nL.

Microfluidic devices or nanofluidic devices may be referred to herein as "microfluidic chips" or "chips"; or "nanofluidic chip" or "chip".

As used herein, "microfluidic channel" or "flow channel" refers to a flow region of a microfluidic device that is significantly longer than the horizontal and vertical dimensions. For example, the flow channel can be at least 5 times the length of the horizontal or vertical dimension, e.g., at least 10 times the length, at least 25 times the length, at least 100 times the length, at least 200 times the length, at least 500 times the length, at least 1,000 times the length, at least 5,000 times the length, or longer. In some embodiments, the length of the flow channel is in the range of about 50,000 micrometers to about 500,000 micrometers, including any range therebetween. In some embodiments, the horizontal dimension is in a range from about 100 microns to about 1000 microns (e.g., about 150 to about 500 microns) and the vertical dimension is in a range from about 25 microns to about 200 microns, e.g., about 40 to about 150 microns. It should be noted that the flow channels may have a variety of different spatial configurations in the microfluidic device and are therefore not limited to perfectly linear elements. For example, the flow channel may include one or more portions having any of the following configurations: curved, bent, spiral, inclined, descending, forked (e.g., multiple distinct flow paths), and any combination thereof. In addition, the flow channel may have different cross-sectional areas along its path, widening and narrowing to provide the desired fluid flow therein.

As used herein, the term "obstruction" generally refers to a protrusion or similar type of structure that is large enough to partially (but not completely) impede movement of a target micro-object between two different regions or conduit elements in a microfluidic device. The two different regions/pipe elements may for example be a microfluidic isolation dock and a microfluidic channel, or a connection region and a separation region of a microfluidic isolation dock.

As used herein, the term "constriction" generally refers to a narrowing of the width of a conduit element (or the interface between two conduit elements) in a microfluidic device. The constriction may be located, for example, at the interface between the microfluidic isolation dock and the microfluidic channel, or at the interface between the separation region and the connection region of the microfluidic isolation dock.

As used herein, the term "transparent" refers to a material that allows the passage of visible light without substantially changing the light when passed.

As used herein, the term "micro-object" generally refers to any micro-object that can be separated and/or processed according to the present invention. Non-limiting examples of micro-objects include: inanimate micro-objects, such as microparticles; microbeads (e.g., polystyrene beads, Luminex)TMBeads, etc.); magnetic beads; a micron rod; microfilaments; quantum dots, and the like; biological micro-objects, such as cells; a biological organelle; a vesicle or complex; synthesizing vesicles; liposomes (e.g., synthetic or derived from membrane preparations); lipid nanorafts (nanorafts), etc.; or a combination of inanimate and biological micro-objects (e.g., cell-attached microbeads, liposome-coated magnetic beads, etc.). The beads may include covalently or non-covalently attached moieties/molecules, such as fluorescent markers, proteins, carbohydrates, antigens, small molecule signaling moieties, or other chemical/biological substances that can be used in assays. Lipid nanorafts have been described, for example, in Ritchie et al (2009) "Regulation of Membrane Proteins in phospholipid Bilayer Nanodiscs," Methods Enzymol, 464: 211-231.

As used herein, the term "cell" may be used interchangeably with the term "biological cell". "non-limiting examples of biological cells include eukaryotic cells; a plant cell; animal cells, such as mammalian cells, reptile cells, avian cells, fish cells, and the like; a prokaryotic cell; a bacterial cell; a fungal cell; protozoan cells, etc.; cells dissociated from tissue (e.g., muscle, cartilage, fat, skin, liver, lung, neural tissue, etc.); immune cells such as T cells, B cells, natural killer cells, macrophages, and the like; embryos (e.g., fertilized eggs); an oocyte; an ovum; a sperm cell; a hybridoma; a cultured cell; cells from a cell line; cancer cells; infected cells; transfected and/or transformed cells; reporter cells, and the like. The mammalian cell can be, for example, a human, mouse, rat, horse, goat, sheep, cow, primate, and the like.

A colony of biological cells is "clonal" if all living cells in the colony that are capable of multiplying are daughter cells derived from a single parent cell. In certain embodiments, all daughter cells in the clonal colony are derived from a single parent cell no more than 10 divisions. In other embodiments, all daughter cells in the clonal colony are derived from no more than 14 divisions of a single parent cell. In other embodiments, all daughter cells in the clonal colony are from a single parent cell for no more than 17 divisions. In other embodiments, all daughter cells in the clonal colony are derived from a single parent cell no more than 20 divisions. The term "clonal cells" refers to cells of the same clonal colony.

As used herein, a "colony" of biological cells refers to 2 or more cells (e.g., about 2 to about 20, about 4 to about 40, about 6 to about 60, about 8 to about 80, about 10 to about 100, about 20 to about 200, about 40 to about 400, about 60 to about 600, about 80 to about 800, about 100 to about 1000, or greater than 1000 cells).

As used herein, the term "maintaining the cell(s)" refers to providing an environment comprising fluid and gas components and optionally surfaces that provide the conditions necessary to keep the cells viable and/or expanded.

As used herein, the term "expansion" when referring to cells refers to an increase in the number of cells.

A "component" of a fluid medium is any chemical or biochemical molecule present in the medium, including solvent molecules, ions, small molecules, antibiotics, nucleotides and nucleosides, nucleic acids, amino acids, peptides, proteins, sugars, carbohydrates, lipids, fatty acids, cholesterol, metabolites, and the like.

As used herein with respect to a fluid medium, "diffusion" and "diffusion" refer to the thermodynamic movement of components of the fluid medium down a concentration gradient.

The phrase "flow of the medium" means that the fluid medium is moved as a whole primarily due to any mechanism other than diffusion. For example, the flow of the medium may include the fluid medium moving from one point to another due to a pressure difference between the points. Such flow may include continuous, pulsed, periodic, random, intermittent, or reciprocating flow of liquid, or any combination thereof. When one fluid medium flows into another fluid medium, turbulence and mixing of the media may result.

The phrase "substantially no flow" refers to a flow rate of the fluid medium that, on average over time, is less than the rate at which a component of the material (e.g., the analyte of interest) diffuses into or within the fluid medium. The diffusion rate of the components of such materials may depend on, for example, the temperature, the size of the components, and the strength of the interaction between the components and the fluid medium.

As used herein with respect to different regions within a microfluidic device, the phrase "fluidically connected" refers to the fluids in each region being connected to form a single liquid when the different regions are substantially filled with a fluid (e.g., a fluidic medium). This does not mean that the fluids (or fluid media) in the different regions are necessarily identical in composition. In contrast, fluids in different fluidically connected regions of a microfluidic device can have different compositions (e.g., different concentrations of solutes, such as proteins, carbohydrates, ions, or other molecules) that are in change as the solutes move down their respective concentration gradients and/or the fluid flows through the device.

Microfluidic (or nanofluidic) devices may include "swept" areas and "unswept" areas. As used herein, a "swept-out" area includes one or more fluidically interconnected tubing elements of a microfluidic tubing, each tubing element being subjected to a flow of a medium as fluid flows through the microfluidic tubing. The conduit elements that sweep the area may include, for example, areas, channels, and all or part of the chamber. As used herein, an "unswept" region includes one or more fluidically interconnected conduit elements of a microfluidic conduit, each conduit element being substantially free of the flow of fluid as the fluid flows through the microfluidic conduit. The unswept region may be fluidly connected to the swept region, provided that the fluid connection is configured to enable diffusion but substantially no media flow between the swept region and the unswept region. Thus, the microfluidic device may be configured to substantially separate the unswept region from the flow of the medium in the swept region, while substantially only diffusive fluid communication is enabled between the swept region and the unswept region. For example, the flow channel of a microfluidic device is an example of a swept area, while the separation area of a microfluidic device (described in further detail below) is an example of an unswept area.

As used herein, "flow path" refers to one or more fluidly connected conduit elements (e.g., channels, regions, chambers, etc.) that define and are constrained by the trajectory of the media flow. Thus, the flow path is an example of a swept (swept) region of a microfluidic device. Other conduit elements (e.g., unswept areas) may be in fluid connection with the conduit elements comprising the flow path, independent of the flow of the medium in the flow path.

As used herein, "B" is used to denote a single nucleotide, being a nucleotide selected from G (guanosine), C (cytidine), and T (thymidine) nucleotides, but excluding a (adenine).

As used herein, "H" is used to denote a single nucleotide, being a nucleotide selected from A, C and T, but excluding G.

As used herein, "D" is used to denote a single nucleotide, being a nucleotide selected from A, G and T, but excluding C.

As used herein, "K" is used to denote a single nucleotide, being a nucleotide selected from G and T.

As used herein, "N" is used to denote a single nucleotide, being a nucleotide selected from A, C, G and T.

As used herein, "R" is used to denote a single nucleotide, being a nucleotide selected from a and G.

As used herein, "S" is used to denote a single nucleotide, being a nucleotide selected from G and C.

As used herein, "V" is used to denote a single nucleotide, is a nucleotide selected from A, G and C, and does not include T.

As used herein, "Y" is used to denote a single nucleotide, being a nucleotide selected from C and T.

As used herein, "I" is used to indicate that the mononucleotide is inosine.

As used herein, A, C, T, G followed by an "", indicates a phosphorothioate substitution in the phosphate linkage of the nucleotide.

As used herein, IsoG is isoguanosine; IsoC is isocytidine; IsodG is isoguanosine deoxyribonucleotide and IsodC deoxyribonucleotide. Each isoguanosine and isocytidine ribonucleotide or deoxyribonucleotide contains a nucleobase that is isomeric to a guanine nucleobase or a cytosine nucleobase, respectively, which is normally incorporated into RNA or DNA.

As used herein, rG denotes a ribonucleotide contained within a nucleic acid, and otherwise contains a deoxyribonucleotide. A nucleic acid containing all ribonucleotides may not include a label to indicate that each nucleotide is a ribonucleotide, but it is clear from the context.

As used herein, a "priming sequence" is an oligonucleotide sequence that is part of a larger oligonucleotide and can serve as a primer in a DNA (or RNA) polymerization reaction when separated from the larger oligonucleotide such that the priming sequence includes a free 3' end.

As used herein: μ m means micron, μm3Refers to cubic microns, pL refers to picoliters, nL refers to nanoliters, and μ L (oruL) means microliter.

And (4) loading method. Loading biological micro-objects or micro-objects (such as, but not limited to, beads) may involve the use of fluid flow, gravity, Dielectrophoresis (DEP) forces, electrowetting, magnetic forces, or any combination thereof, as described herein. DEP forces can be generated optically (e.g., by an optoelectronic tweezers (OET) configuration) and/or electrically (e.g., by activating electrodes/electrode regions in a temporal/spatial manner). Similarly, the electrowetting force may be provided optically (e.g. by an opto-electrowetting (OEW) configuration) and/or electrically (e.g. by activating the electrodes/electrode areas in a time-space manner).

Microfluidic devices and systems for operating and viewing such devices. Fig. 1A shows an example of a microfluidic device 100 and system 150 that can be used to screen and detect antibody-producing cells that secrete antibodies that bind (e.g., specifically bind) to an antigen of interest. A perspective view of the microfluidic device 100 is shown with the cover 110 partially cut away to provide a partial view into the microfluidic device 100. The microfluidic device 100 generally includes a microfluidic circuit 120 having a flow path 106, and a fluidic medium 180 may optionally carry one or more micro-objects (not shown) into and/or through the microfluidic circuit 120 via the flow path 106. Although a single microfluidic circuit 120 is shown in fig. 1A, a suitable microfluidic device may include a plurality (e.g., 2 or 3) of such microfluidic circuits. Regardless, the microfluidic device 100 may be configured as a nanofluidic device. In the embodiment shown in fig. 1A, the microfluidic circuit 120 includes a plurality of microfluidic isolation docks 124, 126, 128, and 130, each of which has an opening (e.g., a single opening) in fluid communication with the flow path 106. As discussed further below, the microfluidic sequestration dock includes various features and structures that have been optimized for retaining micro-objects in a microfluidic device (e.g., microfluidic device 100) even as the medium 180 flows through the flow path 106. However, before the above is described, a brief description of the microfluidic device 100 and system 150 is provided.

As shown generally in fig. 1A, microfluidic circuit 120 is defined by housing 102. Although the housing 102 can be physically configured in different configurations, in the example shown in fig. 1A, the housing 102 is depicted as including a support structure 104 (e.g., a base), a microfluidic conduit structure 108, and a cover 110. The support structure 104, the microfluidic circuit structure 108 and the cover 110 may be attached to each other. For example, the microfluidic circuit structure 108 may be arranged on an inner surface 109 of the support structure 104, and the cover 110 may be arranged over the microfluidic circuit structure 108. The microfluidic circuit structure 108, together with the support structure 104 and the lid 110, may define elements of a microfluidic circuit 120.

As shown in fig. 1A, the support structure 104 may be located at the bottom of the microfluidic circuit 120 and the lid 110 may be located at the top of the microfluidic circuit 120. Alternatively, the support structure 104 and the cover 110 may be configured in other orientations. For example, the support structure 104 may be located at the top of the microfluidic circuit 120 and the lid 110 may be located at the bottom of the microfluidic circuit 120. In any event, there may be one or more ports 107, each port 107 including a passageway into or out of the housing 102. Examples of passageways include valves, gates, through-holes, and the like. As shown, the port 107 is a through hole created by a gap in the microfluidic conduit structure 108. However, the port 107 may be located in other components of the housing 102 (e.g., the cover 110). Only one port 107 is shown in fig. 1A, but the microfluidic circuit 120 may have two or more ports 107. For example, there may be a first port 107 that serves as an inlet for fluid into the microfluidic circuit 120, and there may be a second port 107 that serves as an outlet for fluid out of the microfluidic circuit 120. Whether the port 107 serves as an inlet or an outlet may depend on the direction of fluid flow through the flow path 106.

The support structure 104 may include one or more electrodes (not shown) and a substrate or a plurality of interconnected substrates. For example, the support structure 104 may include one or more semiconductor substrates, each semiconductor substrate being electrically connected to an electrode (e.g., all or a portion of a semiconductor substrate may be electrically connected to a single electrode). The support structure 104 may further include a printed circuit board assembly ("PCBA"). For example, the semiconductor substrate may be mounted on a PCBA.

The microfluidic circuit structure 108 may define a circuit element of a microfluidic circuit 120. When microfluidic circuit 120 is filled with a fluid, such circuit elements may include spaces or regions that may be fluidically interconnected, such as flow regions (which may include or be one or more flow channels), chambers, docks, wells (traps), and the like. In the microfluidic circuit 120 shown in fig. 1A, the microfluidic circuit structure 108 includes a frame 114 and a microfluidic circuit material 116. The frame 114 may partially or completely surround the microfluidic circuit material 116. The frame 114 may be, for example, a relatively rigid structure that substantially encloses the microfluidic circuit material 116. For example, the frame 114 may comprise a metallic material.

The microfluidic circuit material 116 may be patterned with cavities or the like to define circuit elements and interconnections of the microfluidic circuit 120. The microfluidic circuit material 116 may include a flexible material, such as a flexible polymer (e.g., rubber, plastic, elastomer, silicone, polydimethylsiloxane ("PDMS"), etc.), which may be gas permeable. Other examples of materials from which the microfluidic circuit material 116 may be constructed include molded glass; etchable materials such as silicone (e.g., photo-patternable silicone or "PPS"), photoresist (e.g., SU8), and the like. In some embodiments, such materials (and thus the microfluidic circuit material 116) may be rigid and/or substantially gas impermeable. Regardless, the microfluidic circuit material 116 may be disposed on the support structure 104 and within the frame 114.

The cover 110 may be an integral (integral) component of the frame 114 and/or the microfluidic circuit material 116. Alternatively, the cover 110 may be a structurally different element, as shown in FIG. 1A. The cover 110 may comprise the same or different material as the frame 114 and/or the microfluidic circuit material 116. Similarly, the support structure 104 may be a separate structure from the frame 114 or the microfluidic circuit material 116 (as shown), or an integral component of the frame 114 or the microfluidic circuit material 116. Likewise, the frame 114 and the microfluidic circuit material 116 may be separate structures as shown in fig. 1A or integrated components of the same structure.

In some embodiments, the cover 110 may comprise a rigid material. The rigid material may be glass or a material with similar properties. In some embodiments, the cover 110 may include a deformable material. The deformable material may be a polymer, such as PDMS. In some embodiments, the cover 110 may include both a rigid material and a deformable material. For example, one or more portions of the cover 110 (e.g., one or more portions located above the isolation docks 124, 126, 128, 130) may include a deformable material that interfaces with the rigid material of the cover 110. In some embodiments, the cover 110 may further include one or more electrodes. The one or more electrodes may comprise a conductive oxide, such as Indium Tin Oxide (ITO), which may be coated on glass or similar insulating material. Alternatively, the one or more electrodes may be flexible electrodes, such as single-walled nanotubes, multi-walled nanotubes, nanowires, clusters of conductive nanoparticles embedded in a deformable material such as a polymer (e.g., PDMS), or a combination thereof. Flexible electrodes that may be used in microfluidic devices have been described, for example, in US 2012/0325665(Chiou et al), the contents of which are incorporated herein by reference. In some embodiments, the cover 110 may be modified (e.g., by adjusting all or a portion of the surface facing inward toward the microfluidic tubing 120) to support cell adhesion, viability, and/or growth. The modification may include a coating of a synthetic or natural polymer. In some embodiments, the cover 110 and/or the support structure 104 may be optically transparent. The cap 110 may also include at least one gas permeable material (e.g., PDMS or PPS).

Fig. 1A also shows a system 150 for operating and controlling a microfluidic device (e.g., microfluidic device 100). The system 150 includes a power source 192, an imaging device 194 (incorporated within the imaging module 164, where the device 194 is not shown in fig. 1A itself), and a tilt device 190 (incorporated into the tilt module 166, where the device 190 is not shown in fig. 1).

The power source 192 may provide power to the microfluidic device 100 and/or the tilting device 190 to provide a bias voltage or current as desired. The power supply 192 may, for example, include one or more Alternating Current (AC) and/or Direct Current (DC) voltage or current sources. The imaging device 194 (part of the imaging module 164, as discussed below) may include a device for capturing images within the microfluidic circuit 120, such as a digital camera. In some cases, the imaging device 194 further includes a detector with a fast frame rate and/or high sensitivity (e.g., for low light applications). The imaging device 194 may also include a mechanism for directing stimulating radiation and/or light beams into the microfluidic circuit 120 and collecting radiation and/or light beams reflected or emitted from the microfluidic circuit 120 (or micro-objects contained therein). The emitted light beam may be in the visible spectrum and may, for example, include fluorescent emissions. The reflected light beam may comprise reflected emissions from an LED or a broad spectrum lamp such as a mercury lamp (e.g. a high pressure mercury lamp) or a xenon arc lamp. As discussed with respect to fig. 3B, the imaging device 194 may further include a microscope (or optical train), which may or may not include an eyepiece.

The system 150 further includes a tilting device 190 (part of the tilting module 166, as discussed below) configured to rotate the microfluidic device 100 about one or more axes of rotation. In some embodiments, the tilting device 190 is configured to support and/or hold the housing 102 including the microfluidic circuit 120 about at least one axis such that the microfluidic device 100 (and thus the microfluidic circuit 120) can be held in a horizontal orientation (i.e., 0 ° with respect to the x-axis and y-axis), a vertical orientation (i.e., 90 ° with respect to the x-axis and/or y-axis), or any orientation therebetween. The orientation of the microfluidic device 100 (and microfluidic circuit 120) relative to the axis is referred to herein as the "tilt" of the microfluidic device 100 (and microfluidic circuit 120). For example, the tilting device 190 may tilt the microfluidic device 100 relative to the x-axis by 0.1 °, 0.2 °, 0.3 °, 0.4 °, 0.5 °, 0.6 °, 0.7 °, 0.8 °, 0.9 °,1 °,2 °,3 °,4 °,5 °, 10 °, 15 °, 20 °, 25 °, 30 °,35 °, 40 °, 45 °,50 °, 55 °, 60 °, 65 °, 70 °, 75 °, 80 °, 90 °, or any angle therebetween. The horizontal orientation (and thus the x-axis and y-axis) is defined as being perpendicular to the vertical axis defined by gravity. The tilting device may also tilt the microfluidic device 100 (and the microfluidic circuit 120) by any degree greater than 90 ° with respect to the x-axis and/or the y-axis, or tilt the microfluidic device 100 (and the microfluidic circuit 120) by 180 ° with respect to the x-axis or the y-axis, to completely invert the microfluidic device 100 (and the microfluidic circuit 120). Similarly, in some embodiments, the tilting device 190 tilts the microfluidic device 100 (and the microfluidic circuit 120) about an axis of rotation defined by the flow path 106 or some other portion of the microfluidic circuit 120.

In some cases, the microfluidic device 100 is tilted into a vertical orientation such that the flow path 106 is located above or below one or more isolation docks. The term "above" as used herein means that the flow path 106 is positioned higher than the one or more isolation docks on a vertical axis defined by gravity (i.e., an object in an isolation dock above the flow path 106 will have a higher gravitational potential energy than an object in the flow path). The term "below" as used herein means that the flow path 106 is positioned below the one or more isolation docks on a vertical axis defined by gravity (i.e., an object in an isolation dock below the flow path 106 will have a lower gravitational potential energy than an object in the flow path).

In some cases, the tilting device 190 tilts the microfluidic device 100 about an axis parallel to the flow path 106. Furthermore, the microfluidic device 100 may be tilted to an angle of less than 90 ° such that the flow path 106 is located above or below one or more isolation docks, rather than directly above or below the isolation docks. In other cases, the tilting device 190 tilts the microfluidic device 100 about an axis perpendicular to the flow path 106. In still other cases, the tilting device 190 tilts the microfluidic device 100 about an axis that is neither parallel nor perpendicular to the flow path 106.

The system 150 may further include a media source 178. The media source 178 (e.g., container, reservoir, etc.) may include multiple portions or containers, each portion or container for holding a different fluid media 180. Thus, the media source 178 can be a device that is external to and separate from the microfluidic device 100, as shown in fig. 1A. Alternatively, the media source 178 can be located wholly or partially within the housing 102 of the microfluidic device 100. For example, the media source 178 can include a reservoir that is part of the microfluidic device 100.

Fig. 1A also shows a simplified block diagram depicting an example of a control and monitoring apparatus 152 that forms part of the system 150 and that may be used in conjunction with the microfluidic device 100. As shown, examples of such control and monitoring devices 152 include a master controller 154 that may control other controller and monitoring devices, such as a media module 160 for controlling a media source 178; a motion module 162 for controlling movement and/or selection of micro-objects (not shown) and/or media (e.g., media droplets) in the microfluidic circuit 120; an imaging module 164 for controlling an imaging device 194 (e.g., a camera, a microscope, a light source, or any combination thereof) to capture an image (e.g., a digital image); and a tilt module 166 for controlling the tilt device 190. The control apparatus 152 may also include other modules 168 for controlling, monitoring, or performing other functions with respect to the microfluidic device 100. As shown, the device 152 may further include a display 170 and an input/output device 172.

The main controller 154 may include a control module 156 and digital memory 158. The control module 156 may include, for example, a digital processor configured to operate in accordance with machine-executable instructions (e.g., software, firmware, source code, etc.) stored as non-transitory data or signals in the memory 158. Alternatively or additionally, the control module 156 may include hard-wired digital circuitry and/or analog circuitry. Media module 160, motion module 162, imaging module 164, tilt module 166, and/or other modules 168 may be similarly configured. Accordingly, the functions, processes, actions, acts, or steps of the processes discussed herein as being performed with respect to the microfluidic device 100 or any other microfluidic apparatus may be implemented by any one or more of the master controller 154, the substrate module 160, the motion module 162, the imaging module 164, the tilt module 166, and/or the other modules 168 configured as discussed above. Similarly, the master controller 154, the media module 160, the motion module 162, the imaging module 164, the tilt module 166, and/or the other modules 168 may be communicatively coupled to send and receive data used in any of the functions, processes, actions, or steps discussed herein.

The media module 160 controls the media source 178. For example, the media module 160 may control the media source 178 to input a selected fluid media 180 into the housing 102 (e.g., through the inlet port 107). The media module 160 may also control the removal of media from the housing 102 (e.g., through an outlet port (not shown)). Thus, one or more media may be selectively input into and removed from the microfluidic circuit 120. The media module 160 may also control the flow of fluidic media 180 in the flow path 106 within the microfluidic circuit 120. For example, in some embodiments, the media module 160 stops the flow of the media 180 in the flow path 106 and through the housing 102 before the tilt module 166 causes the tilt device 190 to tilt the microfluidic device 100 to a desired tilt angle.

The motion module 162 may be configured to control the selection, trapping, and movement of micro-objects (not shown) in the microfluidic circuit 120. As discussed below with reference to fig. 1B and 1C, the enclosure 102 may include a Dielectrophoresis (DEP), optoelectronic tweezers (OET), and/or optoelectronic wetting (OEW) configuration (not shown in fig. 1A), and the motion module 162 may control activation of electrodes and/or transistors (e.g., phototransistors) to select and move micro-objects (not shown) and/or media droplets (not shown) in the flow path 106 and/or isolation docks 124, 126, 128, 130.

The imaging module 164 may control an imaging device 194. For example, the imaging module 164 may receive and process image data from the imaging device 194. The image data from imaging device 194 may include any type of information captured by imaging device 194 (e.g., the presence or absence of micro-objects, drops of media, accumulation of markers (e.g., fluorescent markers), etc.). Using the information captured by imaging device 194, imaging module 164 may further calculate the locations of objects (e.g., micro-objects, media drops) within microfluidic device 100 and/or the rates of motion of these objects.

The tilt module 166 may control the tilting motion of the tilting device 190. Alternatively or additionally, the tilting module 166 may control the tilting rate and timing to optimize the transfer of micro-objects to one or more isolation docks via gravity. Tilt module 166 is communicatively coupled with imaging module 164 to receive data describing the movement of micro-objects and/or media drops in microfluidic circuit 120. Using this data, tilt module 166 can adjust the tilt of microfluidic circuit 120 in order to adjust the rate at which micro-objects and/or media droplets move in microfluidic circuit 120. Tilt module 166 may also use this data to iteratively adjust the position of micro-objects and/or media drops in microfluidic circuit 120.

In the example shown in fig. 1A, microfluidic circuit 120 is shown to include microfluidic channel 122 and isolation docks 124, 126, 128, 130. Each dock includes a single opening to the channel 122, but otherwise is enclosed so that the dock can substantially separate micro-objects within the dock from the fluid medium 180 and/or micro-objects in the flow path 106 of the channel 122 or other dock. The walls of the isolation dock extend from the inner surface 109 of the base to the inner surface of the cover 110 to provide an enclosure. The opening of the dock to channel 122 is oriented at an angle to the flow 106 of fluid medium 180 so that the flow 106 is not directed into the dock. The flow may be tangential or perpendicular to the plane of the opening of the dock. In some cases, the docks 124, 126, 128, 130 are configured to physically enclose one or more micro-objects within the microfluidic circuit 120. An isolating dock according to the present invention may include various shapes, surfaces and features that are optimized for DEP, OET, OEW, fluid flow and/or gravity as will be discussed and illustrated in detail below.

The microfluidic circuit 120 may include any number of microfluidic isolation docks. Although five isolation docks are shown, microfluidic circuit 120 may have fewer or more isolation docks. As shown, the microfluidic isolation docks 124, 126, 128, and 130 of the microfluidic circuit 120 each include different features and shapes that may provide one or more benefits for screening antibody-producing cells (e.g., separating one antibody-producing cell from another). The microfluidic isolation docks 124, 126, 128, and 130 may provide other benefits, such as facilitating single cell loading and/or growth of colonies of antibody-producing cells (e.g., clonal colonies). In some embodiments, microfluidic circuit 120 includes a plurality of identical microfluidic isolation docks.

In some embodiments, microfluidic circuit 120 includes a plurality of microfluidic isolation docks, wherein two or more of the isolation docks include different structures and/or features that provide different benefits of screening for antibody-producing cells. The microfluidic device for screening antibody-producing cells may include any of the isolated docks 124, 126, 128, and 130, or variations thereof, and/or may include docks similar to the dock configurations shown in fig. 2B, 2C, 2D, 2E, and 2F, as discussed below.

In the embodiment shown in fig. 1A, a single channel 122 and flow path 106 are shown. However, other embodiments may contain multiple channels 122, each configured to include a flow path 106. The microfluidic circuit 120 further includes an inlet valve or port 107 in fluid communication with the flow path 106 and the fluidic medium 180, whereby the fluidic medium 180 may enter the channel 122 via the inlet port 107. In some cases, the flow path 106 comprises a single path. In some cases, the single paths are arranged in a zigzag pattern whereby the flow paths 106 pass through the microfluidic device 100 in alternating directions two or more times.

In some cases, microfluidic circuit 120 includes a plurality of parallel channels 122 and flow paths 106, wherein fluidic medium 180 within each flow path 106 flows in the same direction. In some cases, the fluid medium within each flow path 106 flows in at least one of a forward direction or a reverse direction. In some cases, multiple isolation docks are configured (e.g., relative to channel 122) such that the isolation docks can load the target micro-object in parallel.

In some embodiments, microfluidic circuit 120 further comprises one or more micro-object wells 132. The wells 132 are generally formed in the walls that border the channel 122 and may be disposed opposite the openings of one or more of the microfluidic isolation stations 124, 126, 128, 130. In some embodiments, the trap 132 is configured to receive or capture a single micro-object from the flow path 106. In some embodiments, the trap 132 is configured to receive or capture a plurality of micro-objects from the flow path 106. In some cases, well 132 includes a volume approximately equal to the volume of a single target micro-object.

The well 132 may also include an opening configured to assist the flow of the target micro-object into the well 132. In some cases, well 132 includes an opening having a height and width approximately equal to the dimensions of a single target micro-object, thereby preventing larger micro-objects from entering the micro-object well. The well 132 may further include other features configured to help retain the target micro-object within the well 132. In some cases, wells 132 are aligned with respect to the opening of the microfluidic isolation dock and are located on opposite sides of channel 122 such that when microfluidic device 100 is tilted about an axis parallel to channel 122, trapped micro-objects exit wells 132 in a trajectory that causes the micro-objects to fall into the opening of the isolation dock. In some cases, the well 132 includes side channels 134 that are smaller than the target micro-object in order to facilitate flow through the well 132, thereby increasing the likelihood of micro-objects being trapped in the well 132.

In some embodiments, Dielectrophoretic (DEP) forces are applied to the fluidic medium 180 (e.g., in the flow path and/or in the isolation dock) via one or more electrodes (not shown) to manipulate, transport, separate, and sort micro-objects located therein. For example, in some embodiments, DEP forces are applied to one or more portions of the microfluidic circuit 120 in order to transfer individual micro-objects from the flow path 106 into a desired microfluidic isolation dock. In some embodiments, DEP forces are used to prevent micro-objects within an isolation dock (e.g., isolation dock 124, 126, 128, or 130) from being replaced therefrom. Further, in some embodiments, DEP forces are used to selectively remove micro-objects previously collected according to the teachings of the present invention from the isolation dock. In some embodiments, the DEP force comprises an optoelectronic tweezers (OET) force.

In other embodiments, an electro-optical wetting (OEW) force is applied to one or more locations (e.g., locations that help define a flow path and/or a plurality of isolation docks) in the support structure 104 (and/or lid 110) of the microfluidic device 100 by one or more electrodes (not shown) to manipulate, transport, separate, and sort droplets located in the microfluidic circuit 120. For example, in some embodiments, OEW forces are applied to one or more locations in the support structure 104 (and/or the lid 110) to transfer individual droplets from the flow path 106 into a desired microfluidic isolation dock. In some embodiments, OEW forces are used to prevent droplets within an isolation dock (e.g., isolation dock 124, 126, 128, or 130) from being replaced therefrom. Furthermore, in some embodiments, OEW forces are used to selectively remove droplets previously collected according to the teachings of the present invention from the isolation dock.

In some embodiments, DEP and/or OEW forces are combined with other forces (e.g., flow and/or gravity) in order to manipulate, transport, separate, and sort micro-objects and/or droplets within the microfluidic circuit 120. For example, the housing 102 can be tilted (e.g., by the tilting device 190) to position the flow path 106 and micro-objects located therein above the microfluidic isolation dock, and gravity can transport the micro-objects and/or droplets into the dock. In some embodiments, DEP and/or OEW forces may be applied before other forces are applied. In other embodiments, DEP and/or OEW forces may be applied after other forces are applied. In other cases, DEP and/or OEW forces may be applied simultaneously with or in an alternating manner with other forces.

Figures 1B, 1C, and 2A-2H illustrate various embodiments of microfluidic devices that can be used to implement the present invention. Fig. 1B depicts an embodiment of an electrokinetic device in which the microfluidic device 200 is configured to be optically actuated. A variety of optically actuated electrokinetic devices are known in the art, including devices having an opto-electronic tweezers (OET) configuration and devices having an opto-electronic wetting (OEW) configuration. Examples of suitable OET configurations are shown in the following U.S. patent documents, all of which are incorporated herein by reference in their entirety: U.S. Pat. No. RE44,711(Wu et al) (originally issued in U.S. Pat. No. 7,612,355); and U.S. Pat. No. 7,956,339(Ohta et al). Examples of OEW configurations are shown in U.S. patent No. 6,958,132(Chiou et al) and U.S. patent application publication No. 2012/0024708(Chiou et al), both of which are incorporated herein by reference in their entirety. Another example of a light actuated electrodynamic device includes a combined OET/OEW configuration, examples of which are shown in U.S. patent publication nos. 20150306598(Khandros et al) and 20150306599(Khandros et al) and their corresponding PCT publications WO2015/164846 and WO2015/164847, both of which are incorporated herein by reference in their entirety.

Examples of microfluidic devices with docks in which antibody producing cells may be placed, cultured, monitored and/or screened have been described, for example, in US application nos. 14/060,117, 14/520,568 and 14/521,447, each of which is incorporated herein by reference in its entirety. Each of the aforementioned applications further describes a microfluidic device configured to generate Dielectrophoretic (DEP) forces, such as an optoelectronic tweezers (OET), or configured to provide optoelectronic wetting (OEW). For example, the optoelectronic tweezers device shown in fig. 2 of US application No. 14/060,117 is an example of a device that may be used to select and move a single biological micro-object or a group of biological micro-objects in an embodiment of the present invention.

A microfluidic device motion configuration. As mentioned above, the control and monitoring device of the system may comprise a motion module for selecting and moving objects, such as micro-objects or droplets, in the microfluidic circuit of the microfluidic device. Microfluidic devices may have various motion configurations depending on the type of object being moved and other considerations. For example, a Dielectrophoresis (DEP) configuration can be used to select and move micro-objects in a microfluidic circuit. Accordingly, the support structure 104 and/or the lid 110 of the microfluidic device 100 may comprise a DEP configuration for selectively inducing DEP forces on micro-objects in the fluidic medium 180 in the microfluidic circuit 120, thereby selecting, capturing and/or moving individual micro-objects or groups of micro-objects. Alternatively, the support structure 104 and/or the cover 110 of the microfluidic device 100 can comprise an Electrowetting (EW) configuration for selectively inducing EW forces on droplets in the fluidic medium 180 in the microfluidic circuit 120 to select, capture, and/or move individual droplets or groups of droplets.

One example of a microfluidic device 200 containing a DEP configuration is shown in fig. 1B and IC. Although fig. 1B and IC show a side cross-sectional view and a top cross-sectional view, respectively, of a portion of the housing 102 of the microfluidic device 200 with an open region/chamber 202 for purposes of simplicity, it should be understood that the region/chamber 202 may be part of a fluid conduit element having a more detailed structure, such as a growth chamber, an isolation dock, a flow region, or a flow channel. In addition, the microfluidic device 200 may include other fluid conduit elements. For example, the microfluidic device 200 may include multiple growth chambers or isolation docks and/or one or more flow regions or flow channels, such as those described herein with respect to the microfluidic device 100. DEP configurations can be incorporated into any such fluid conduit element of the microfluidic device 200, or selected portions thereof. It should also be understood that any of the microfluidic device components and system components described above or below may be incorporated into the microfluidic device 200 and/or used in combination with the microfluidic device 200. For example, the system 150 including the control and monitoring device 152 described above may be used with a microfluidic device 200, the microfluidic device 200 including one or more of a media module 160, a motion module 162, an imaging module 164, a tilt module 166, and other modules 168.

As shown in fig. 1B, the microfluidic device 200 includes a support structure 104 having a bottom electrode 204 and an electrode activation substrate 206 covering the bottom electrode 204, and a lid 110 having a top electrode 210, the top electrode 210 being spaced apart from the bottom electrode 204. The top electrode 210 and the electrode activation substrate 206 define opposing surfaces of the region/chamber 202. Thus, the dielectric 180 contained in the region/chamber 202 provides a resistive connection between the top electrode 210 and the electrode activation substrate 206. Also shown is a power supply 212 configured to connect to the bottom electrode 204 and the top electrode 210 and generate a bias voltage between these electrodes as required to generate DEP forces in the region/chamber 202. The power source 212 may be, for example, an Alternating Current (AC) power source.

In certain embodiments, the microfluidic device 200 shown in fig. 1B and 1C can have a light-actuated DEP configuration. Thus, changing the pattern of light 218 from the light source 216 (which may be controlled by the motion module 162) may selectively activate and deactivate the changing pattern of DEP electrodes at the regions 214 of the inner surface 208 of the electrode activation substrate 206. (hereinafter, the region 214 of the microfluidic device having the DEP configuration is referred to as the "DEP electrode region") as shown in fig. 1C, a light pattern 218 directed at the inner surface 208 of the electrode activation substrate 206 may illuminate a selected DEP electrode region 214a (shown in white) in a pattern such as a square. The non-illuminated DEP electrode regions 214 (cross-hatched) are referred to hereinafter as "dark" DEP electrode regions 214. The relative electrical impedance through the DEP electrode activation substrate 206 (i.e., from the bottom electrode 204 up to the inner surface 208 of the electrode activation substrate 206 interfacing with the medium 180 in the flow region 106) is greater than the relative electrical impedance through the medium 180 in the region/chamber 202 at each dark DEP electrode region 214 (i.e., from the inner surface 208 of the electrode activation substrate 206 to the top electrode 210 of the lid 110). However, the illuminated DEP electrode regions 214a exhibit a reduced relative impedance through the electrode activation substrate 206 that is less than the relative impedance through the medium 180 in the region/chamber 202 at each illuminated DEP electrode region 214 a.

With the power supply 212 activated, the aforementioned DEP configuration creates an electric field gradient in the fluid medium 180 between the illuminated DEP electrode region 214a and the adjacent dark DEP electrode region 214, which in turn creates a local DEP force that attracts or repels nearby micro-objects (not shown) in the fluid medium 180. Thus, by varying the light pattern 218 projected from the light source 216 into the microfluidic device 200, DEP electrodes that attract or repel micro-objects in the fluidic medium 180 can be selectively activated and deactivated at many different such DEP electrode regions 214 at the inner surface 208 of the region/chamber 202. Whether the DEP force attracts or repels nearby micro-objects may depend on parameters such as the frequency of the power source 212 and the dielectric properties of the medium 180 and/or micro-objects (not shown).

The square pattern 220 of the illuminated DEP electrode regions 214a shown in fig. 1C is merely an example. Any pattern of DEP electrode regions 214 may be illuminated (and thus activated) by a light pattern 218 projected into the device 200, and the pattern of the illuminated/activated DEP electrode regions 214 may be repeatedly changed by changing or moving the light pattern 218.

In some embodiments, the electrode activation substrate 206 may include or consist of a photoconductive material. In such embodiments, the inner surface 208 of the electrode activation substrate 206 may be featureless. For example, the electrode activation substrate 206 may include or consist of a hydrogenated amorphous silicon (a-Si: H) layer. H may contain, for example, about 8% to 40% hydrogen (calculated as 100 x the number of hydrogen atoms/total number of hydrogen and silicon atoms). The a-Si: H layer may have a thickness of about 500nm to about 2.0 □ m. In such embodiments, DEP electrode regions 214 may be formed in any pattern anywhere on the inner surface 208 of the electrode activation substrate 206, according to the light pattern 218. Thus, the number and pattern of DEP electrode regions 214 need not be fixed, but may correspond to the light pattern 218. Examples of microfluidic devices having DEP configurations comprising a photoconductive layer (such as those described above) have been described, for example, in U.S. patent No. RE44,711(Wu et al) (originally issued as U.S. patent No. 7,612,355), the entire contents of which are incorporated herein by reference.

In other embodiments, the electrode activation substrate 206 may comprise a substrate comprising a plurality of doped layers, electrically insulating layers (or regions), and electrically conductive layers forming a semiconductor integrated circuit, such as is known in the semiconductor arts. For example, the electrode activation substrate 206 may include a plurality of phototransistors, including, for example, lateral bipolar phototransistors, each corresponding to a DEP electrode region 214. Alternatively, the electrode activation substrate 206 can include electrodes (e.g., conductive metal electrodes) controlled by the phototransistor switches, wherein each such electrode corresponds to a DEP electrode region 214. The electrode activation substrate 206 may include a pattern of such phototransistors or phototransistor-controlled electrodes. For example, the pattern may be an array of substantially square phototransistors or phototransistor-controlled electrodes arranged in rows and columns, as shown in fig. 2B. Alternatively, the pattern may be an array of substantially hexagonal phototransistors or phototransistor-controlled electrodes forming a hexagonal lattice (hexagonal lattice). Regardless of the pattern, the circuit elements can form electrical connections between the DEP electrode regions 214 at the inner surface 208 of the electrode activation substrate 206 and the bottom electrode 210, and those electrical connections (i.e., phototransistors or electrodes) can be selectively activated and deactivated by the light pattern 218. When not activated, each electrical connection can have a high impedance such that the relative impedance through the electrode activation substrate 206 (i.e., from the bottom electrode 204 to the inner surface 208 of the electrode activation substrate 206 interfacing with the dielectric 180 in the region/chamber 202) is greater than the relative impedance through the dielectric 180 at the corresponding DEP electrode region 214 (i.e., from the inner surface 208 of the electrode activation substrate 206 to the top electrode 210 of the cover 110). However, when activated by light in the light pattern 218, the relative impedance through the electrode activation substrate 206 is less than the relative impedance through the medium 180 at each illuminated DEP electrode region 214, thereby activating the DEP electrode at the respective DEP electrode region 214, as described above. Thus, DEP electrodes that attract or repel micro-objects (not shown) in the medium 180 can be selectively activated and deactivated at a number of different DEP electrode regions 214 at the inner surface 208 of the electrode activation substrate 206 in the region/chamber 202, in a manner determined by the light pattern 218.

Examples of microfluidic devices having electrode-activated substrates including phototransistors have been described, for example, in U.S. Pat. No. 7,956,339(Ohta et al) (see, e.g., device 300 shown in fig. 21 and 22 and the description thereof), the entire contents of which are incorporated herein by reference. Examples of microfluidic devices having electrode activated substrates including electrodes controlled by phototransistor switches have been described, for example, in U.S. patent publication No. 2014/0124370(Short et al) (see, e.g., devices 200, 400, 500, 600, and 900 and the description thereof shown throughout the figures), the entire contents of which are incorporated herein by reference.

In some embodiments of DEP configured microfluidic devices, the top electrode 210 is part of a first wall (or lid 110) of the housing 102, and the electrode activation substrate 206 and the bottom electrode 204 are part of a second wall (or support structure 104) of the housing 102. The region/chamber 202 may be located between the first wall and the second wall. In other embodiments, the electrode 210 is part of the second wall (or support structure 104) and one or both of the electrode activation substrate 206 and/or the electrode 210 is part of the first wall (or cover 110). Further, the light source 216 may alternatively be used to illuminate the housing 102 from below.

With the microfluidic device 200 of fig. 1B-1C having a DEP configuration, the motion module 162 can select a micro-object (not shown) in the medium 180 in the region/chamber 202 by projecting a light pattern 218 into the device 200 to activate a first set of one or more DEP electrodes at the DEP electrode region 214a of the inner surface 208 of the electrode activation substrate 206 in a pattern (e.g., a square pattern 220) that surrounds and captures the micro-object. The motion module 162 can then move the captured micro-objects by moving the light pattern 218 relative to the device 200 to activate the second set of one or more DEP electrodes at the DEP electrode region 214. Alternatively, the device 200 may be moved relative to the light pattern 218.

In other embodiments, the microfluidic device 200 can have a DEP configuration that does not rely on photo-activation of DEP electrodes at the inner surface 208 of the electrode activation substrate 206. For example, the electrode activation substrate 206 can include selectively addressable and energizable electrodes located opposite a surface (e.g., the lid 110) that includes at least one electrode. A switch (e.g., a transistor switch in a semiconductor substrate) can be selectively opened and closed to activate or deactivate the DEP electrode at the DEP electrode region 214, thereby creating a net DEP force on a micro-object (not shown) in the region/chamber 202 near the activated DEP electrode. Depending on characteristics such as the frequency of the power source 212 and the dielectric properties of the medium (not shown) and/or micro-objects in the region/chamber 202, DEP forces may attract or repel nearby micro-objects. One or more micro-objects in the region/chamber 202 can be captured and moved in the region/chamber 202 by selectively activating and deactivating a set of DEP electrodes (e.g., at a set of DEP electrode regions 214 forming a square pattern 220). The motion module 162 in fig. 1A can control such switches to activate and deactivate individual DEP electrodes to select, trap, and move specific micro-objects (not shown) around the area/chamber 202. Microfluidic devices having DEP configurations comprising selectively addressable and excitable electrodes are known in the art and have been described, for example, in U.S. patent nos. 6,294,063(Becker et al) and 6,942,776(Medoro), the entire contents of which are incorporated herein by reference.

As yet another example, the microfluidic device 200 can have an Electrowetting (EW) configuration, which can replace the DEP configuration, or can be located in a portion of the microfluidic device 200 separate from the portion having the DEP configuration. The EW configuration can be an electro-wetting configuration or an electro-wetting on dielectric (EWOD) configuration, both of which are known in the art. In some EW configurations, the support structure 104 has an electrode activation substrate 206 sandwiched between a dielectric layer (not shown) and the bottom electrode 204. The dielectric layer may comprise and/or may be coated with a hydrophobic material. For microfluidic devices 200 having the EW configuration, the inner surface 208 of the support structure 104 is the inner surface of the dielectric layer or hydrophobic coating thereof.

The dielectric layer (not shown) may include one or more oxide layers and may have a thickness of about 50nm to about 250nm (e.g., about 125nm to about 175 nm). In certain embodiments, the dielectric layer may include an oxide layer, such as a metal oxide (e.g., aluminum oxide or hafnium oxide) layer. In certain embodiments, the dielectric layer may comprise a dielectric material other than a metal oxide, such as silicon oxide or nitride. Regardless of the exact composition and thickness, the dielectric layer may have an impedance of about 10k ohms to about 50k ohms.

In some embodiments, the surface of the dielectric layer facing inward toward the region/chamber 202 is coated with a hydrophobic material. The hydrophobic material may comprise, for example, carbon fluoride molecules. Examples of fluorinated carbon molecules include perfluoropolymers, such as polytetrafluoroethylene (e.g.,

Figure BDA0002104386230000241

) Or poly (2, 3-difluoromethylene-perfluorotetrahydrofuran) (e.g., CYTOP)TM). Molecules constituting the hydrophobic material may be covalently bonded to the surface of the dielectric layer. For example, molecules of the hydrophobic material may be covalently bonded to the surface of the dielectric layer by means of a linking group such as a siloxane group, a phosphonic acid group or a thiol group. Thus, in some embodiments, the hydrophobic material may comprise an alkyl-terminated siloxane, an alkyl-terminated phosphonic acid, or an alkyl-terminated thiol. The alkyl group can be a long chain hydrocarbon (e.g., a chain having at least 10 carbons or at least 16, 18, 20, 22 or more carbons). Alternatively, fluorinated (or perfluorinated) carbon chains may be used instead of alkyl groups. Thus, for example, the hydrophobic material may comprise a fluoroalkyl terminated siloxane, a fluoroalkyl terminated phosphonic acid, or a fluoroalkyl terminated thiol. In some embodiments, the hydrophobic coating has a thickness of about 10nm to about 50 nm. In other embodiments, the hydrophobic coating has a thickness of less than 10nm (e.g., less than 5nm, or about 1.5 to 3.0 nm).

In some embodiments, the cover 110 of the microfluidic device 200 having an electrowetting configuration is also coated with a hydrophobic material (not shown). The hydrophobic material may be the same hydrophobic material used to coat the dielectric layer of the support structure 104, and the hydrophobic coating may have a thickness that is substantially the same as the thickness of the hydrophobic coating on the dielectric layer of the support structure 104. In addition, the lid 110 may include an electrode activation substrate 206 sandwiched between a dielectric layer and a top electrode 210 in the manner of the support structure 104. The dielectric layers of the electrode activation substrate 206 and the cap 110 may have the same composition and/or dimensions as the dielectric layers of the electrode activation substrate 206 and the support structure 104. Thus, the microfluidic device 200 may have two electrowetting surfaces.

In some embodiments, the electrode activation substrate 206 may include a photoconductive material, such as those described above. Thus, in certain embodiments, the electrode activation substrate 206 may comprise or consist of a hydrogenated amorphous silicon layer (a-Si: H). H may contain, for example, about 8% to 40% hydrogen (calculated as 100 x the number of hydrogen atoms/total number of hydrogen and silicon atoms). The a-Si: H layer may have a thickness of about 500nm to about 2.0 μm. Alternatively, as described above, the electrode activation substrate 206 may include an electrode (e.g., a conductive metal electrode) controlled by a phototransistor switch. Microfluidic devices having electro-optical wetting configurations are known in the art and/or may be constructed with electrode-activated substrates known in the art. For example, U.S. Pat. No. 6,958,132(Chiou et al), the entire contents of which are incorporated herein by reference, discloses a electrowetting configuration having a photoconductive material such as a-Si: H, while U.S. Pat. No. 2014/0124370(Short et al), cited above, discloses an electrode activated substrate having electrodes controlled by phototransistor switches.

Thus, microfluidic device 200 can have a photo-electro-wetting configuration, and light pattern 218 can be used to activate a photoconductive EW region or a photo-responsive EW electrode in electrode activation substrate 206. Such activated EW regions or EW electrodes of the electrode activation substrate 206 can generate electrowetting forces at the inner surface 208 of the support structure 104 (i.e., the inner surface that covers the dielectric layer or hydrophobic coating thereof). By varying the light pattern 218 incident on the electrode-activated substrate 206 (or moving the microfluidic device 200 relative to the light source 216), droplets (e.g., containing an aqueous medium, solution, or solvent) in contact with the inner surface 208 of the support structure 104 may be moved through an immiscible fluid (e.g., an oil medium) present in the region/chamber 202.

In other embodiments, the microfluidic device 200 may have an EWOD configuration, and the electrode activation substrate 206 may include selectively addressable and excitable electrodes that do not rely on light for activation. Thus, the electrode activation substrate 206 can include a pattern of such Electrowetting (EW) electrodes. For example, the pattern can be an array of substantially square EW electrodes arranged in rows and columns, as shown in figure 2B. Alternatively, the pattern can be an array of substantially hexagonal EW electrodes forming a hexagonal lattice of dots. Regardless of the pattern, the EW electrode can be selectively activated (or deactivated) by an electrical switch (e.g., a transistor switch in a semiconductor substrate). By selectively activating and deactivating the EW electrodes in the electrode activation substrate 206, droplets (not shown) in contact with the inner surface 208 of the covered dielectric layer or hydrophobic coating thereof can be moved within the region/chamber 202. The motion module 162 in figure 1A can control such switches to activate and deactivate individual EW electrodes to select and move specific droplets around the region/chamber 202. Microfluidic devices having EWOD configurations of selectively addressable and excitable electrodes are known in the art and have been described, for example, in U.S. patent No. 8,685,344(Sundarsan et al), the entire contents of which are incorporated herein by reference.

Regardless of the configuration of the microfluidic device 200, the power supply 212 may be used to provide a potential (e.g., an AC voltage potential) that powers the circuitry of the microfluidic device 200. The power supply 212 may be the same as or a component of the power supply 192 referenced in FIG. 1. The power supply 212 may be configured to provide an AC voltage and/or current to the top electrode 210 and the bottom electrode 204. For AC voltages, the power supply 212 may provide a range of frequencies and a range of average or peak powers (e.g., voltages or currents): which, as described above, is sufficient to generate a net DEP force (or electrowetting force) strong enough to trap and move individual micro-objects (not shown) in the region/chamber 202, and/or which, as also described above, is sufficient to alter the wetting properties of the inner surface 208 of the support structure 104 (i.e., the dielectric layer and/or the hydrophobic coating on the dielectric layer) in the region/chamber 202. Such frequency ranges and average or peak power ranges are known in the art. See, for example, U.S. Pat. No. 6,958,132(Chiou et al), U.S. Pat. No. RE44,711(Wu et al) (originally issued as U.S. Pat. No. 7,612,355), and U.S. patent application publication Nos. US2014/0124370(Short et al), US2015/0306598(Khandros et al), and US2015/0306599(Khandros et al).

Isolating the dock. Non-limiting examples of general isolation docks 224, 226 and 228 are shown within microfluidic device 230 depicted in fig. 2A-2C. Each isolation dock 224, 226, and 228 may include a separation structure 232 defining a separation region 240 and a connecting region 236 fluidly connecting the separation region 240 to the channel 122. The connecting region 236 may include a proximal opening 234 leading to the channel 122 and a distal opening 238 leading to the separating region 240. The connection region 236 may be configured such that a maximum penetration depth of a flow of fluid medium (not shown) from the channel 122 into the isolation dock 224, 226, 228 does not extend into the separation region 240. Thus, due to the connection region 236, micro-objects (not shown) or other materials (not shown) disposed in the separation region 240 of the isolation docks 224, 226, 228 may be separated from and substantially unaffected by the flow of the medium 180 in the channel 122.

The isolation docks 224, 226 and 228 of fig. 2A-2C each have a single opening that leads directly to the channel 122. The opening of the isolation dock opens laterally from the channel 122. Electrode activation substrate 206 is below both channel 122 and isolation docks 224, 226 and 228. The upper surface of the electrode activation substrate 206 within the housing of the isolation dock (forming the floor of the isolation dock) is disposed at the same or substantially the same level as the upper surface of the electrode activation substrate 206 within the channel 122 (or flow area if no channel is present) (forming the floor of the flow channel (or flow area) of the microfluidic device). The electrode activation substrate 206 may be featureless or may have an irregular or patterned surface that varies from its highest elevation to its lowest depression by less than about 3 microns, 2.5 microns, 2 microns, 1.5 microns, 1 micron, 0.9 microns, 0.8 microns, 0.7 microns, 0.6 microns, 0.5 microns, 0.4 microns, 0.3 microns, 0.2 microns, 0.1 microns, or less. The height variation in the upper surface of the substrate across the channel 122 (or flow region) and isolation dock may be less than about 3%, 2%, 1%, 0.9%, 0.8%, 0.5%, 0.3%, or 0.1% of the height of the wall of the isolation dock or the wall of the microfluidic device. Although the microfluidic device 200 is described in detail, this also applies to any of the microfluidic devices 100, 230, 250, 280, 290 described herein.

Thus, the channel 122 may be an example of a swept area, and the separate area 240 separating the docks 224, 226, 228 may be an instance of an unswept area. It should be noted that the channel 122 and the partitions 224, 226, 228 may be configured to contain one or more fluid media 180. In the example shown in fig. 2A-2B, port 222 is connected to channel 122 and allows for the introduction or removal of fluidic medium 180 into or from microfluidic device 230. The microfluidic device may be loaded with a gas, such as carbon dioxide gas, prior to introduction of the fluid medium 180. Once microfluidic device 230 contains fluidic medium 180, stream 242 of fluidic medium 180 in channel 122 may be selectively generated and stopped. For example, as shown, the ports 222 may be arranged at different locations (e.g., opposite ends) of the channel 122, and a flow 242 of the medium may be formed from one port 222 serving as an inlet to another port 222 serving as an outlet.

Fig. 2C shows a detailed view of an example of an isolated dock 224 according to the present invention. An example of a micro-object 246 is also shown.

As is known, the passage of a flow 242 of fluidic media 180 in the microfluidic channel 122 through the proximal opening 234 of the isolation dock 224 may cause a secondary flow 244 of media 180 to enter and/or exit the isolation dock 224. To separate micro-objects 246 in separation region 240 of isolation dock 224 from secondary flow 244, length L of connection region 236 of isolation dock 224con(i.e., from the proximal opening 234 to the distal opening 238) should be greater than the penetration depth D of the secondary flow 244 into the union region 236p. Depth of penetration D of secondary flow 244pDepending on the velocity of the fluid medium 180 flowing in the channel 122 and various parameters related to the configuration of the channel 122 and the proximal opening 234 to the connection region 236 of the channel 122. Of the channel 122 and opening 234 for a given microfluidic deviceThe configuration will be fixed and the rate of flow 242 of fluid medium 180 in passage 122 will be variable. Thus, for each isolation dock 224, the maximum velocity V of the flow 242 of fluid medium 180 in the channel 122 may be identifiedmaxEnsuring the penetration depth D of the secondary flow 244pNot exceeding the length L of the connecting region 236con. As long as the velocity of the flow 242 of fluid medium 180 in the passage 122 does not exceed the maximum velocity VmaxThe resulting secondary flow 244 may be confined to the channel 122 and the union region 236 and remain outside of the separation region 240. Thus, the flow 242 of medium 180 in the channel 122 will not drag the micro-objects 246 out of the separation region 240. In contrast, micro-objects 246 located in separation region 240 will reside in separation region 240 regardless of the flow 242 of fluid medium 180 in channel 122.

Further, as long as the velocity of the stream 242 of the medium 180 in the channel 122 does not exceed VmaxThe flow 242 of fluid medium 180 in the channel 122 does not move the intermixed particles (e.g., microparticles and/or nanoparticles) from the channel 122 into the separation region 240 of the isolation dock 224. Thus, the length L of the connecting region 236 is madeconGreater than the maximum penetration depth D of the secondary flow 244pOne isolation dock 224 may be prevented from being contaminated by miscellaneous particles from the channel 122 or another isolation dock (e.g., isolation docks 226, 228 in fig. 2D).

Because the channel 122 and the connection region 236 of the isolation docks 224, 226, 228 may be affected by the flow 242 of the medium 180 in the channel 122, the channel 122 and the connection region 236 may be considered a swept (or flow) region of the microfluidic device 230. On the other hand, the separation area 240 of the isolation docks 224, 226, 228 may be considered an unswept (or no-flow) area. For example, a component (not shown) in first fluid medium 180 in channels 122 may mix with second fluid medium 248 in separation zone 240 substantially only by diffusion of the component of first medium 180 from channels 122 through connection zone 236 and into second fluid medium 248 in separation zone 240. Similarly, the components (not shown) of the second media 248 in the separation zone 240 may mix with the first media 180 in the channels 122 substantially only by diffusion of the components of the second media 248 from the separation zone 240, through the connection zone 236, and into the first media 180 in the channels 122. In some embodiments, the degree of fluid medium exchange by diffusion between the separation region and the flow region of the isolation dock is about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more of the total fluid exchange. The first media 180 may be the same media as the second media 248 or a different media. In addition, the first media 180 and the second media 248 may be initially the same and then become different (e.g., by conditioning the second media 248 by separating one or more cells in the region 240, or by changing the media 180 flowing through the channel 122).

As described above, the maximum penetration depth D of the secondary flow 244 caused by the flow 242 of the fluid medium 180 in the passage 122pMay depend on a number of parameters. Examples of such parameters include: the shape of the channel 122 (e.g., the channel may direct media into the connecting region 236, divert media from the connecting region 236, or direct media in a direction substantially perpendicular to the proximal opening 234 to the connecting region 236 of the channel 122); width W of channel 122 at proximal opening 234ch(or cross-sectional area); and the width W of the attachment region 236 at the proximal opening 234con(or cross-sectional area); velocity V of flow 242 of fluid medium 180 in passage 122; the viscosity of the first medium 180 and/or the second medium 248, and so on.

In some embodiments, the dimensions of the channel 122 and isolation docks 224, 226, 228 may be oriented relative to the vector of the flow 242 of fluid medium 180 in the channel 122 as follows: width W of channelch(or cross-sectional area of the channel 122) may be substantially perpendicular to the flow 242 of the medium 180; width W of connecting region 236 at opening 234con(or cross-sectional area) may be substantially parallel to the flow 242 of media 180 in the channel 122; and/or length L of the connecting regionconMay be substantially perpendicular to the flow 242 of the medium 180 in the channel 122. The foregoing are examples only, and the relative positions of the channel 122 and isolation docks 224, 226, 228 may be otherwise oriented with respect to one another.

As shown in FIG. 2C, the width W of the land area 236conFrom the proximal endThe port 234 may be uniform to the distal opening 238. Thus, the width W of the attachment region 236 at the distal opening 238conMay be referred to herein as the width W of the attachment region 236 at the proximal opening 234conAny range identified. Alternatively, the width W of the attachment region 236 at the distal opening 238conMay be greater than the width W of the union region 236 at the proximal opening 234con

As shown in FIG. 2C, the width W of the separation region 240 at the distal opening 238isoMay be connected to the width W of the region 236 at the proximal opening 234conAre substantially the same. Thus, the width W of the separation region 240 at the distal opening 238isoMay be referred to herein as the width W of the attachment region 236 at the proximal opening 234conAny range identified. Alternatively, the width W of the separation region 240 at the distal opening 238isoMay be greater than or less than the width W of the union region 236 at the proximal opening 234con. Further, the distal opening 238 may be smaller than the proximal opening 234, and the width W of the connecting region 236conMay narrow between the proximal opening 234 and the distal opening 238. For example, using a variety of different geometries (e.g., beveling, etc.) the connection region 236 may narrow between the proximal and distal openings. Further, any portion or sub-portion of the connecting region 236 may be narrowed (e.g., a portion of the connecting region adjacent the proximal opening 234).

Fig. 2D-2F depict another exemplary embodiment of a microfluidic device 250 comprising microfluidic tubing 262 and a flow channel 264, which are variations of the respective microfluidic device 100, tubing 132, and channel 134 of fig. 1. The microfluidic device 250 also has a plurality of isolation docks 266, which are additional variations of the isolation docks 124, 126, 128, 130, 224, 226, or 228 described above. In particular, it should be understood that the isolating dock 266 of the device 250 shown in fig. 2D-2F may replace any of the isolating docks 124, 126, 128, 130, 224, 226, or 228 described above in the devices 100, 200, 230, 280, 290, or 320. Similarly, the microfluidic device 250 is another variation of the microfluidic device 100, and may also have the same or different DEP configuration as the microfluidic devices 100, 200, 230, 280, 290, 320 described above, as well as any of the other microfluidic system components described herein.

The microfluidic device 250 of fig. 2D-2F includes a support structure (not visible in fig. 2D-2F, but can be the same as or substantially similar to the support structure 104 of the device 100 depicted in fig. 1A), a microfluidic conduit structure 256, and a lid (not visible in fig. 2D-2F, but can be the same as or substantially similar to the lid 122 of the device 100 depicted in fig. 1A). The microfluidic circuit structure 256 includes a frame 252 and a microfluidic circuit material 260, which may be the same as or substantially similar to the frame 114 and the microfluidic circuit material 116 of the device 100 depicted in fig. 1A. As shown in fig. 2D, the microfluidic circuit 262 defined by the microfluidic circuit material 260 may include a plurality of channels 264 (two are shown, but there may be more) to which a plurality of isolation docks 266 are fluidly connected.

Each isolation dock 266 may include a separation structure 272, a separation region 270 within separation structure 272, and a connection region 268. The connecting region 268 fluidly connects the channel 264 to the separation region 270 from a proximal opening 274 at the channel 264 to a distal opening 276 at the separation structure 272. Generally, the flow 278 of the first fluidic medium 254 in the channel 264 may generate a secondary flow 282 of the first medium 254 from the channel 264 into and/or out of the respective connection regions 268 of the isolation dock 266, in accordance with the discussion of fig. 2B and 2C above.

As shown in fig. 2E, the connection region 268 of each isolation dock 266 generally includes a region extending between a proximal opening 274 to the channel 264 and a distal opening 276 to the separation structure 272. Length L of connecting region 268conMay be greater than the maximum penetration depth D of the secondary flow 282pIn this case, the secondary flow 282 would extend into the union region 268 without being redirected toward the separation region 270 (as shown in FIG. 2D). Alternatively, as shown in FIG. 2F, the connection region 268 may have less than the maximum penetration depth DpLength L ofconIn this case, secondary flow 282 would extend through connection area 268 and be redirected toward separation area 270. In the latter case, the length L of the connecting region 268c1And Lc2And greater than the maximum penetration depth DpSo that the secondary flow 282 do not extend into the separation region 270. Regardless of the length L of the connecting region 268conWhether greater than the penetration depth DpOr length L of connecting region 268c1And Lc2Whether the sum of (A) and (B) is greater than the penetration depth DpNo more than a maximum velocity V of first medium 254 in passage 264maxWill produce a flow 278 having a penetration depth DpAnd micro-objects (not shown, but may be the same as or substantially similar to micro-objects 246 shown in fig. 2C) in separation region 270 of isolation dock 266 are not dragged out of separation region 270 by flow 278 of first medium 254 in channel 264. The flow 278 in the channel 264 also does not drag mixed material (not shown) from the channel 264 into the separation region 270 of the isolation dock 266. As such, diffusion is the only mechanism by which a component in first medium 254 in channel 264 can move from channel 264 into second medium 258 in separation region 270 of isolation dock 266. Similarly, diffusion is the only mechanism by which a component in second medium 258 in separation region 270 of isolation dock 266 can move from separation region 270 into first medium 254 in channel 264. First media 254 may be the same media as second media 258 or first media 254 may be a different media than second media 258. Alternatively, first medium 254 and second medium 258 may be initially the same and then become different, for example, by conditioning the second medium with one or more cells in separation region 270, or by altering the medium flowing through channel 264.

As shown in FIG. 2E, the width W of the channels 264ch(i.e., transverse to the direction of fluid medium flow through the channel, as indicated by arrow 278 in FIG. 2D) may be substantially perpendicular to the width W of the proximal opening 274con1And thus substantially parallel to the width W of the distal opening 276con2. However, width W of proximal opening 274con1And width W of distal opening 276con2And need not be substantially perpendicular to each other. For example, width W of proximal opening 274con1The width W of the shaft (not shown) and distal opening 276 oriented thereoncon2The angle between the further axes oriented thereon may be different from perpendicular and thus not 90 deg.. Examples of selectable angles of orientation includeAn angle in any one of the following ranges: about 30 ° to about 90 °, about 45 ° to about 90 °, about 60 ° to about 90 °, and the like.

In various embodiments of an isolated dock chamber (e.g., 124, 126, 128, 130, 224, 226, 228, or 266), an isolation region (e.g., 240 or 270) is configured to contain a plurality of micro-objects. In other embodiments, the separation region may be configured to contain only one, two, three, four, five, or a similar relatively small number of micro-objects. Thus, the volume of the separation region may be, for example, at least 5 × 105、8×105、1×106、2×106、4×106、6×106Cubic microns or larger.

In various embodiments of the isolation dock, the width W of the channel (e.g., 122) at the proximal opening (e.g., 234)chMay be in any of the following ranges: about 50-1000 microns, 50-500 microns, 50-400 microns, 50-300 microns, 50-250 microns, 50-200 microns, 50-150 microns, 50-100 microns, 70-500 microns, 70-400 microns, 70-300 microns, 70-250 microns, 70-200 microns, 70-150 microns, 90-400 microns, 90-300 microns, 90-250 microns, 90-200 microns, 90-150 microns, 100-300 microns, 100-250 microns, 100-200 microns, 100-150 microns, 100-120 microns. In some other embodiments, the width W of the channel (e.g., 122) at the proximal opening (e.g., 234)chMay be in the range of about 200-800 microns, 200-700 microns, or 200-600 microns. The above are examples only, and the width W of the channel 122chMay be within other ranges (e.g., within a range defined by any of the endpoints listed above). Further, W of channel 122 is in the area of the channel other than the proximal opening that isolates the dockchMay be selected to be within any of these ranges.

In some embodiments, the height of the isolation dock is about 30 to about 200 microns or about 50 to about 150 microns. In some embodiments, the cross-sectional area of the isolation dock is about 1x 104To about 3X 106Square micron, about 2 x 104To about 2X 106Square micron, about 4 x 104To about 1X 106Square micron, about 2 x 104To about 5×105Square micron, about 2 x 104To about 1X 105Square micron and about 2 x 105To about 2X 106Square micron. In some embodiments, the connecting region has a cross-sectional width of about 20 to about 100 microns, about 30 to about 80 microns, or about 40 to about 60 microns.

In various implementations of the isolation dock, the height H of the channel (e.g., 122) at the proximal opening (e.g., 234)chMay be in any of the following ranges: 20-100 microns, 20-90 microns, 20-80 microns, 20-70 microns, 20-60 microns, 20-50 microns, 30-100 microns, 30-90 microns, 30-80 microns, 30-70 microns, 30-60 microns, 30-50 microns, 40-100 microns, 40-90 microns, 40-80 microns, 40-70 microns, 40-60 microns, or 40-50 microns. The foregoing are examples only, and the height H of the channel (e.g., 122)chMay be within other ranges (e.g., ranges defined by any of the endpoints listed above). Height H of channel 122 in the area of the channel other than the proximal opening that isolates the dockchMay be selected to be within any of these ranges.

In various embodiments of the isolation dock, the cross-sectional area of the channel (e.g., 122) at the proximal opening (e.g., 234) can be within any of the following ranges: 500-50,000 square micron, 500-40,000 square micron, 500-30,000 square micron, 500-25,000 square micron, 500-20,000 square micron, 500-15,000 square micron, 500-10,000 square micron, 500-7,500 square micron, 500-5,000 square micron, 1,000-25,000 square micron, 1,000-20,000 square micron, 1,000-15,000 square micron, 1,000-10,000 square micron, 1,000-7,500 square micron, 1,000-5,000 square micron, 2,000-20,000 square micron, 2,000-15,000 square micron, 2,000-10,000 square micron, 2,000-7,500 square micron, 2,000-6,000 square micron, 3,000-20,000 micron, 3,000 square micron, 10,000 square micron, 3,000-3,000 square micron, 3,000 square micron. The foregoing are examples only, and the cross-sectional area of the channel (e.g., 122) at the proximal opening (e.g., 234) can be within other ranges (e.g., within a range defined by any of the endpoints listed above).

In various embodiments of the isolation dock, the length L of the connection region (e.g., 236)conMay be in any of the following ranges: about 20 to about 300 microns, about 40 to about 250 microns, about 60 to about 200 microns, about 80 to about 150 microns, about 20 to about 500 microns, about 40 to about 400 microns, about 60 to about 300 microns, about 80 to about 200 microns, or about 100 to about 150 microns. The foregoing are examples only, and the length L of the connection region (e.g., 236)conMay be in a different range than the preceding examples (e.g., within a range defined by any of the endpoints listed above).

In various embodiments of the isolation dock, the width W of the connection region (e.g., 236) at the proximal opening (e.g., 234)conMay be in any of the following ranges: about 20 to about 150 microns, about 20 to about 100 microns, about 20 to about 80 microns, about 20 to about 60 microns, about 30 to about 150 microns, about 30 to about 100 microns, about 30 to about 80 microns, about 30 to about 60 microns, about 40 to about 150 microns, about 40 to about 100 microns, about 40 to about 80 microns, about 40 to about 60 microns, about 50 to about 150 microns, about 50 to about 100 microns, about 50 to about 80 microns, about 60 to about 150 microns, about 60 to about 100 microns, about 60 to about 80 microns, about 70 to about 150 microns, about 70 to about 100 microns, about 80 to about 150 microns, and about 80 to about 100 microns. The foregoing are examples only, and the width W of the connection region (e.g., 236) at the proximal opening (e.g., 234)conMay be different from the foregoing examples (e.g., within a range defined by any of the endpoints listed above).

In various embodiments of the isolation dock, the width W of the connection region (e.g., 236) at the proximal opening (e.g., 234)conMay be at least as large as the largest dimension of the micro-object (e.g., biological cell, which may be an immune cell, such as a B cell or T cell, or hybridoma cell, etc.) for which the sequestration dock is intended. For example, the width W of the connection region 236 at the proximal opening 234 of the isolation dock where immune cells (e.g., B cells) will be placedconAny of the following widths may be used: about 20 microns, about 25 microns, about 30 microns, about 35 microns, about 40 microns, about 45 microns, about 50 microns, about 55 microns, about 60 microns, about 65 microns, about 70 micronsRice, about 75 microns, or about 80 microns. The foregoing are examples only, and the width W of the connection region (e.g., 236) at the proximal opening (e.g., 234)conMay be different from the foregoing examples (e.g., within a range defined by any of the endpoints listed above).

In various embodiments of the isolation dock, the length L of the connection region (e.g., 236)conWidth W of the connection region (e.g., 236) at the proximal opening 234conThe ratio of (d) may be greater than or equal to any of the following ratios: 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0, or more. The above are examples only, and the length L of the connecting region 236conAnd the width W of the attachment region 236 at the proximal opening 234conThe ratio of (d) may be different from the previous examples.

In various embodiments of the microfluidic devices 100, 200, 230, 250, 280, 290, 320, VmaxMay be set to about 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 μ L/sec.

In various embodiments of microfluidic devices with isolation docks, the volume of the isolation region (e.g., 240) of the isolation dock can be, for example, at least 5 x 105、8×105、1×106、2×106、4×106、6×106、8×106、1×107Cubic microns or larger. In various embodiments of microfluidic devices with isolation docks, the volume of the isolation dock may be about 5 x 105、6×105、8×105、1×106、2×106、4×106、8×106、1×107Cubic microns or larger. In some other embodiments, the volume of the isolating dock can be about 0.5 nanoliter to about 10 nanoliters, about 1.0 nanoliter to about 5.0 nanoliters, about 1.5 nanoliters to about 4.0 nanoliters, about 2.0 nanoliters to about 3.0 nanoliters, or about 2.5 nanoliters, or any range defined by the two aforementioned endpoints.

In various embodiments, the microfluidic device has isolated docks configured as in any of the embodiments discussed herein, wherein the microfluidic device has about 5 to about 10 isolated docks, about 10 to about 50 isolated docks, about 100 to about 500 isolated docks; about 200 to about 1000 isolated docks, about 500 to about 1500 isolated docks, about 1000 to about 2000 isolated docks, or about 1000 to about 3500 isolated docks. The isolation docks need not all be the same size and may include multiple configurations (e.g., different widths, different features within the isolation dock).

In some other embodiments, the isolation dock of the microfluidic device is configured as any of the embodiments discussed herein, wherein the microfluidic device has from about 1500 to about 3000 isolation docks, from about 2000 to about 3500 isolation docks, from about 2500 to about 4000 isolation docks, from about 3000 to about 4500 isolation docks, from about 3500 to about 5000 isolation docks, from about 4000 to about 5500 isolation docks, from about 4500 to about 6000 isolation docks, from about 5000 to about 6500 isolation docks, from about 5500 to about 7000 isolation docks, from about 6000 to about 7500 isolation docks, from about 6500 to about 8000 isolation docks, from about 7000 to about 8500 isolation docks, from about 7500 to about 9000 isolation docks, from about 8000 to about 9500 isolation docks, from about 8500 to about 10,000 isolation docks, from about 9000 to about 10,500 isolation docks, from about 11,000 isolation docks, from about 9500 to about 9500 isolation docks, from about 11,000 to about 500,000 isolation docks, from about 11,000 isolation docks, from about 11,500 to about 11,000 isolation docks, from about 11,12 to about 500,13, from about 500,000 isolation docks, from about 11,000 isolation docks, from about 500,13, from about 500,000 isolation docks, and about 500,000 isolation docks, About 12,500 to about 14,000 isolated docks, about 13,000 to about 14,500 isolated docks, about 13,500 to about 15,000 isolated docks, about 14,000 to about 15,500 isolated docks, about 14,500 to about 16,000 isolated docks, about 15,000 to about 16,500 isolated docks, about 15,500 to about 17,000 isolated docks, about 16,000 to about 17,500 isolated docks, about 16,500 to about 18,000 isolated docks, about 17,000 to about 18,500 isolated docks, about 17,500 to about 19,000 isolated docks, about 18,000 to about 19,500 isolated docks, about 18,500 to about 20,000 isolated docks, about 19,000 to about 20,500 isolated docks, about 19,500 to about 21,000 isolated docks, or about 20,000 to about 21,500 isolated docks.

Fig. 2G shows a microfluidic device 280 according to an embodiment. The microfluidic device 280 shown in fig. 2G is a stylized schematic of the microfluidic device 100. In the implementation of the method, the first step of the method,the microfluidic device 280 and its constituent plumbing components (e.g., channel 122 and isolation dock 128) will have dimensions as discussed herein. The microfluidic circuit 120 shown in fig. 2G has two ports 107, four different channels 122, and four different flow paths 106. Microfluidic device 280 further includes a plurality of isolation docks that open into each channel 122. In the microfluidic device shown in fig. 2G, the isolation dock has a similar geometry to the dock shown in fig. 2C, and thus has both a connection region and a separation region. Thus, microfluidic circuit 120 includes a swept area (e.g., channel 122 and connecting region 236 at maximum penetration depth D of secondary flow 244pInner portion) and non-swept areas (e.g., separation area 240 and union area 236 are not at the maximum penetration depth D of the secondary stream 244pInner portion).

Fig. 3A-3B illustrate various embodiments of a system 150 that can be used to operate and view microfluidic devices (e.g., 100, 200, 230, 280, 250, 290, 320) according to the present invention. As shown in fig. 3A, the system 150 may include a structure ("nest") 300 configured to hold a microfluidic device 100 (not shown) or any other microfluidic device described herein. Nest 300 can include a socket 302 that can interface with a microfluidic device 320 (e.g., a light-actuated electrokinetic device 100) and provide an electrical connection from power source 192 to microfluidic device 320. Nest 300 may also include an integrated electrical signal generation subsystem 304. The electrical signal generation subsystem 304 may be configured to provide a bias voltage to the receptacle 302 such that when the receptacle 302 holds the microfluidic device 320, a bias voltage is applied across a pair of electrodes in the microfluidic device 320. Thus, the electrical signal generation subsystem 304 may be part of the power supply 192. The ability to apply a bias voltage to the microfluidic device 320 does not mean that the bias voltage is always applied when the socket 302 holds the microfluidic device 320. In contrast, in most cases, the bias voltage will be applied intermittently, e.g., only when needed to facilitate generation of an electrokinetic force (e.g., dielectrophoresis or electrowetting) in the microfluidic device 320.

As shown in fig. 3A, nest 300 may include a Printed Circuit Board Assembly (PCBA) 322. The electrical signal generation subsystem 304 may be mounted on the PCBA 322 and electrically integrated therein. The example support also includes a socket 302 mounted on the PCBA 322.

Typically, electrical signal generation subsystem 304 will include a waveform generator (not shown). The electrical signal generation subsystem 304 can also include an oscilloscope (not shown) and/or a waveform amplification circuit (not shown) configured to amplify waveforms received from the waveform generator. The oscilloscope (if any) may be configured to measure the waveform supplied to the microfluidic device 320 held by the receptacle 302. In certain embodiments, the oscilloscope measures the waveform at a location near the microfluidic device 320 (and away from the waveform generator), thereby ensuring a more accurate measurement of the waveform actually applied to the device. Data obtained from oscilloscope measurements may be provided, for example, as feedback to a waveform generator, and the waveform generator may be configured to adjust its output based on such feedback. An example of a suitable combined waveform generator and oscilloscope is Red PitayaTM

In certain embodiments, nest 300 further includes a controller 308, such as a microprocessor for detecting and/or controlling electrical signal generating subsystem 304. Examples of suitable microprocessors include ArduinoTMMicroprocessors, e.g. ArduinonanoTM. The controller 308 may be used to perform functions and analyses or may communicate with the external master controller 154 (shown in FIG. 1A) to perform functions and analyses. In the embodiment shown in fig. 3A, the controller 308 communicates with the master controller 154 through an interface 310 (e.g., a plug or connector).

In some embodiments, nest 300 may include an electrical signal generation subsystem 304, which includes Red PitayaTMA waveform generator/oscilloscope cell ("Red Pitaya cell") and a waveform amplification circuit that amplifies the waveform generated by the Red Pitaya cell and transmits the amplified voltage to the microfluidic device 100. In some embodiments, the Red Pitaya cell is configured to measure the amplified voltage at the microfluidic device 320 and then adjust its own output voltage as needed so that the voltage measured at the microfluidic device 320 is a desired value. In some embodiments, waveform amplificationThe circuit may have a +6.5V to-6.5V power supply generated by a pair of DC-DC converters mounted on the PCBA 322, producing a signal at the microfluidic device 100 of up to 13 Vpp.

As shown in fig. 3A, the support structure 300 may further include a thermal control subsystem 306. The thermal control subsystem 306 may be configured to regulate the temperature of the microfluidic device 320 held by the support structure 300. For example, the thermal control subsystem 306 may include a Peltier thermoelectric device (not shown) and a cooling unit (not shown). The Peltier thermoelectric device may have a first surface configured to interface with at least one surface of the microfluidic device 320. The cooling unit may be, for example, a cooling block (not shown), such as a liquid cooled aluminum block. A second surface (e.g., a surface opposite the first surface) of the Peltier thermoelectric device may be configured to interface with a surface of such a cooling block. The cooling block may be connected to a fluid path 314, the fluid path 314 configured to circulate a cooled fluid through the cooling block. In the embodiment shown in fig. 3A, the support structure 300 includes an inlet 316 and an outlet 318 to receive cooled fluid from an external reservoir (not shown), introduce the cooled fluid into the fluid path 314 and through the cooling block, and return the cooled fluid to the external reservoir. In some embodiments, a Peltier thermoelectric device, a cooling unit, and/or a fluid path 314 may be mounted on the housing 312 of the support structure 300. In some embodiments, the thermal control subsystem 306 is configured to regulate the temperature of the Peltier thermoelectric device to achieve a target temperature for the microfluidic device 320. Temperature regulation of the Peltier thermoelectric device can be effected, for example, by a thermoelectric power supply, for example, by PololuTMThermoelectric power supply (Pololu semiconductors and Electronics Corp.) was implemented. The thermal control subsystem 306 may include feedback circuitry, such as temperature values provided by analog circuitry. Alternatively, the feedback circuit may be provided by a digital circuit.

In some embodiments, nest 300 may include a thermal control subsystem 306 having a feedback circuit that is an analog voltage divider including a resistor (e.g., having an impedance of 1k ohms +/-0.1%, +/-0.02ppm/C0 temperature coefficient) and an NTC thermistor (e.g., having a nominal impedance of 1k ohms +/-0.01%)An electrical circuit (not shown). In some cases, thermal control subsystem 306 measures the voltage from the feedback circuit and then uses the calculated temperature value as an input to the on-board PID control loop algorithm. The output from the PID control loop algorithm may drive, for example, PololuTMDirectional and pulse width modulated signal pins on a motor driver (not shown) to actuate the thermoelectric power supply to control the Peltier thermoelectric device.

Nest 300 may include a serial port 324 that allows the microprocessor of controller 308 to communicate with external master controller 154 via interface 310 (not shown). Additionally, the microprocessor of the controller 308 may be in communication with the electrical signal generation subsystem 304 and the thermal control subsystem 306 (e.g., via a Plink tool (not shown)). Thus, the electrical signal generation subsystem 304 and the thermal control subsystem 306 may communicate with the external master controller 154 via a combination of the controller 308, the interface 310, and the serial port 324. In this manner, the main controller 154 may assist the electrical signal generation subsystem 304 by performing, among other things, scaling calculations for output voltage regulation. A Graphical User Interface (GUI) (not shown) provided via a display device 170 coupled to the external master controller 154 may be configured to plot the temperature and waveform data obtained from the thermal control subsystem 306 and the electrical signal generation subsystem 304, respectively. Alternatively or additionally, the GUI may allow for updating the controller 308, the thermal control subsystem 306, and the electrical signal generation subsystem 304.

As discussed above, the system 150 may include an imaging device 194. In some embodiments, the imaging device 194 includes a light modulation subsystem 330 (see fig. 3B). The light modulation subsystem 330 may include a Digital Mirror Device (DMD) or a micro-shutter array system (MSA), either of which may be configured to receive light from the light source 332 and transmit a portion of the received light into the optical train of the microscope 350. Alternatively, light modulation subsystem 330 may include a device that generates its own light (and thus does not require light source 332), such as an organic light emitting diode display (OLED), a Liquid Crystal On Silicon (LCOS) device, a ferroelectric liquid crystal on silicon device (FLCOS), or a transmissive Liquid Crystal Display (LCD). The light modulation subsystem 330 may beSuch as a projector. Thus, the light modulation subsystem 330 is capable of emitting structured light and unstructured light. One example of a suitable light modulation subsystem 330 is from Andor TechnologiesTMMosaic ofTMProvided is a system. In certain embodiments, the imaging module 164 and/or the motion module 162 of the system 150 may control the light modulation subsystem 330.

In certain embodiments, the imaging device 194 further comprises a microscope 350. In such embodiments, the nest 300 and the light modulation subsystem 330 may be individually configured to be mounted on the microscope 350. The microscope 350 may be, for example, a standard research grade optical microscope or a fluorescent microscope. Thus, the nest 300 can be configured to mount on the stage 344 of the microscope 350 and/or the light modulation subsystem 330 can be configured to mount on a port of the microscope 350. In other embodiments, the nest 300 and the light modulation subsystem 330 described herein may be integrated components of the microscope 350.

In certain embodiments, the microscope 350 may further include one or more detectors 348. In some embodiments, detector 348 is controlled by imaging module 164. Detector 348 may include an eyepiece, a Charge Coupled Device (CCD), a camera (e.g., a digital camera), or any combination thereof. If there are at least two detectors 348, one detector may be, for example, a fast frame rate camera and the other detector may be a high sensitivity camera. Further, the microscope 350 can include an optical train configured to receive light reflected and/or emitted from the microfluidic device 320 and focus at least a portion of the reflected and/or emitted light onto the one or more detectors 348. The optical train of the microscope may also include different tube lenses (not shown) for different detectors so that the final magnification on each detector may be different.

In certain embodiments, the imaging device 194 is configured to use at least two light sources. For example, a first light source 332 may be used to generate structured light (e.g., via the light modulation subsystem 330), and a second light source 334 may be used to provide unstructured light. The first light source 332 may generate structured light for optically driven electrical motion and/or fluorescence excitation, and the second light source 334 may be used to provide bright field illumination. In these embodiments, the motion module 164 may be used to control the first light source 332 and the imaging module 164 may be used to control the second light source 334. The optical train of the microscope 350 can be configured to (1) receive structured light from the light modulation subsystem 330 and focus the structured light on at least a first area in a microfluidic device (e.g., a light-actuated electro-kinetic device) when the device is held by the nest 300, and (2) receive light reflected and/or emitted from the microfluidic device and focus at least a portion of such reflected and/or emitted light onto the detector 348. The optical train can be further configured to receive unstructured light from the second light source and focus the unstructured light on at least a second area of the microfluidic device when the microfluidic device is held by the nest 300. In certain embodiments, the first and second regions of the microfluidic device may be overlapping regions. For example, the first region may be a subset of the second region.

In fig. 3B, the first light source 332 is shown providing light to the light modulation subsystem 330, which provides structured light to an optical train (not shown) of the microscope 350 of the system 355. The second light source 334 is shown providing unstructured light to the optical train via a beam splitter 336. The structured light from the light modulation subsystem 330 and the unstructured light from the second light source 334 travel together through an optical train from the beam splitter 336 to a second beam splitter (or dichroic filter 338, depending on the light provided by the light modulation subsystem 330), where the light is reflected down through the objective lens 336 to the sample plane 342. Light reflected and/or emitted from sample plane 342 is then returned back up through objective lens 340, through beam splitter and/or dichroic filter 338, and back to dichroic filter 346. Only a portion of the light that reaches the dichroic filter 346 passes through to the detector 348.

In some embodiments, the second light source 334 emits blue light. With an appropriate dichroic filter 346, blue light reflected from the sample plane 342 can pass through the dichroic filter 346 and reach a detector 348. In contrast, structured light from the light modulation subsystem 330 reflects from the sample plane 342, but does not pass through the dichroic filter 346. In this example, the dichroic filter 346 filters out visible light having a wavelength longer than 495 nm. This filtering of light from the light modulation subsystem 330 is accomplished (as shown) only if the light emitted from the light modulation subsystem does not include any wavelengths shorter than 495 nm. In an implementation, if the light from light modulation subsystem 330 includes wavelengths shorter than 495nm (e.g., a blue wavelength), some of the light from the light modulation subsystem passes through filter 346 to detector 348. In such embodiments, the filter 346 acts to change the balance between the amount of light reaching the detector 348 from the first light source 332 and the second light source 334. This may be beneficial if the first light source 332 is significantly stronger than the second light source 334. In other embodiments, the second light source 334 may emit red light, and the dichroic filter 346 may filter out visible light other than red light (e.g., visible light having a wavelength shorter than 650 nm).

Coating solution and coating agent. Without wishing to be bound by theory, when one or more interior surfaces of the microfluidic device have been conditioned or coated so as to present a layer of organic and/or hydrophilic molecules that provides a primary interface between the microfluidic device and the micro-objects (e.g., biological cells) maintained therein, culturing of biological cells (e.g., immune cells, such as B cells or T cells) within the microfluidic device may be facilitated (i.e., the micro-objects exhibit increased viability, greater expansion, and/or greater portability within the microfluidic device). In some embodiments, one or more internal surfaces of the microfluidic device (e.g., an internal surface of an electrode-activated substrate of the DEP-configured microfluidic device, a surface of a lid and/or tubing material of the microfluidic device) are treated with a coating solution and/or a coating agent to produce the desired layer of organic and/or hydrophilic molecules. In some embodiments, the micro-objects (e.g., biological cells) that are cultured in the microfluidic device and optionally allowed to expand are input into a coating solution comprising one or more coating agents.

In other embodiments, the interior surface of a microfluidic device (e.g., a DEP configured microfluidic device) is treated or "primed" with a coating solution comprising a coating agent prior to introducing the micro-objects (e.g., biological cells) into the microfluidic device. Any convenient coating agent/paint may be usedCoating solutions, including but not limited to: serum or serum factors, Bovine Serum Albumin (BSA), polymers, detergents, enzymes, and any combination thereof. In some specific embodiments, the coating agent will be used to treat the interior surfaces of the microfluidic device. In one example, a polymer containing alkylene ether moieties is included in the coating solution as a coating agent. Many alkylene ether containing polymers may be suitable. One non-limiting exemplary class of alkylene ether-containing polymers is the amphoteric nonionic block copolymers, which comprise blocks of Polyoxyethylene (PEO) and polyoxypropylene (PPO) subunits having different proportions and positions within the polymer chain.

Figure BDA0002104386230000411

Polymers (BASF) are such block copolymers and are known in the art to be suitable for use when in contact with living cells. Average molecular weight M of the PolymerwAnd may range from about 2000Da to about 20 KDa. In some embodiments, the PEO-PPO block copolymer may have a hydrophilic-lipophilic balance (HLB) of greater than about 10 (e.g., 12-18). Specific for producing coated surfaces

Figure BDA0002104386230000412

The polymer comprisesL44, L64, P85 and F127 (including F127 NF). Another class of alkylene ether-containing polymers is polyethylene glycol (PEG M)w<100,000Da) or alternatively, polyoxyethylene (PEO, M)w>100,000). In some embodiments, the PEG can have an M of about 1000Da, 5000Da, 10,000Da, or 20,000Daw

In some embodiments, the coating solution may comprise a plurality of proteins and/or peptides as coating agents. In a particular embodiment, the coating solution used in the present disclosure comprises as coating agent a protein, such as albumin (e.g. BSA) and/or a serum (or a combination of different sera) comprising albumin and/or one or more other similar proteins. The serum may be from any convenient source, including but not limited to fetal bovine serum, sheep serum, goat serum, horse serum, and the like. In certain embodiments, BSA is present in a blocking solution (blocking solution) at a concentration ranging from about 1mg/mL to about 100mg/mL, including 5mg/mL, 10mg/mL, 20mg/mL, 30mg/mL, 40mg/mL, 50mg/mL, 60mg/mL, 70mg/mL, 80mg/mL, 90mg/mL, or any value therebetween. In certain embodiments, serum may be present in the coating solution at a concentration ranging from about 20% (v/v) to about 50% v/v, including 25%, 30%, 35%, 40%, 45%, or higher or any value in between. In some embodiments, BSA may be present as a coating agent in the coating solution at 5mg/mL, while in other embodiments, BSA may be present as a coating agent in the coating solution at 70 mg/mL. In certain embodiments, serum is present at 30% in the coating solution as a coating agent.

And (3) coating materials. Depending on the embodiment, any of the above-described coating agents/coating solutions can be replaced with, or used in combination with, a variety of coating materials for coating one or more interior surfaces of a microfluidic device (e.g., a DEP-configured and/or EW-configured microfluidic device). In some embodiments, at least one surface of the microfluidic device comprises a coating material that provides an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells). In some embodiments, substantially all of the interior surfaces of the microfluidic device comprise a coating material. The coated interior surface may include a flow region (e.g., a channel), a surface of a chamber or isolation dock, or a combination thereof. In some embodiments, each of the plurality of isolated docks has at least one interior surface coated with a coating material. In other embodiments, each of the plurality of flow regions or channels has at least one interior surface coated with a coating material. In some embodiments, at least one interior surface of each of the plurality of isolated docks and each of the plurality of channels is coated with a coating material.

A polymer-based coating material. At least one of the inner surfaces may include a coating material comprising a polymer. The polymer may be covalently or non-covalently bound (or linked) to at least one surface. The polymers can have a variety of structural motifs such as found in block (and copolymers), star (star copolymers), and graft or comb (graft copolymers), all of which can be adapted for use in the methods disclosed herein.

The polymer may comprise a polymer comprising alkylene ether moieties. A wide variety of alkylene ether-containing polymers may be suitable for use in the microfluidic devices described herein. One non-limiting exemplary class of alkylene ether-containing polymers is the amphoteric nonionic block copolymers, which comprise blocks of Polyoxyethylene (PEO) and polyoxypropylene (PPO) subunits having different proportions and positions within the polymer chain.

Figure BDA0002104386230000421

Polymers (BASF) are such block copolymers and are known in the art to be suitable for use when in contact with living cells. Average molecular weight M of the PolymerwFrom about 2000Da to about 20 kDa. In some embodiments, the PEO-PPO block copolymer may have a hydrophilic-lipophilic balance (HLB) of greater than about 10 (e.g., 12-18). Specific for producing coated surfaces

Figure BDA0002104386230000422

The polymer comprises

Figure BDA0002104386230000423

L44, L64, P85 and F127 (including F127 NF). Another class of alkylene ether-containing polymers is polyethylene glycol (PEG M)w<100,000Da) or alternatively, polyoxyethylene (PEO, M)w>100,000). In some embodiments, the PEG can have an M of about 1000Da, 5000Da, 10,000Da, or 20,000Daw

In other embodiments, the coating material may include a polymer containing carboxylic acid moieties. The carboxylic acid subunits may be subunits containing alkyl, alkenyl or aromatic moieties. One non-limiting example is polylactic acid (PLA).

In other embodiments, the coating material may include a polymer containing sulfonic acid moieties. The sulfonate subunits may be subunits containing alkyl, alkenyl or aromatic moieties. One non-limiting example is polystyrene sulfonic acid (PSSA) or polyanetholesulfonic acid. These latter exemplary polymers are polyelectrolytes and may alter the characteristics of the surface to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects, such as cells, e.g., immune cells (e.g., B cells) or hybridoma cells.

In some embodiments, the coating material may include a urethane moiety-containing polymer, such as, but not limited to, a polyurethane.

In other embodiments, the coating material may include a polymer that includes a phosphate moiety at a terminus of or pendant from the backbone of the polymer.

In other embodiments, the coating material may include a polymer containing sugar moieties. In one non-limiting example, a polysaccharide (e.g., derived from seaweed or a fungal polysaccharide, such as xanthan or dextran) may be suitable for forming a material that can reduce or prevent cell adhesion in a microfluidic device. For example, dextran polymers having a size of about 3kDa may be used to provide a coating material for surfaces within a microfluidic device.

In other embodiments, the coating material may comprise a polymer containing nucleotide moieties, i.e., nucleic acids, which may have ribonucleotide moieties or deoxyribonucleotide moieties. Nucleic acids may contain only natural nucleotide moieties or may contain non-natural nucleotide moieties that comprise nucleobase, ribose, or phosphate moiety analogs, such as, but not limited to, 7-deazaadenine, pentose, methylphosphonate, or phosphorothioate moieties. The nucleic acid-containing polymer may comprise a polyelectrolyte that may provide a layer of organic and/or hydrophilic molecules suitable for maintaining and/or expanding biological micro-objects, such as cells, e.g. immune cells (e.g. B-cells) or hybridoma cells.

In other embodiments, the coating material may include a polymer containing amino acid moieties. Polymers containing amino acid moieties may include polymers containing natural amino acids or polymers containing unnatural amino acids, which may each include peptides, polypeptides, or proteins. In one non-limiting example, the protein can be Bovine Serum Albumin (BSA). In some embodiments, extracellular matrix (ECM) proteins may be provided within the coating material for obtaining optimized cell adhesion to promote cell growth. Cell matrix proteins that may be included in the coating material may include, but are not limited to, collagen, elastin, RGD-containing peptides (e.g., fibronectin), or laminin. In other embodiments, growth factors, cytokines, hormones, or other cell signaling substances may be provided within the coating material of the microfluidic device.

In further embodiments, the coating material can include a polymer containing amine moieties. The polyamino polymer may comprise a natural polyamine polymer or a synthetic polyamine polymer. Examples of natural polyamines include spermine, spermidine and putrescine.

In some embodiments, the coating material can include a polymer that includes more than one of an alkylene oxide moiety, a carboxylic acid moiety, a sulfonic acid moiety, a phosphate moiety, a sugar moiety, a nucleotide moiety, or an amino acid moiety. In other embodiments, the polymer conditioned surface may comprise a mixture of more than one polymer, each polymer having alkylene oxide moieties, carboxylic acid moieties, sulfonic acid moieties, phosphate moieties, sugar moieties, nucleotide moieties, and/or amino acid moieties, which may be incorporated into the coating material independently or simultaneously.

A covalently linked coating material. In some embodiments, at least one internal surface comprises covalently linked molecules that provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) within a microfluidic device to provide a conditioned surface for such cells. The covalently linked molecules include a linking group, wherein the linking group is covalently linked to one or more surfaces of the microfluidic device. The linking group is also covalently attached to a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell). The surface to which the linking group is attached can comprise a substrate surface of a microfluidic device, which can comprise silicon and/or silicon dioxide for embodiments in which the microfluidic device comprises a DEP configuration. In some embodiments, the covalently attached coating material coats substantially all of the interior surfaces of the microfluidic device.

In some embodiments, a covalently attached moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological microbial object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell) may include an alkyl or fluoroalkyl (including perfluoroalkyl) moiety; mono-or polysaccharides (which may include, but are not limited to, dextran); alcohols (including but not limited to propargyl alcohol); polyols, including but not limited to polyvinyl alcohol; alkylene ethers including, but not limited to, polyethylene glycol; polyelectrolytes (including but not limited to polyacrylic acid or polyvinylphosphonic acid); amino groups (including derivatives thereof such as, but not limited to, alkylated amines, hydroxyalkylated amino groups, guanidinium salts, and heterocyclic groups containing a non-aromatic nitrogen ring atom such as, but not limited to, morpholinyl or piperazinyl); carboxylic acids, including but not limited to propiolic acid (which may provide a carboxylate anionic surface); phosphonic acids, including but not limited to ethynylphosphonic acid (which can provide a phosphonate anionic surface); a sulfonate anion; a carboxybetaine; a sulfobetaine; sulfamic acid; or an amino acid.

The covalently linked moieties configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) in a microfluidic device can be any of the polymers described herein, and can include polymers comprising alkylene oxide moieties, carboxylic acid moieties, sugar moieties, sulfonic acid moieties, phosphate moieties, amino acid moieties, nucleotide moieties, or amino moieties.

In other embodiments, a moiety configured to provide covalent attachment of an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell) in a microfluidic device can include a non-polymeric moiety, such as an alkyl moiety, a substituted alkyl moiety (e.g., a fluoroalkyl moiety (including but not limited to a perfluoroalkyl moiety)), an amino acid moiety, an alcohol moiety, an amino moiety, a carboxylic acid moiety, a phosphonic acid moiety, a sulfonic acid moiety, an aminosulfonic acid moiety, or a sugar moiety.

In some embodiments, the covalently linked moiety can be an alkyl group comprising carbon atoms that form a straight chain (e.g., a straight chain of at least 10 carbons or at least 14, 16, 18, 20, 22 or more carbons). Thus, the alkyl group may be an unbranched alkyl group. In some embodiments, alkyl groups may include substituted alkyl groups (e.g., some carbons in an alkyl group may be fluorinated or perfluorinated). The alkyl group can include a linear chain of substituted (e.g., fluorinated or perfluorinated) carbons bonded to an unsubstituted carbon. For example, an alkyl group can include a first segment (which can include a perfluoroalkyl group) that is linked to a second segment (which can include an unsubstituted alkyl group). The first and second segments may be linked together directly or indirectly (e.g., via an ether linkage). The first segment of the alkyl group may be located distal to the linking group and the second segment of the alkyl group may be located proximal to the linking group. In other embodiments, the alkyl group may include branched alkyl groups, and may also have one or more arylene groups interrupting the alkyl backbone of the alkyl group. In some embodiments, the branched or arylene interrupted portion of the alkyl or fluoroalkyl group is located distal to the linking group to the surface and the covalent bond.

In other embodiments, the covalently linked moiety may comprise at least one amino acid, which may comprise more than one type of amino acid. Thus, the covalently linked moiety may comprise a peptide or a protein. In some embodiments, the covalently linked moiety may comprise an amino acid, which may provide a zwitterionic surface to support cell growth, viability, portability (portal), or any combination thereof.

The covalently linked moiety may comprise one or more sugars. The covalently linked sugar may be a monosaccharide, disaccharide or polysaccharide. The covalently linked sugar may be modified to introduce reactive pairing moieties that allow coupling or processing for attachment of the surface. Exemplary reactive partner moieties may include aldehyde, alkyne, or halogen moieties. The polysaccharide may be modified in a random manner, wherein each saccharide monomer or only a portion of the saccharide monomers within the polysaccharide may be modified to provide reactive partner moieties that may be coupled directly or indirectly to a surface. One example may include dextran polysaccharides, which may be indirectly coupled to a surface via an unbranched linker moiety.

The covalently linked moiety may include one or more amino groups. The amino group can be a substituted amine moiety, guanidine moiety, nitrogen-containing heterocyclic moiety, or heteroaryl moiety. The amino-containing moiety can have a structure that allows for pH modification of the environment within the microfluidic device and optionally within the sequestration dock and/or flow region (e.g., channel).

The coating material may comprise only one type of covalently linked moiety, or may comprise more than one different type of covalently linked moiety. For example, a fluoroalkyl-conditioned surface (including perfluoroalkyl) may have a plurality of covalently attached moieties that are all the same, e.g., having the same attachment group and covalent attachment to the surface, the same total length, and the same number of fluoromethylene units, including fluoroalkyl moieties. Alternatively, the coating material may have more than one type of covalently linked moiety attached to the surface. For example, the coating material may include molecules having covalently linked alkyl or fluoroalkyl moieties having a specified number of methylene or fluoromethylene units, and may also include another group of molecules having charged moieties covalently linked to alkyl or fluoroalkyl chains having a greater number of methylene or fluoromethylene units. In some embodiments, a coating material having more than one covalently attached moiety can be designed such that a first set of molecules having a greater number of backbone atoms and thus a longer distance from covalent attachment to the surface can provide the ability to present a larger portion on the coated surface, while a second set of molecules having different, less spatially demanding ends and fewer backbone atoms can help to functionalize the entire substrate surface, preventing undesired adhesion or contact with the silicon or aluminum oxide comprising the substrate itself. In another example, the covalently linked moieties can provide a zwitterionic surface that exhibits alternating charges on the surface in a random manner.

Conditioned surface properties. In some embodiments, the covalently linked moieties can form a monolayer when covalently linked to a surface of the microfluidic device (e.g., a substrate surface of a DEP configuration). In some embodiments, the conditioned surface formed by covalently linked moieties can have a thickness of less than 10nm (e.g., less than 5nm, or about 1.5 to 3.0 nm). In other embodiments, the conditioned surface formed by covalently linked moieties may have a thickness of about 10nm to about 50 nm. In some embodiments, the conditioned surface does not require a perfectly formed monolayer to function properly for operation within a DEP configured microfluidic device.

In various embodiments, the coating material of the microfluidic device can provide desired electrical properties. Without wishing to be bound by theory, one factor that affects the robustness of a surface coated with a particular coating material is inherent charge trapping. Different coating materials may trap electrons, which may lead to destruction of the coating material. Defects in the coating material may increase charge trapping and lead to further damage of the coating material. Similarly, different coating materials have different dielectric strengths (i.e., minimum applied electric field that results in dielectric breakdown), which may affect charge trapping. In certain embodiments, the coating material can have a bulk structure (e.g., a close-packed monolayer structure) that reduces or limits the amount of charge trapping.

In addition to the composition of the coating material, other factors, such as the physical (and electrical) thickness of the coating material, can influence the generation of DEP and electrowetting forces by the substrate of the microfluidic device. Various factors may alter the physical and electrical thickness of the coating material, including the manner in which the coating material is deposited on the substrate (e.g., vapor deposition, liquid deposition, spin coating, and electrostatic coating). The physical thickness and uniformity of the coating material can be measured using an ellipsometer.

In addition to its electrical properties, the coating material may have properties that are beneficial for use with biomolecules. For example, coating materials containing fluoro (or perfluoro) alkyl groups may provide benefits in reducing the amount of surface fouling relative to unsubstituted alkyl groups. Surface fouling, as used herein, refers to the amount of any substance deposited on the surface of a microfluidic device, which may include permanent or semi-permanent deposition of biological substances (e.g., proteins and their degradation products, nucleic acids and their degradation products). Such fouling can increase the amount of adhesion of biological micro-objects to the surface.

In addition to the composition of the conditioned surface, other factors (e.g., the physical thickness of the hydrophobic material) may affect the DEP force. Various factors may alter the physical thickness of the conditioned surface, such as the manner in which the conditioned surface is formed on the substrate (e.g., vapor deposition, liquid deposition, spin coating, flooding, and electrostatic coating). The physical thickness and uniformity of the conditioned surface can be measured using an ellipsometer.

In addition to its electrical properties, the conditioned surface may also have properties that are beneficial for use with biomolecules. For example, a conditioned surface containing fluorinated (or perfluorinated) carbon chains may provide benefits in reducing the amount of surface fouling relative to alkyl terminated chains. Surface fouling, as used herein, refers to the amount of any substance deposited on the surface of a microfluidic device, which may include permanent or semi-permanent deposition of biological materials (e.g., proteins and their degradation products, nucleic acids and respective degradation products, and the like).

The following table includes a number of properties of the conditioned surface that can be used in DEP. As shown, for entries 1 to 7 (all of which were covalently attached to the conditioned surface as described herein), the thickness as measured by the ellipsometric technique was consistently thinner than that of entry 8 (a CYTOP surface formed by non-covalent spin coating) (N/a represents unavailable data throughout the table). Fouling was found to be more dependent on the chemistry of the surface than the manner of formation, since fluorinated surfaces are generally less prone to fouling than alkyl (hydrocarbon) conditioned surfaces.

Table 1. properties of various conditioned surfaces prepared by covalently modifying a surface compared to a non-covalently formed surface CYTOP.

Figure BDA0002104386230000481

Figure BDA0002104386230000491

1. Spin coating, non-covalent.

A surface linking group. The covalent linking moieties forming the coating material are attached to the surface via a linking group. The linking group can be a siloxy linking group formed by reaction of a siloxane-containing reagent with an oxide of the substrate surface, which can include silicon oxide (e.g., a substrate for DEP configuration) or aluminum oxide or hafnium oxide (e.g., a substrate for EW configuration). In some other embodiments, the linking group can be a phosphate ester formed by reacting a phosphonic acid-containing reagent with an oxide of the substrate surface.

A multi-part conditioned surface. As described below, the covalently linked coating material may be formed by the reaction of molecules (e.g., alkyl siloxane reagents or fluoro-substituted alkyl siloxane reagents, which may include perfluoroalkyl siloxane reagents) that already contain moieties configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) in a microfluidic device. Alternatively, the covalently linked coating material may be formed by coupling a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) to a surface modifying ligand, which itself is covalently linked to the surface.

A method of making a covalently linked coating material. In some embodiments, a coating material covalently attached to a surface of a microfluidic device (e.g., at least one surface comprising an isolation dock and/or a flow region) has the structure of formula 1.

Figure BDA0002104386230000492

The coating material may be covalently attached to the oxide on the surface of the DEP configured substrate. The DEP configured substrate may comprise silicon or aluminum oxide or hafnium oxide, and the oxide may be present as part of the initial chemical structure of the substrate, or may be introduced as discussed below.

The coating material may be attached to the oxide via a linking group ("LG"), which may be a siloxy or phosphonate group formed from the reaction of a siloxane or phosphonate group with the oxide. The portion configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) in a microfluidic device may be any portion described herein. The linking group LG may be directly or indirectly linked to a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g. a cell, such as an immune cell (e.g. a B cell) or a hybridoma cell) in a microfluidic device. When the linking group LG is directly linked to the moiety, there is no optional linking moiety ("L") and n is 0. When the linking group LG is indirectly linked to the moiety, there is a linking moiety L and n is 1. The linking moiety L may have a linear portion, wherein the backbone of the linear portion may include 1 to 200 non-hydrogen atoms selected from any combination of silicon, carbon, nitrogen, oxygen, sulfur, and phosphorus atoms, subject to chemical bonding limitations known in the art. In some non-limiting examples, it may be interrupted by any combination of one or more moieties that may be selected from ether, amino, carbonyl, amido, or phosphonate groups. In addition, the linking moiety L may have one or more arylene, heteroarylene, or heterocyclyl groups interrupting the backbone of the linking group. In some embodiments, the backbone of the linking moiety L may comprise 10 to 20 atoms. In other embodiments, the backbone of the linking moiety L may comprise from about 5 atoms to about 200 atoms; about 10 atoms to about 80 atoms; about 10 atoms to about 50 atoms; or from about 10 atoms to about 40 atoms. In some embodiments, the backbone atoms are all carbon atoms. In other embodiments, the backbone atoms are not all carbon, and may include any possible combination of silicon, carbon, nitrogen, oxygen, sulfur, and phosphorus atoms, subject to the limitations of chemical bonding known in the art.

When a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g. cells, such as immune cells (e.g. B cells) or hybridoma cells) in a microfluidic device is added to the substrate surface in a one-step process, the molecules of formula 2 may be used to introduce a coating material:

part- (L) n-LG.

Formula 2

In some embodiments, the portion configured to provide the layer of organic and/or hydrophilic molecules suitable for maintaining and/or expanding biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) in a microfluidic device may be added to the surface of the substrate in a multi-step process. When the moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding biological micro-objects (e.g. cells such as immune cells (e.g. B-cells) or hybridoma cells) is coupled to the surface in a stepwise manner, the linking moiety L may further comprise a coupling group CG, as shown in formula 3.

Figure BDA0002104386230000511

In some embodiments, the coupling group CG is represented by a reactive moiety RxAnd a reactive partner Rpx(i.e., configured to react with the reactive moiety R)xPart of the reaction). For example, a typical coupling group CG may include a carboxamide group that is the result of the reaction of an amino group with a carboxylic acid derivative (e.g., an activated ester, an acid chloride, etc.). Other CGs may include triazolylene, carboxamido, thioamido, oxime, mercapto, disulfide, ether or alkenyl groups, or any other suitable group that may be formed upon reaction of a reactive moiety with its corresponding reactive partner moiety. The coupling group CG may be located at the second end of the linking group L (i.e., adjacent to a region configured to provide a biological microorganism (e.g., a cell, such as an immune cell (e.g., B)) suitable for maintaining and/or expanding in a microfluidic deviceCells) or hybridoma cells) are used in the presence of a hydrophilic molecular layer). In some other embodiments, the coupling group CG may interrupt the backbone of the linking group L. In some embodiments, the coupling group CG is a triazolylene group, which may be obtained by reaction between an alkyne group and an azide group, any of which may be a reactive moiety RxAnd a reactive partner RpxAs known in the art for Click coupling reactions. For example, the dibenzocyclooctenyl-fused triazolylene group may be conjugated to a dibenzocyclooctynyl reactive partner Rpx(ii) an azido-reactive moiety R with a surface-modifying moleculexThe reaction of (a), which is described in more detail in the following paragraphs. A variety of dibenzocyclooctynyl-modified molecules are known in the art, or can be synthesized to incorporate moieties that are configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological microorganism (e.g., a cell, such as an immune cell (e.g., B cell) or a hybridoma cell).

When the coating material is formed in a multi-step process, moieties configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or amplifying biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) in a microfluidic device may be introduced by reacting a reagent containing the moiety (formula 5) with a substrate having a surface modifying ligand covalently attached thereto (formula 6).

The modified surface of formula 4 has a surface modifying ligand attached thereto having the formula-LG- (L') j-RxWhich is connected to the oxide of the substrate and is formed similarly as described above for the conditioned surface of equation 1. The surface of the substrate may be a substrate surface of the DEP configuration as described above, and may comprise the substrate itself or an oxide incorporated therein. The linking group LG is as described above. The linking moiety L "may be present (j ═ 1) or absent (j ═ 0). The linking moiety L' may have a linear portion wherein the backbone of the linear portion may comprise from 1 to 100 non-hydrogen atoms selected from silicon, carbon, nitrogen,any combination of oxygen, sulfur, and phosphorus atoms, subject to the limitations of chemical bonding known in the art. In some non-limiting examples, it may be interrupted by any combination of ether, amino, carbonyl, amido, or phosphonate groups. In addition, the linking moiety L "may have one or more arylene, heteroarylene, or heterocyclic groups that interrupt the backbone of the linking moiety. In some embodiments, the backbone of the linking moiety L "may comprise 10 to 20 carbon atoms. In other embodiments, the backbone of the linking moiety L "can comprise from about 5 atoms to about 100 atoms; from about 10 atoms to about 80 atoms, from about 10 atoms to about 50 atoms, or from about 10 atoms to about 40 atoms. In some embodiments, all of the backbone atoms are carbon atoms. In other embodiments, the backbone atoms are not all carbon, and may include any possible combination of silicon, carbon, nitrogen, oxygen, sulfur, or phosphorus atoms, subject to the limitations of chemical bonding known in the art.

Reactive moiety RxIs present at the end of the surface modifying ligand remote from the covalent attachment of the surface modifying ligand to the surface. Reactive moiety RxIs any suitable reactive moiety that can be used in a coupling reaction to introduce a moiety that provides an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell) in a microfluidic device. In some embodiments, the reactive moiety RxCan be an azido, amino, bromo, thiol, activated ester, succinimidyl, or alkynyl moiety.

A reagent containing a moiety. The reagent containing moiety (formula 5) is configured to supply a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or amplifying a biological micro-object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell) in a microfluidic device.

Moiety- (L')m-Rpx

Formula 5

Configured to provide, in a reagent containing a moiety, an organism and/or a cell suitable for maintaining and/or expanding a biological microorganism (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell)Part of the hydrophilic molecular layer is formed by a reactive pair RpxWith reactive moieties RxThe reaction of (a) is linked to a surface modifying ligand. Reactive partner RpxIs any suitable reactive group configured to react with a corresponding reactive moiety RxAnd (4) reacting. In a non-limiting example, a suitable reactive partner RpxCan be an alkyne, a reactive moiety RxMay be an azide. Reactive partner RpxMay alternatively be an azide moiety, the corresponding reactive moiety RxMay be an alkyne. In other embodiments, the reactive partner RpxCan be an active ester functional group, a reactive moiety RxMay be an amino group. In other embodiments, the reactive partner RpxMay be an aldehyde, a reactive moiety RxMay be an amino group. Other reactive moiety-reactive partner combination are possible, and these examples are in no way limiting.

The portion of the reagent containing moiety of formula 5 configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g., a cell, such as an immune cell (e.g., a B cell) or a hybridoma cell) can include any of the moieties described herein, including alkyl or fluoroalkyl (including perfluoroalkyl) moieties; mono-or polysaccharides (which may include, but are not limited to, dextran); alcohols (including but not limited to propargyl alcohol); polyols, including but not limited to polyvinyl alcohol; alkylene ethers including, but not limited to, polyethylene glycol; polyelectrolytes (including but not limited to polyacrylic acid or polyvinylphosphonic acid); amino (including derivatives thereof such as, but not limited to, alkylated amines, hydroxyalkylated amino, guanidinium salts, and heterocyclic groups containing a nitrogen ring atom that is not aromatic, such as, but not limited to, morpholinyl or piperazinyl); carboxylic acids, including but not limited to propiolic acid (which can provide a carboxylate anionic surface); phosphonic acids, including but not limited to ethynylphosphonic acid (which can provide a phosphonic acid anionic surface); a sulfonate anion; a carboxybetaine; a sulfobetaine; sulfamic acid; or an amino acid.

The configuration of the reagent containing moieties of formula 5 provides a reagent suitable for maintaining and/or amplifying a biological microorganism (e.g., a cell)E.g. an immune cell (e.g. a B cell) or a hybridoma cell) may be linked directly (i.e. L', where m ═ 0) or indirectly to the reactive partner moiety Rpx. When the reactive partner R ispxIndirectly linked to a moiety configured to provide an organic and/or hydrophilic molecular layer suitable for maintaining and/or expanding a biological micro-object (e.g. a cell, such as an immune cell (e.g. a B cell) or a hybridoma cell), a reactive partner RpxMay be connected to the connecting portion L' (m ═ 1). Reactive partner RpxA moiety that can be attached to a first end of the linking moiety L 'and that is configured to reduce surface fouling and/or prevent or reduce cell adhesion can be attached to a second end of the linking moiety L'. The linking moiety L' may have a linear portion, wherein the backbone of the linear portion comprises 1 to 100 non-hydrogen atoms selected from any combination of silicon, carbon, nitrogen, oxygen, sulfur and phosphorus atoms, subject to chemical bonding limitations known in the art. In some non-limiting examples, it may be interrupted by any combination of ether, amino, carbonyl, amido, or phosphonate groups. In addition, linker L 'may have one or more arylene, heteroarylene, or heterocyclic groups that interrupt the backbone of linker L'. In some embodiments, the backbone of the linking moiety L' may comprise 10 to 20 carbon atoms. In other embodiments, the backbone of the linking moiety L' may comprise from about 5 atoms to about 100 atoms; about 10 atoms to about 80 atoms; about 10 atoms to about 50 atoms; or from about 10 atoms to about 40 atoms. In some embodiments, all of the backbone atoms are carbon atoms. In other embodiments, the backbone atoms are not all carbon, and may include any possible combination of silicon, carbon, nitrogen, oxygen, sulfur, or phosphorus atoms, subject to the limitations of chemical bonding known in the art.

When the reagent containing moieties (formula 5) reacts with the surface having surface modifying ligands (formula 3), a substrate having a conditioned surface of formula 2 is formed. The linking moiety L 'and linking moiety L' are then formally part of the linking moiety L, and the reactive partner RpxWith a reactive moiety RxTo produce the coupling group of formula 2The cluster CG.

A surface modifier. The surface modifier is of the structure LG- (L')j-RxA compound of (formula 4). The linking group LG is covalently linked to the oxide on the surface of the substrate. The substrate may be a DEP configured substrate and may comprise silicon or aluminum oxide or hafnium oxide, and the oxide may be present as part of the native chemical structure of the substrate or may be incorporated as discussed herein. The linking group LG can be any linking group described herein, such as a siloxy or phosphonate group, formed from the reaction of a siloxane or phosphonate group with an oxide on the surface of a substrate. Reactive moiety RxAs described above. Reactive moiety RxThe linking group LG may be directly linked to (L ", j ═ 0) or indirectly linked to (j ═ 1) through a linking moiety L". The linking group LG may be linked to a first end of the linking moiety L' and the reactive moiety RxCan be attached to the second end of the linking moiety L', the reactive moiety R once the surface modifying reagent has been attached to the surface as shown in formula 6xWill be located distally of the substrate surface.

Figure BDA0002104386230000551

The linking moiety L "may have a linear portion, wherein the backbone of the linear portion comprises 1 to 100 non-hydrogen atoms selected from any combination of silicon, carbon, nitrogen, oxygen, sulfur and phosphorus atoms. In some non-limiting examples, it may be interrupted by any combination of ether, amino, carbonyl, amido, or phosphonate groups. In addition, the linking moiety L "may have one or more arylene, heteroarylene, or heterocyclic groups that interrupt the backbone of the linking moiety L". In some embodiments, the backbone of the linking moiety L "may comprise 10 to 20 carbon atoms. In other embodiments, the backbone of the linking moiety L "can comprise from about 5 atoms to about 100 atoms; from about 10 atoms to about 80 atoms, from about 10 atoms to about 50 atoms, or from about 10 atoms to about 40 atoms. In some embodiments, all of the backbone atoms are carbon atoms. In other embodiments, the backbone atoms are not all carbon, and may include any possible combination of silicon, carbon, nitrogen, oxygen, sulfur, or phosphorus atoms, subject to the limitations of chemical bonding known in the art.

In some embodiments, the coating material (or surface-modified ligand) is deposited on the inner surface of the microfluidic device using chemical vapor deposition. By chemical vapor deposition, the coating material may achieve a close-packed monolayer, wherein molecules comprising the coating material are covalently bonded to molecules of the inner surface of the microfluidic device. To achieve the desired packing density, molecules comprising, for example, alkyl-terminated siloxanes can be vapor deposited at a temperature of at least 110 ℃ (e.g., at least 120 ℃, 130 ℃, 140 ℃, 150 ℃, 160 ℃, etc.) for at least 15 hours (e.g., at least 20, 25, 30, 35, 40, 45, or more hours). Such vapor deposition is typically carried out under vacuum and in a water source (e.g., hydrated sulfate salts (e.g., MgSO)4·7H2O)) in the presence of oxygen. Generally, increasing the temperature and duration of the vapor deposition results in improved properties of the hydrophobic coating material.

The vapor deposition process can optionally be modified, for example, by pre-cleaning the lid 110, the microfluidic conduit material 116, and/or the substrate (e.g., the inner surface 208 of the electrode activation substrate 206 of a DEP configured substrate, or the dielectric layer of the support structure 104 of an EW configured substrate). For example, the pre-clean may include a solvent bath, such as an acetone bath, an ethanol bath, or a combination thereof. The solvent bath may include sonication. Alternatively or additionally, such pre-cleaning may include treating the cover 110, the microfluidic circuit material 116, and/or the substrate in an oxygen plasma cleaner, which may remove various impurities while introducing an oxidized surface (e.g., an oxide on the surface, which may be covalently modified as described herein). The oxygen plasma cleaner may be operated, for example, at 100W under vacuum for 60 seconds. Alternatively, a liquid phase treatment, which includes an oxidizing agent (e.g., hydrogen peroxide) to oxidize the surface, may be used in place of the oxygen plasma cleaner. For example, a mixture of hydrochloric acid and hydrogen peroxide or a mixture of sulfuric acid and hydrogen peroxide (e.g., a piranha solution, which may have a ratio range of sulfuric acid to hydrogen peroxide of about 3:1 to about 7: 1) is substituted for the oxygen plasma cleaner.

In some embodiments, vapor deposition is used to coat the inner surfaces of the microfluidic device 200 after the microfluidic device 200 has been assembled to form the housing 102 defining the microfluidic circuit 120. Depositing a coating material comprising a tightly packed monolayer on a fully assembled microfluidic circuit 120 may be beneficial in providing a variety of functional properties. Without wishing to be bound by theory, depositing such a coating material on the fully assembled microfluidic circuit 120 may be beneficial to prevent delamination caused by weakened bonding between the microfluidic circuit material 116 and the electrode activation substrate 206 dielectric layer and/or the cap 110.

Fig. 2H depicts a cross-sectional view of a microfluidic device 290, the microfluidic device 290 including an example class of coating materials. As shown, the coating material 298 (shown schematically) may comprise a close-packed molecular monolayer covalently bonded to both the inner surface 294 of the substrate 286 and the inner surface 292 of the cover 288 of the microfluidic device 290. The coating material 298 may be disposed on all of the interior surfaces 294, 292 of the housing 284 adjacent to and inwardly facing the microfluidic device 290, in some embodiments and as discussed above, including surfaces of microfluidic tubing material (not shown) for defining tubing elements and/or structures within the microfluidic device 290. In alternative embodiments, the coating material 298 may be disposed only on one or some of the interior surfaces of the microfluidic device 290.

In the embodiment shown in fig. 2H, the coating material 298 comprises a monolayer of alkyl-terminated siloxane molecules, each molecule covalently bonded to an interior surface 292, 294 of the microfluidic device 290 via a siloxy linker 296. However, any of the above-described coating materials 298 (e.g., alkyl-terminated phosphonate molecules) can be used. More specifically, the alkyl group can comprise a straight chain of at least 10 carbon atoms (e.g., 10, 12, 14, 16, 18, 20, 22, or more carbon atoms) and, optionally, can be a substituted alkyl group. As described above, the coating material 298 comprising a tightly packed molecular monolayer may have beneficial functional properties for the DEP configured microfluidic device 290, such as minimal charge trapping, reduced physical/electrical thickness, and a substantially uniform surface.

In another particular embodiment, the coating material 298 may include a fluoroalkyl group (e.g., fluoroalkyl or perfluoroalkyl) at its end facing the housing (i.e., the portion of the monolayer of coating material 298 that is not bonded to the inner surfaces 292, 294 and is proximate to the housing 284). As described above, the coating material 298 may include a single layer of fluoroalkyl terminated siloxane or fluoroalkyl terminated phosphonate, where fluoroalkyl groups are present at the end of the coating material 298 facing the housing. Such a coating material 298 provides improved functional benefits of maintenance and/or expansion of biological micro-objects (e.g., cells, such as immune cells (e.g., B cells) or hybridoma cells) by separating or "shielding" the biological micro-objects from non-biological molecules (e.g., silicon and/or silicon oxide of the substrate).

In other particular embodiments, the coating material 298 used to coat the interior surfaces 292, 294 of the microfluidic device 290 may include anionic, cationic, or zwitterionic moieties, or any combination thereof. Without wishing to be bound by theory, by providing cationic, anionic, and/or zwitterionic moieties on the inner surface of the housing 284 of the microfluidic circuit 120, the coating material 298 may form strong hydrogen bonds with water molecules, such that the resulting hydrated water acts as a layer (or "shield") separating the core (nucleoei) from interactions with non-biological molecules (e.g., silicon and/or silicon oxide of the substrate). Additionally, in embodiments where the coating material 298 is used in conjunction with a blocking agent, the anion, cation, and/or zwitterion of the coating material 298 may form an ionic bond with a charged portion of a non-covalent coating agent (e.g., a protein in solution) present in the medium 180 (e.g., a coating solution) in the housing 284.

In another specific embodiment, the coating material may comprise or be chemically modified to provide a hydrophilic coating agent at its end facing the housing. In some embodiments, the coating agent may be an alkylene ether containing polymer, such as PEG. In some embodiments, the coating agent may be a polysaccharide, such as dextran. As with the charged moieties discussed above (e.g., anionic, cationic, and zwitterionic moieties), the hydrophilic coating agent can form strong hydrogen bonds with water molecules, such that the resulting water of hydration acts as a layer (or "shield") separating the core from interactions with non-biological molecules (e.g., silicon and/or silicon oxide of the substrate).

Methods for detecting expression of an antibody. The methods disclosed herein include methods of detecting or identifying biological cells that express antibodies that specifically bind to an antigen of interest. The antigen of interest may be a protein, a carbohydrate group or chain, a biological or chemical agent other than a protein or carbohydrate, or any combination thereof. The antigen of interest can be, for example, an antigen associated with a pathogen (e.g., a virus, a bacterial pathogen, a fungal pathogen, a protozoan pathogen, etc.). Alternatively, the antigen of interest may be associated with a cancer (e.g., lung cancer, breast cancer, melanoma, etc.). In another alternative, the antigen may be associated with an autoimmune disease (e.g., multiple sclerosis or type I diabetes). As used herein, the term "associated with a pathogen" when used with an antigen of interest means that the antigen of interest is produced directly by the pathogen or by an interaction between the pathogen and the host.

Methods of detecting biological cells expressing antibodies that specifically bind to an antigen of interest can be performed in the microfluidic devices described herein. In particular, the microfluidic device may comprise a housing having a flow region that may comprise one or more microfluidic channels and an isolation dock (or multiple isolation docks). The isolation dock may include a separation region and a connection region that provides a fluidic connection between the separation region and the flow region/microfluidic channel. The volume of the isolation dock can be about 0.5nL to about 5.0nL or any range therein (e.g., about 0.5nL to about 1.0nL, about 0.5nL to about 1.5nL, about 0.5nL to about 2.0nL, about 1.0nL to about 1.5nL, about 1.0nL to about 2.0nL, about 1.0nL to about 2.5nL, about 1.5nL to about 2.0nL, about 1.5nL to about 2.5nL, about 1.5nL to about 3.0nL, about 2.0nL to about 2.5nL, about 2.0nL to about 3.0nL, about 2.0nL to about 3.5nL, about 2.5nL to about 3.5nL, about 3.5nL to about 3.0 to about 3.5nL, about 3.5nL to about 3.5nL, about 3.0 to about 3.5nL, about 3.5nL to about 3.5nL, about 3.0 to about 3.5nL). The connection region may have a width W as generally described hereincon(e.g., from about 20 microns to about 100 microns, or from about 30 microns to about 60 microns). Width W of the separation regionisoMay be greater than the width W of the connection regioncon. In certain embodiments, the width W of the separation regionisoFrom about 50 microns to about 250 microns.

The flow region, the isolation dock, and/or the isolation area of the isolation dock may comprise at least one surface coated with a coating material that promotes viability of the biological cells and/or reduces interaction with the biological cells. Thus, for example, the coating material may promote the viability of hybridoma cells, and/or promote the viability of B-cell lymphocytes (e.g., memory B cells or plasma cells), and/or the ability to move any such cells into the microfluidic device. As used herein, "promoting viability" means that the viability of the antibody-expressing biological cells is better on the coated surface compared to an uncoated equivalent surface. In certain embodiments, the flow region, isolation dock, and/or isolation region has a plurality of surfaces, each surface coated with a coating material that promotes viability of and/or reduces interaction with antibody-expressing cells. The coating material can be any suitable coating material known in the art and/or described herein. The coating material may for example comprise hydrophilic molecules. The hydrophilic molecule may be selected from the group consisting of a polymer comprising polyethylene glycol (PEG), a polymer comprising a carbohydrate group, a polymer comprising an amino acid (e.g., a protein, such as BSA), and combinations thereof.

The flow region, the isolation dock, and/or the isolation region of the isolation dock may comprise at least one conditioned surface that promotes viability of the antibody-expressing biological cells and/or reduces interaction with the antibody-expressing biological cells. Thus, for example, the conditioned surface may promote the viability of hybridoma cells, and/or promote the viability of B-cell lymphocytes (e.g., memory B cells or plasma cells), and/or promote the ability to move any cells into the microfluidic device. As used herein, "promoting viability" means that the viability of the antibody-expressing biological cells is better on the conditioned surface compared to an unregulated equivalent surface. In certain embodiments, the flow region, the spacer dock, and/or the separation region has a plurality of conditioned surfaces, each conditioned surface capable of promoting viability of the antibody-expressing cell and/or reducing interaction with the antibody-expressing cell. The conditioned surface may comprise covalently attached molecules. The covalently linked molecule can be any suitable molecule known in the art and/or disclosed herein, including, for example, covalently linked hydrophilic molecules. The hydrophilic molecule may be selected from the group consisting of a polymer comprising polyethylene glycol (PEG), a polymer comprising a carbohydrate group, a polymer comprising an amino acid, and combinations thereof. As described herein, the hydrophilic molecules may form a covalently linked layer of hydrophilic molecules. Alternatively, the covalently linked molecules may comprise perfluoroalkanes (e.g., covalently linked perfluoroalkane layers).

A method of detecting a biological cell expressing an antibody that specifically binds to an antigen of interest may comprise the steps of: introducing a sample containing antibody-expressing biological cells into a microfluidic device; loading antibody-expressing biological cells into a separation region of a separation dock in a microfluidic device; introducing a target antigen into the microfluidic device such that the target antigen is in the vicinity of the antibody-expressing biological cells; and monitoring the binding of the antigen of interest to the antibody expressed by the biological cell.

The antibody-expressing biological cell can be, for example, a hybridoma cell. Alternatively, the antibody-expressing biological cell may be a B cell lymphocyte. The B cell lymphocyte may be, for example, CD27+B cells or CD138+B cells. In some embodiments, the B cell is a memory B cell. In other embodiments, the B cell is a plasma cell.

Introducing the antibody-expressing biological cells into the microfluidic device can involve obtaining a sample containing the antibody-expressing cells. For embodiments in which the antibody-expressing biological cells are B cell lymphocytes, the sample containing B cell lymphocytes can be obtained from a mammal, such as a human, a rodent (e.g., mouse, rat, guinea pig, gerbil, hamster), a rabbit, a ferret, a livestock (e.g., goat, sheep, pig, horse, cow), a llama, a camel, a monkey, or from an avian species, such as a chicken and turkey. In some embodiments, the mammal has been immunized against an antigen of interest. In some embodiments, the animal has been exposed to or infected with a pathogen associated with the antigen of interest. In some embodiments, the animal has a cancer associated with an antigen of interest. In other embodiments, the animal has an autoimmune disease associated with an antigen of interest. The sample containing B cell lymphocytes may be a peripheral blood sample (e.g., PBMCs), spleen biopsy, bone marrow biopsy, lymph node biopsy, tumor biopsy, or any combination thereof.

A sample containing B cell lymphocytes can be processed (e.g., sorted, negative and/or positive) to enrich for desired B cell lymphocytes. In some embodiments, the desired B cell lymphocyte is a memory B cell. In other embodiments, the desired B cell lymphocyte is a plasma cell. In some embodiments, the desired B cell lymphocytes express IgG-type antibodies. Thus, for example, the sample may be depleted of cell types other than B-cell lymphocytes. Methods for depleting non-B cell types from a sample are well known in the art and include, for example, using DYNABEADSTMUnmodified B cell reagent (DYNABEADS)TMUntouched Human B Cells reagent, Thermo Fisher), B Cell Isolation Kit (Miltenyi), easy Sep B Cell Enrichment Kit (easy Sep), Rosetesep Human B Cell Enrichment Cocktain (Rosetesep Human B Cell Enrichment Cocktain, Stem Cell Technologies), and the like. Alternatively, or in addition, samples containing B cell lymphocytes can be sorted by fluorescence-associated cell sorting (FACS) to remove unwanted cell types and enrich for desired cell types. FACS sorting may be negative and/or positive. For example, FACS sorting can deplete B cell lymphocyte samples expressing IgM antibodies, IgA antibodies, IgD antibodies, IgG antibodies, or any combination thereof. Alternatively, or in addition, FACS sorting may enrich a sample for B cell lymphocytes expressing CD27 (or some other memory B cell marker) or B cell lymphocytes expressing CD138 (or some other plasma cell marker). The sample containing B cell lymphocytes can be provided in an enriched state (i.e., pretreated) such that it is not necessary to perform a site of enrichment for the desired B cell lymphocytesAs part of the method. Alternatively, treatment of a sample containing B cell lymphocytes to enrich for desired B cell lymphocytes can be performed as part of the method of the invention.

The sample containing B cell lymphocytes can be treated to reduce cell adhesion to the microfluidic device in the sample. For example, DNase may be used (e.g.Nucleic (Millipore)) treatment of the sample. Preferably, the dnase contains minimal protease activity.

Introduction of the antibody-expressing biological cells into the microfluidic device can be performed by flowing a sample containing the biological cells into an inlet of the microfluidic device and through a portion of a flow region of the microfluidic device. The flow of sample through the microfluidic device can then be stopped to allow loading of antibody-expressing biological cells (e.g., B-cell lymphocytes) into the isolated region of the sequestration dock. Loading of antibody-expressing cells into the separation region can be performed by any technique known in the art or disclosed herein, for example, using gravitational forces and/or DEP forces. In certain embodiments, a single antibody-expressing cell (e.g., a B cell lymphocyte) is loaded into the isolation region. In certain embodiments, a single antibody-expressing cell (e.g., a B-cell lymphocyte) is loaded into a separate region of each of a plurality of isolated docks in a microfluidic device.

Methods of detecting biological cells that express an antibody that specifically binds to an antigen of interest can include the step of contacting a B cell lymphocyte with a stimulating agent that stimulates B cell activation. The stimulating agent may be a CD40 agonist, such as CD40L, a derivative thereof, or an anti-CD 40 antibody. The stimulant may comprise CD40L+Feeder cells, consisting essentially of, or consisting of. CD40L+The feeder cells may be T cells (e.g., Jurkat D1.1 cells) or derivatives thereof. Alternatively, the feeder cells may be cell lines transfected/transformed with the CD40L expression construct (e.g., NIH-3T3 cells). The stimulating agent may further comprise a B Cell Receptor (BCR) superantigen, such as protein a, protein G, or any other BCR superantigen. The BCR superantigen may be linked toMicro-objects such as beads, lipid vesicles, lipid nanorafts, etc. Thus, the superantigen-coated microobjects can interact with CD40L+Feeder cells were mixed. The mixture may have a ratio of about 1:1 feeder cells to micro-objects, or a ratio of about 1:5 feeder cells to micro-objects, or any ratio therebetween. Alternatively, the mixture may have a ratio of about 1:2 feeder cells to micro-objects, or a ratio of about 2:10 feeder cells to micro-objects, or any ratio therebetween. The stimulating agent may further comprise a toll-like receptor (TLR) agonist (e.g., a TLR9 agonist), which may be combined with a CD40 agonist and optionally a BCR superantigen. The TLR agonist may be, for example, a CpG oligonucleotide (e.g., CpG 2006). The CpG oligonucleotide may be used at a concentration of about 1 microgram/mL to about 20 microgram/mL (e.g., about 1.5 to about 15 microgram/mL, about 2.0 to about 10 microgram/mL, or about 2.5 to about 5.0 microgram/mL). The B cell lymphocytes can be contacted (e.g., substantially continuously, or periodically/intermittently) with the stimulating agent for 1 to 10 days (e.g., 2 to 8 days, 3 to 7 days, or 4 to 6 days). B cell lymphocytes can be contacted with a stimulating agent in a sequestration dock into which the B cell lymphocytes are loaded. This contact may occur after the B cell lymphocytes are loaded into the isolation dock.

The method of detecting a biological cell that expresses an antibody that specifically binds to an antigen of interest can further comprise the step of providing the antibody-expressing biological cell (e.g., a B cell lymphocyte) with a culture/activation medium comprising one or more growth inducing agents that promote B cell activation and/or expansion. The one or more growth-inducing agents may include at least one agent selected from the group consisting of CpG oligonucleotides, IL-2, IL-4, IL-6, IL-10, IL-21, BAFF, and April. IL-2 can be provided at a concentration of about 2ng/mL to about 5ug/mL, or about 50ng/mL to about 2ug/mL, or about 100ng/mL to about 1.5ug/mL, or about 500ng/mL to about 1ug/mL, or about 1 ug/mL. IL-4 may be provided at a concentration of about 2ng/mL to about 20ng/mL, or about 5ng/mL to about 10ng/mL, or about 5 ng/mL. IL-6, IL-10 and/or IL-21 may be provided at a concentration of about 2ng/mL to about 50ng/mL, or about 5ng/mL to about 20ng/mL, or about 10 ng/mL. BAFF and/or April may be provided at a concentration of about 10ng/mL to about 100ng/mL, or about 10ng/mL to about 50ng/mL, or about 10ng/mL to about 20ng/mL, or about 10 ng/mL. CpG oligonucleotides may be used at a concentration of about 1 microgram/mL to about 20 microgram/mL, about 1.5 to about 15 microgram/mL, about 2.0 to about 10 microgram/mL, or about 2.0 microgram/mL. In certain embodiments, the antibody-expressing biological cells are provided with culture medium for 1 to 10 days (e.g., 2 to 8 days, 3 to 7 days, or 4 to 6 days). The culture medium may comprise a stimulating agent (e.g., a CD40 agonist and/or a BCR superantigen). Thus, for example, where the antibody-producing cell is a B cell lymphocyte, the providing of the culture medium to the B cell lymphocyte can be performed while contacting the B cell lymphocyte with the activating agent. In certain embodiments, the steps of contacting the B cell lymphocytes with the stimulating agent and providing the culture medium to the B cell lymphocytes are performed within an overlapping time period (e.g., within a substantially coextensive time period).

In certain embodiments, introducing the antigen of interest into the microfluidic device such that the antigen of interest is located in the vicinity of the antibody-expressing biological cell comprises locating the antigen of interest within 1 millimeter (mm) of the biological cell (e.g., within 750 microns, within 600 microns, within 500 microns, within 400 microns, within 300 microns, within 200 microns, within 100 microns, or within 50 microns of the biological cell). In certain embodiments, the method may comprise introducing a micro-object or a plurality of micro-objects into a flow region/microfluidic channel connected to an isolation dock. The micro-object may comprise an antibody-specific binding agent, such as an anti-IgG antibody or other IgG binding agent. See, for example, fig. 6C. In such embodiments, monitoring binding of the antigen of interest to the antibody expressed by the biological cell comprises detecting indirect binding of the labeled antigen of interest to the micro-object via the antibody expressed by the antibody expressing biological cell. The labeled target antigen may be soluble and may include a detectable label, such as a fluorescent label. The micro-objects may be any suitable micro-objects (e.g., cells, liposomes, lipid nanorafts, or beads) known in the art and/or described herein. The step of providing the antigen of interest may comprise placing such micro-objects near or within the connection region of the isolation dock, wherein the antibody-expressing biological cell is located. Alternatively, the step of providing the antigen of interest may comprise loading such micro-objects into separate regions of the sequestration dock, in which the antibody-expressing biological cells are located. The micro-objects and the target antigen may be provided simultaneously as a mixture, or sequentially (if the micro-objects are first placed in a separate dock).

Alternatively, in certain embodiments, the method may comprise introducing a micro-object or a plurality of micro-objects into a flow region/microfluidic channel connected to an isolation dock, wherein the antigen of interest binds to the micro-object. In such embodiments, a soluble labeled antibody-specific binding agent, such as an anti-IgG antibody or other IgG binding agent, can also be provided, and monitoring binding of the antigen of interest to the antibody expressed by the biological cell comprises detecting indirect binding of the labeled antibody-specific binding agent to the micro-object via the antibody expressed by the antibody-expressing biological cell. The labeled antibody-specific binding agent may comprise a detectable label, such as a fluorescent label. The micro-objects may be any suitable micro-objects (e.g., cells, liposomes, lipid nanorafts, or beads) known in the art and/or described herein. The step of providing the antigen of interest may comprise placing such micro-objects near or within the connection region of the isolation dock, in which the antibody-expressing biological cell is located. The step of providing the antigen of interest may further comprise loading such micro-objects into a separate region of the sequestration dock, wherein the antibody-expressing biological cells are located. The micro-objects and antibody-specific binding agent may be provided simultaneously as a mixture, or sequentially (if the micro-objects are first placed in a separate dock). Methods of screening for expression of a molecule of interest (e.g., an antibody) have been described, for example, in U.S. patent publication No. US2015/0151298, which is incorporated by reference herein in its entirety.

In some embodiments, the method further comprises providing a second antibody-specific binding agent prior to or concurrently with the first antibody-specific binding agent. See, e.g., fig. 6C. The second antibody binding agent may be an anti-IgG antibody or other type of antibody binding agent, and may be labeled (e.g., with a fluorescent label). In certain embodiments, the labeled second antibody-specific binding agent is provided in a mixture with the antigen of interest and the first antibody-specific binding agent. In other embodiments, the labeled second antibody-specific binding agent is provided after the antigen of interest and/or the first antibody-specific binding agent is provided.

In certain embodiments, providing the target antigen can include flowing a solution comprising a soluble target antigen through a flow region of the microfluidic device and diffusing the soluble antigen into a sequestration dock in which the antibody-expressing biological cell is located. Such soluble antigen may be covalently bound to a detectable label (e.g., a fluorescent label). A general method of screening for expression of molecules of interest (including antibodies) in this manner has been described in, for example, international application PCT/US2017/027795 filed on 2017, 4/14, which is incorporated herein by reference in its entirety.

In certain embodiments, the method can further comprise detecting binding of an antigen of interest to an antibody expressed by the biological cell (e.g., a B cell lymphocyte), and identifying an antibody-expressing biological cell (e.g., a B cell lymphocyte) that expresses an antibody that specifically binds the antigen of interest.

Obtaining antibody sequences from the recognized B cell lymphocytes. Also disclosed herein are methods of providing sequencing libraries and/or obtaining heavy and light chain antibody sequences from antibody-expressing cells. Alternatively, obtaining a sequencing library from a target B cell lymphocyte can be performed by methods other than those described herein. Other suitable, but non-limiting, methods are described in PCT/US2017/054628, filed on 29/9/2017, and the entirety of which is incorporated herein by reference for all purposes.

Capture/priming oligonucleotides. The capture/priming oligonucleotide may comprise a first priming sequence and a capture sequence. The capture/priming oligonucleotide may comprise a 5 'endmost nucleotide and a 3' endmost nucleotide.

And (5) capturing the sequence. The capture sequence is an oligonucleotide sequence configured to capture nucleic acids from lysed cells. In various embodiments, the capture sequence may be near or include the 3' endmost nucleotide of the capture/priming oligonucleotide. The capture sequence can have from about 6 to about 50 nucleotides. In some embodiments, the capture sequence captures the nucleic acid by hybridizing to the nucleic acid released from the target cell. In some of the methods described herein, the nucleic acid released from the target B cell may be mRNA. The capture sequence that can capture and hybridize to the mRNA can include a polyT sequence, which mRNA has a PolyA sequence at the 3' end of the mRNA. The polyT sequence may have from about 20T nucleotides to more than 100T nucleotides. In some embodiments, the polyT sequence may have from about 30 to about 40 nucleotides. The polyT sequence may further contain two nucleotides VI at its 3' end.

A first priming sequence. The first priming sequence of the capture/priming oligonucleotide may be: 5 'of the capture sequence, proximal to the 5' endmost nucleotide of the capture/priming oligonucleotide; or the 5' endmost nucleotide comprising a capture/priming oligonucleotide. The first priming sequence may be a universal sequence or a sequence specific priming sequence. The first priming sequence may be bound to a primer that, upon binding, primes the reverse transcriptase. The first priming sequence may comprise from about 10 to about 50 nucleotides.

Additional priming and/or adaptor sequences. The capture oligonucleotide may optionally have one or more additional priming/adaptor sequences that provide a landing site for primer extension (which may include extension by a polymerase) or a site for immobilization to a complementary hybridization anchor site within a massively parallel sequencing array or flow cell. In the methods herein, the second (or additional) priming sequence may be a P1 sequence (e.g., AAGCAGTGGTATCAACGCAGAGT (SEQ ID No.1), as used in Illumina sequencing chemistry), but the methods are not so limited. Any suitable priming sequence for use in the preparation of other types of NGS libraries may be included. In some embodiments, when the P1 sequence is included as an additional priming sequence, it may be 5' to the first priming sequence. The P1 additional priming sequence may also be 5' to the capture sequence.

Template switching oligonucleotides. Template switch oligonucleotide as used herein refers to an oligonucleotide that allows terminal transferase activity of a suitable reverse transcriptase, such as but not limited to Moloney Murine Leukemia Virus (MMLV), to anchor the template switch oligonucleotide with added deoxycytidine nucleotides. After base pairing between the template switch oligonucleotide and the additional deoxycytidine, the reverse transcriptase "switches" the template strand from the captured RNA to the template switch oligonucleotide and proceeds to copy to the 5' end of the template switch oligonucleotide. Thus, the entire 5' end of the transcribed NA is included, and additional priming sequences for further amplification may be introduced. In addition, the cDNA is transcribed in a sequence-independent manner.

BCR gene sequence. The B cell receptor gene sequence includes several subregions to include variable (V), diversity (D), junction (J) and constant (C) segments in 5 'to 3' order in the released RNA. The constant region is exactly 5' to the polyA sequence. In many methods of sequencing BCRs, it may be desirable to construct a selection strategy that results in amplicons for sequencing that do not contain poly a sequences (tails). Furthermore, it may be desirable to generate amplicons that retain few constant regions. Limiting amplification to exclude these segments of released nucleic acid sequence may allow for more robust sequencing of V, D (if present) and J segments of BCR.

To better understand this approach, turning to FIGS. 8A-8H, each of FIGS. 8A-8H represents a single species or a set of related double stranded species present at different points in the method of obtaining a BCR sequencing library from a single cell. The method can be multiplexed such that many individual cells can be processed to provide a sequencing library that can be traced back to a particular start site within the well plate. Knowledge of this location can be further traced to a single isolation dock within the microfluidic device from which the cells have been exported. Thus, biological cells can be assayed for the desired ability to produce desired products or stimulate other cells, and then traceably exported, traceably processed to provide sequencing libraries, and the resulting genomic data resulting from the sequencing can be identifiably correlated back to the source cells in the microfluidic device.

In fig. 8A, mRNA 810 is released by lysing the biological cells. The released mRNA 810 may include the target gene sequence 805 and have a poly a segment 815 at its 3' end. Capture/priming oligonucleotide 820 may include P1 priming sequence 825 and a PolyT capture sequence (shown in fig. 8A as T30NI, which represents a sequence of 30T nucleotides and has the double nucleotide sequence NI at the 3' end of capture/priming oligonucleotide 820). In some embodiments, the N nucleotides in the T30NI sequence of the PolyT capture sequence may be selected from G, C and a (e.g., T nucleotides may be excluded). The capture/priming oligonucleotide can bind to the polyA sequence 815 of the released mRNA 810.

In fig. 8B, the initial process of reverse transcription is shown, wherein reverse transcriptase extends the capture/priming oligonucleotide using mRNA 810 as a template, thereby introducing the target gene 805 into the transcript. Upon reaching the 3' end of mRNA 810, the reverse transcriptase adds several C (here shown as three C) nucleotides.

In fig. 8C, a transcript conversion oligonucleotide (TSO)835 is present in the reverse transcription reaction mixture, where the TSO includes P1 sequence 825 and may also include a tetranucleotide (N4) first barcode 802. In the embodiments described below, TSO835 can be an oligonucleotide having the sequence of SEQ ID No. 3. The first barcode 802 may be used for several experiments for multiplexing during sequencing, and the method is not limited to requiring the presence of the first barcode 802. The first barcode 802 is not limited to having four nucleotides, but can have any suitable number of nucleotides to make the sequencing library products identifiable. In some embodiments, the first (multiplex) barcode may have from about 3 nucleotides to about 10 nucleotides. The TSO may also include biotin attached to the 5' end of the oligonucleotide to improve efficiency.

In reverse transcription, the TSO aligns to the 5 'end of mRNA 810 and allows reverse transcriptase to "switch templates" and extends cDNA 830 beyond the three C nucleotides of its 3' end using the deoxynucleotides of the TSO as a template to incorporate the first four nucleotide barcode 802(N4) and P1 sequence 825 of the TSO as shown in fig. 8D. The fully extended cDNA product 840 now includes the P1 priming sequence, the target gene 805 and optionally the first barcode 802(N4) at both ends. cDNA product 840 also includes polyT-NI sequence incorporated from the capture sequence of capture/priming oligonucleotide 820.

In FIG. 8E, cDNA 840 can be amplified using forward and reverse P1 primers 845 to amplify the captured intact mRNA. In the examples described below, the P1 primer may have biotin at its 5' end and may have the sequence of SEQ id No. 4. The sequence of the amplified product (amplified cDNA 840) retains all of the features of the transcript resulting from the reverse transcription step, including the 5 'and 3' end P1 sequences, the target gene sequence 805 and the first barcode 802. The first barcode is incorporated 5' to the target gene sequence 805 and 3' to the P1 priming sequence at the 5' end of the amplification product 840.

A first Polymerase Chain Reaction (PCR) is then performed. Fig. 8F shows a schematic of the primer arrangement used to selectively amplify the focus region of the BCR region. The forward primer 850 was designed to bind to the portion of the 5 'region of the cDNA amplification product 840 that was 3' to the P1 sequence 825 that had been used for the amplification of fig. 8E. The forward primer 850 may also include a second 6-nucleotide barcode 804, which may be used as part of a system to identify the source wells of a well plate. Although a 6 nucleotide second barcode 804 is shown in fig. 8E-8H, the second barcode can have any suitable number of nucleotides, and can have from about 3 nucleotides to about 10 nucleotides. The reverse primer 855 is directed to bind a subregion of the large constant region of the BCR, near the 5 'end of the constant region, where it follows the 3' end of the junction (J) region of the BCR. This ensures that all variable (V), diversity (D) (if present) and junction (J) sub-regions fall within the sequencing portion of the amplicon. This removes the T30NI sequence and the P1 sequence 825 introduced from the capture/priming sequence 820. To ensure coverage of the heavy chain and the kappa and lambda light chains, a mixture of reverse primers 855 was used.

The selected and truncated amplification product 860 was subjected to a second PCR amplification, as shown in FIG. 8G. The amplification product 860 contains the target gene sequence 805, optionally a first (multiplex) barcode sequence 802(N4) and a second (well plate) barcode 804 (N6). The forward primer 865 for the second PCR binds to the sequence of the 5' of the second (well plate) barcode 804 into which the forward primer 850 was introduced. Reverse primer 870 binds to a consensus sequence introduced by reverse primers 855 (the 5' portion of each reverse primer 855). The reverse primer 870 also includes a third (well plate) barcode 806 having 6 nucleotides (N6). Although fig. 8G-8H show a 6 nucleotide third (well plate) barcode 806, the third barcode can have any suitable number of nucleotides, and can have from about 3 nucleotides to about 10 nucleotides.

The final amplicon 880 is shown in fig. 8H and contains a first optional multiplex barcode 802, a gene of interest sequence 805, a second (well plate) barcode 804, and a third (well plate) barcode 806. Additional adapters may be present for a particular sequencing chemistry.

Barcodes 2 and 3 are used across the wells of the output well plate to unambiguously identify each source well to determine the cells that generated the sequencing library. One economical approach may be to use unique barcodes distributed across 8X12 of wells of a well plate, thus only 20 unique barcodes are required to identify each well. The first (multiplex) barcode may be used if multiple well plate samples are combined in a sequencing run, but is not necessary if only one well plate is sequenced in a sequencing run.

Examples

93页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于检测大肠癌的生物标志物

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!